1
|
Pečar Fonović U, Kos J, Mitrović A. Compensational role between cathepsins. Biochimie 2024; 226:62-76. [PMID: 38663456 DOI: 10.1016/j.biochi.2024.04.010] [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: 01/23/2024] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
Cathepsins, a family of lysosomal peptidases, play a crucial role in maintaining cellular homeostasis by regulating protein turnover and degradation as well as many specific regulatory actions that are important for proper cell function and human health. Alterations in the activity and expression of cathepsins have been observed in many diseases such as cancer, inflammation, neurodegenerative disorders, bone remodelling-related conditions and others. These changes are not exclusively harmful, but rather appear to be a compensatory response on the lack of one cathepsin in order to maintain tissue integrity. The upregulation of specific cathepsins in response to the inhibition or dysfunction of other cathepsins suggests a fine-tuned system of proteolytic balance and understanding the compensatory role of cathepsins may improve therapeutic potential of cathepsin's inhibitors. Selectively targeting one cathepsin or modulating their activity could offer new treatment strategies for a number of diseases. This review emphasises the need for comprehensive research into cathepsin biology in the context of disease. The identification of the specific cathepsins involved in compensatory responses, the elucidation of the underlying molecular mechanisms and the development of targeted interventions could lead to innovative therapeutic approaches.
Collapse
Affiliation(s)
- Urša Pečar Fonović
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia.
| | - Janko Kos
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia; Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000, Ljubljana, Slovenia.
| | - Ana Mitrović
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia; Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Theeke LA, Liu Y, Wang S, Luo X, Navia RO, Xiao D, Xu C, Wang K. Plasma Proteomic Biomarkers in Alzheimer's Disease and Cardiovascular Disease: A Longitudinal Study. Int J Mol Sci 2024; 25:10751. [PMID: 39409080 PMCID: PMC11477191 DOI: 10.3390/ijms251910751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
The co-occurrence of Alzheimer's disease (AD) and cardiovascular diseases (CVDs) in older adults highlights the necessity for the exploration of potential shared risk factors. A total of 566 adults were selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, including 111 individuals with AD, 383 with mild cognitive impairment (MCI), and 410 with CVD. The multivariable linear mixed model (LMM) was used to investigate the associations of AD and CVD with longitudinal changes in 146 plasma proteomic biomarkers (measured at baseline and the 12-month follow-up). The LMM showed that 48 biomarkers were linked to AD and 46 to CVD (p < 0.05). Both AD and CVD were associated with longitudinal changes in 14 biomarkers (α1Micro, ApoH, β2M, BNP, complement C3, cystatin C, KIM1, NGAL, PPP, TIM1, THP, TFF3, TM, and VEGF), and both MCI and CVD were associated with 12 biomarkers (ApoD, AXL, BNP, Calcitonin, CD40, C-peptide, pM, PPP, THP, TNFR2, TTR, and VEGF), suggesting intricate connections between cognitive decline and cardiovascular health. Among these, the Tamm Horsfall Protein (THP) was associated with AD, MCI, CVD, and APOE-ε4. This study provides valuable insights into shared and distinct biological markers and mechanisms underlying AD and CVD.
Collapse
Affiliation(s)
- Laurie A. Theeke
- Department of Community of Acute and Chronic Care, School of Nursing, The George Washington University, Ashburn, VA 20147, USA;
| | - Ying Liu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN 37614, USA;
| | - Silas Wang
- Department of Statistics & Data Science, Dietrich College of Humanities and Social Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06516, USA;
| | - R. Osvaldo Navia
- Department of Medicine and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA;
| | - Danqing Xiao
- Department of STEM, School of Arts and Sciences, Regis College, Weston, MA 02493, USA;
| | - Chun Xu
- Department of Health and Biomedical Sciences, College of Health Affairs, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA;
| | - Kesheng Wang
- Department of Biobehavioral Health & Nursing Science, College of Nursing, University of South Carolina, Columbia, 1601 Greene Street, Columbia, SC 29208, USA
| | | |
Collapse
|
3
|
Sun H, Tang Q, Yan X, Xie W, Xu Y, Zhang W. Cathepsins and neurological diseases: a Mendelian randomization study. Front Neurosci 2024; 18:1454369. [PMID: 39420987 PMCID: PMC11484041 DOI: 10.3389/fnins.2024.1454369] [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: 06/25/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Background The causal relationship between cathepsins and neurological diseases remains uncertain. To address this, we utilized a two-sample Mendelian randomization (MR) approach to assess the potential causal effect of cathepsins on the development of neurological diseases. Methods This study conducted a two-sample two-way MR study using pooled data from published genome-wide association studies to evaluate the relationship between 10 cathepsins (B, D, E, F, G, H, L2, O, S, and Z) and 7 neurological diseases, which included ischemic stroke, cerebral hemorrhage, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, and epilepsy. The analysis employed various methods such as inverse variance weighting (IVW), weighted median, MR Egger regression, MR pleiotropy residual sum and outlier, Cochran Q statistic, and leave-one-out analysis. Results We found a causal relationship between cathepsins and neurological diseases, including Cathepsin B and Parkinson's disease (IVW odds ratio (OR): 0.89, 95% confidence interval (CI): 0.83, 0.95, p = 0.001); Cathepsin D and Parkinson's disease (OR: 0.80, 95%CI: 0.68, 0.95, p = 0.012); Cathepsin E and ischemic stroke (OR: 1.05, 95%CI: 1.01, 1.09, p = 0.015); Cathepsin O and ischemic stroke (OR: 1.05, 95%CI: 1.01, 1.10, p = 0.021). Reverse MR analyses revealed that multiple sclerosis and Cathepsin E (OR: 1.05, 95%CI: 1.01, 1.10, p = 0.030). There is currently no significant relationship has been found between other cathepsins and neurological diseases. Conclusion Our study reveals a causal relationship between Cathepsins B, D, E, and O and neurological diseases, offering valuable insights for research aimed at improving the diagnosis and treatment of such conditions.
Collapse
Affiliation(s)
- Haitao Sun
- Changchun University of Chinese Medicine, Changchun, China
| | - Qingqing Tang
- Changchun University of Chinese Medicine, Changchun, China
| | - Xue Yan
- The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Wanying Xie
- Changchun University of Chinese Medicine, Changchun, China
| | - Yueshan Xu
- Changchun University of Chinese Medicine, Changchun, China
| | - Weimin Zhang
- Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
4
|
Zeng R, Zhou Z, Liao W, Guo B. Genetic insights into the role of cathepsins in cardiovascular diseases: a Mendelian randomization study. ESC Heart Fail 2024; 11:2707-2718. [PMID: 38714485 PMCID: PMC11424349 DOI: 10.1002/ehf2.14826] [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: 01/30/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 05/10/2024] Open
Abstract
AIMS This study aimed to explore the causal relationships between cathepsins and cardiovascular diseases (CVDs) by Mendelian randomization (MR) analysis. METHODS AND RESULTS Single nucleotide polymorphisms (SNPs) associated with nine cathepsin types (cathepsins B, E, F, G, H, O, S, L2, and Z) were obtained from the INTERVAL study (3301 individuals). CVDs data were acquired from the UK Biobank (coronary atherosclerosis: 14 334 cases, 346 860 controls) and a genome-wide association study (GWAS) (myocardial infarction: 20 917 cases, 440 906 controls; myocarditis: 633 cases, 427 278 controls; chronic heart failure: 14 262 cases, 471 898 controls; angina pectoris: 30 025 cases, 440 906 controls; stable angina pectoris: 17 894 cases, 325 132 controls; unstable angina pectoris: 9481 cases, 446 987 controls; pericarditis: 1795 cases, 453 370 controls). Inverse variance weighted (IVW), MR-Egger, weighted median methods were adopted to conduct univariable MR (UVMR), reverse MR, multivariable MR (MVMR) analyses to estimate causality. The UVMR analyses demonstrated significant causal relationships between higher cathepsin E levels and increased risk of coronary atherosclerosis [IVW: P = 0.0051, odds ratio (OR) = 1.0033, 95% confidence interval (CI) = 1.0010-1.0056] and myocardial infarction (IVW: P = 0.0097, OR = 1.0553, 95% CI = 1.0131-1.0993), while elevated cathepsin L2 levels were causally related to reduced risk of myocarditis (IVW: P = 0.0120, OR = 0.6895, 95% CI = 0.5158-0.9216) and chronic heart failure (IVW: P = 0.0134, OR = 0.9316, 95% CI = 0.8807-0.9854). Reverse MR analyses revealed that myocardial infarction increased cathepsin O levels (IVW: P = 0.0400, OR = 1.0708, 95% CI = 1.0031-1.1431). MVMR analyses treating nine cathepsins together revealed that the positive causality between cathepsin E levels and coronary atherosclerosis risk (IVW: P = 0.0390, OR = 1.0030, 95% CI = 1.0000-1.0060), and the protective effect of cathepsin L2 levels on myocarditis (IVW: P = 0.0030, OR = 0.6610, 95% CI = 0.5031-0.8676) and chronic heart failure (IVW: P = 0.0090, OR = 0.9259, 95% CI = 0.8737-0.9812) remained, as higher cathepsin O levels were found to be causally related to increased risks of myocarditis (IVW: P = 0.0030, OR = 1.6145, 95% CI = 1.1829-2.2034) and chronic heart failure (IVW: P = 0.0300, OR = 1.0779, 95% CI = 1.0070-1.1537). CONCLUSIONS The study highlights the causalities of cathepsin E, L2, and O on CVDs, offering insights into their roles in cardiovascular biomarkers and therapeutic targets development. Further research is required to apply these genetic findings clinically.
Collapse
Affiliation(s)
- Ruiqi Zeng
- Guangzhou Medical UniversityGuangzhou511436China
- Department of Clinical Medicine, Nanshan CollegeGuangzhou Medical UniversityGuangzhouChina
| | - Zhiyi Zhou
- Guangzhou Medical UniversityGuangzhou511436China
- Department of Clinical Medicine, Third Clinical SchoolGuangzhou Medical UniversityGuangzhouChina
| | - Wanzhe Liao
- Guangzhou Medical UniversityGuangzhou511436China
- Department of Clinical Medicine, Nanshan CollegeGuangzhou Medical UniversityGuangzhouChina
| | - Beian Guo
- Guangzhou Medical UniversityGuangzhou511436China
- Department of Clinical Medicine, Nanshan CollegeGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
5
|
Khalifa O, Al-Akl NS, Arredouani A. Differential expression of cardiometabolic and inflammation markers and signaling pathways between overweight/obese Qatari adults with high and low plasma salivary α-amylase activity. Front Endocrinol (Lausanne) 2024; 15:1421358. [PMID: 39411310 PMCID: PMC11473332 DOI: 10.3389/fendo.2024.1421358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/02/2024] [Indexed: 10/19/2024] Open
Abstract
Background The relationship between salivary α-amylase activity (sAAa) and susceptibility to cardiovascular disorders lacks a definitive consensus in available studies. To fill this knowledge gap, the present study endeavors to investigate this association among overweight/obese otherwise healthy Qatari adults. The study specifically categorizes participants based on their sAAa into high and low subgroups, aiming to provide a more comprehensive understanding of the potential link between sAAa levels and cardiovascular and inflammation markers in this population. Methods Plasma samples of 264 Qatari overweight/obese (Ow/Ob) participants were used to quantify the sAAa and to profile the proteins germane to cardiovascular, cardiometabolic, metabolism, and organ damage in low sAAa (LsAAa) and high sAAa (HsAAa) subjects using the Olink technology. Comprehensive statistical tools as well as chemometric and enrichments analyses were used to identify differentially expressed proteins (DEPs) and their associated signaling pathways and cellular functions. Results A total of ten DEPs were detected, among them five were upregulated (QPCT, LCN2, PON2, DPP7, CRKL) while five were down regulated in the LsAAa subgroup compared to the HsAAa subgroup (ARG1, CTSH, SERPINB6, OSMR, ALDH3A). Functional enrichment analysis highlighted several relevant signaling pathways and cellular functions enriched in the DEPs, including myocardial dysfunction, disorder of blood pressure, myocardial infraction, apoptosis of cardiomyocytes, hypertension, chronic inflammatory disorder, immunes-mediated inflammatory disease, inflammatory response, activation of leukocytes and activation of phagocytes. Conclusion Our study unveils substantial alterations within numerous canonical pathways and cellular or molecular functions that bear relevance to cardiometabolic disorders among Ow/Ob Qatari adults exhibiting LsAAa and HsAAa in the plasma. A more comprehensive exploration of these proteins and their associated pathways and functions offers the prospect of elucidating the mechanistic underpinnings inherent in the documented relationship between sAAa and metabolic disorders.
Collapse
Affiliation(s)
- Olfa Khalifa
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Neyla S. Al-Akl
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Abdelilah Arredouani
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| |
Collapse
|
6
|
Ajani TA, Magwebu ZE, Chauke CG, Obikeze K. Advances in Cathepsin S Inhibition: Challenges and Breakthroughs in Drug Development. PATHOPHYSIOLOGY 2024; 31:471-487. [PMID: 39311309 PMCID: PMC11417842 DOI: 10.3390/pathophysiology31030035] [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: 07/05/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024] Open
Abstract
Cathepsin S (CatS) is a proteolytic enzyme and a member of the cysteine protease family of proteolytic enzymes. Cathepsins S, K, and L are particularly similar in terms of their amino acid sequences and interactions with substrates, and this has made it difficult to develop inhibitors with specificity for either CatS, CatK, or CatL. The involvement of CatS in various disease pathophysiologies (autoimmune disorders, cardiovascular diseases, cancer, etc.) has made it a very important target in drug development. Efforts have been made since the early 1990s to develop a specific CatS inhibitor without any major success. Following many failed efforts to develop an inhibitor for CatS, it was discovered that interactions with the amino acid residues at the S2 and S3 pockets of CatS are critical for the identification of CatS-specific inhibitors. Amino acid residues at these pockets have been the target of recent research focused on developing a non-covalent, reversible, and specific CatS inhibitor. Methods applied in the identification of CatS inhibitors include molecular modeling, in-vitro screening, and in-vivo studies. The molecular modeling process has proven to be very successful in the identification of CatS-specific inhibitors, with R05459072 (Hoffmann-La Roche) and LY3000328 (Eli Lilly Company) which has completed phase 1 clinical trials. CatS inhibitors identified from 2011 to 2023 with promising prospects are discussed in this article.
Collapse
Affiliation(s)
- Temitope A. Ajani
- School of Pharmacy, University of the Western Cape, Cape Town 7535, South Africa;
| | - Zandisiwe E. Magwebu
- South African Medical Research Council, Primate Unit and Delft Animal Centre (PUDAC), Cape Town 7100, South Africa; (Z.E.M.); (C.G.C.)
| | - Chesa G. Chauke
- South African Medical Research Council, Primate Unit and Delft Animal Centre (PUDAC), Cape Town 7100, South Africa; (Z.E.M.); (C.G.C.)
| | - Kenechukwu Obikeze
- School of Pharmacy, University of the Western Cape, Cape Town 7535, South Africa;
| |
Collapse
|
7
|
Li Q, Zhou Z, Xu T, Gao X, Lou Y, Chen Z, Zhang M, Fang Q, Tan J, Huang J. Relationship between cathepsins and cardiovascular diseases: a Mendelian randomized study. Front Pharmacol 2024; 15:1370350. [PMID: 39027333 PMCID: PMC11254818 DOI: 10.3389/fphar.2024.1370350] [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: 01/14/2024] [Accepted: 06/04/2024] [Indexed: 07/20/2024] Open
Abstract
Background: Cardiovascular diseases (CVDs) are the leading age-related disorders worldwide, with their prevalence increasing annually. Cathepsins are protein-degrading enzymes essential for processes such as intracellular protein breakdown, apoptosis, and immune responses. Recent studies suggest a potential link between cathepsins and CVDs, yet the exact causal relationship remains to be elucidated. To address this, we propose using Mendelian randomization (MR) to explore the causal relationships between cathepsins and CVDs. Methods: We obtained single nucleotide polymorphism (SNP) data for cathepsins from the INTERVAL study, a publicly accessible genome-wide association study (GWAS) dataset. Outcome SNP data were sourced from seven distinct GWAS datasets, ensuring a comprehensive analysis across multiple cardiovascular outcomes. For MR analysis, we primarily employed the inverse variance weighted (IVW) method, known for its efficiency when all SNPs are valid instruments. This was supplemented by the weighted median and MR-Egger methods to provide robustness against potential violations of MR assumptions, such as pleiotropy. The IVW method offers precision and efficiency, the weighted median method adds robustness against invalid instruments, and the MR-Egger method helps identify and correct for pleiotropic biases. Cochran's Q test was utilized to assess heterogeneity, and sensitivity analyses were conducted using MR-PRESSO and the leave-one-out approach. Results: The strength of the associations between exposure and outcome was measured using odds ratios (ORs), and results were presented with 95% confidence intervals (CIs). The cathepsin E increases the risk of myocardial infarction (MI) (OR = 1.053%, 95% CI: 1.007-1.101, p = 0.024) and ischemic stroke (IS) (OR = 1.06%, 95% CI: 1.019-1.103, p = 0.004). Conversely, cathepsin L2 decreases the risk of chronic heart failure (CHF) (OR = 0.922%, 95% CI: 0.859-0.99, p = 0.025) and atrial fibrillation (AF) (OR = 0.956%, 95% CI: 0.918-0.996, p = 0.033). Cathepsin O was associated with an increased risk of IS (OR = 1.054%, 95% CI: 1.008-1.102, p = 0.021) and AF (OR = 1.058%, 95% CI: 1.02-1.098, p = 0.002). Conclusion: Our MR analysis reveals that cathepsin E is a risk factor for MI and IS, cathepsin L2 offers protective effects against CHF and AF, and cathepsin O increases the risk for IS and AF.
Collapse
Affiliation(s)
- Qiaoqiao Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongzheng Zhou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Teng Xu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xueping Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yake Lou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zijun Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Muzi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghua Fang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Tan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Filippova TA, Masamrekh RA, Khudoklinova YY, Shumyantseva VV, Kuzikov AV. The multifaceted role of proteases and modern analytical methods for investigation of their catalytic activity. Biochimie 2024; 222:169-194. [PMID: 38494106 DOI: 10.1016/j.biochi.2024.03.006] [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: 09/25/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
We discuss the diverse functions of proteases in the context of their biotechnological and medical significance, as well as analytical approaches used to determine the functional activity of these enzymes. An insight into modern approaches to studying the kinetics and specificity of proteases, based on spectral (absorption, fluorescence), mass spectrometric, immunological, calorimetric, and electrochemical methods of analysis is given. We also examine in detail electrochemical systems for determining the activity and specificity of proteases. Particular attention is given to exploring innovative electrochemical systems based on the detection of the electrochemical oxidation signal of amino acid residues, thereby eliminating the need for extra redox labels in the process of peptide synthesis. In the review, we highlight the main prospects for the further development of electrochemical systems for the study of biotechnologically and medically significant proteases, which will enable the miniaturization of the analytical process for determining the catalytic activity of these enzymes.
Collapse
Affiliation(s)
- Tatiana A Filippova
- Institute of Biomedical Chemistry, 10 bld. 8, Pogodinskaya str., 119121, Moscow, Russia; Pirogov Russian National Research Medical University, 1, Ostrovityanova Street, Moscow, 117513, Russia
| | - Rami A Masamrekh
- Institute of Biomedical Chemistry, 10 bld. 8, Pogodinskaya str., 119121, Moscow, Russia; Pirogov Russian National Research Medical University, 1, Ostrovityanova Street, Moscow, 117513, Russia
| | - Yulia Yu Khudoklinova
- Pirogov Russian National Research Medical University, 1, Ostrovityanova Street, Moscow, 117513, Russia
| | - Victoria V Shumyantseva
- Institute of Biomedical Chemistry, 10 bld. 8, Pogodinskaya str., 119121, Moscow, Russia; Pirogov Russian National Research Medical University, 1, Ostrovityanova Street, Moscow, 117513, Russia
| | - Alexey V Kuzikov
- Institute of Biomedical Chemistry, 10 bld. 8, Pogodinskaya str., 119121, Moscow, Russia; Pirogov Russian National Research Medical University, 1, Ostrovityanova Street, Moscow, 117513, Russia.
| |
Collapse
|
9
|
Wu Y, Jiang D, Liu Q, Yan S, Liu X, Wu T, Sun W, Li G. Cathepsin L induces cellular senescence by upregulating CUX1 and p16 INK4a. Aging (Albany NY) 2024; 16:10749-10764. [PMID: 38944813 PMCID: PMC11272106 DOI: 10.18632/aging.205955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 04/18/2024] [Indexed: 07/01/2024]
Abstract
Cathepsin L (CTSL) has been implicated in aging and age-related diseases, such as cardiovascular diseases, specifically atherosclerosis. However, the underlying mechanism(s) is not well documented. Recently, we demonstrated a role of CUT-like homeobox 1 (CUX1) in regulating the p16INK4a-dependent cellular senescence in human endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) via its binding to an atherosclerosis-associated functional SNP (fSNP) rs1537371 on the CDKN2A/B locus. In this study, to determine if CTSL, which was reported to proteolytically activate CUX1, regulates cellular senescence via CUX1, we measured the expression of CTSL, together with CUX1 and p16INK4a, in human ECs and VSMCs undergoing senescence. We discovered that CUX1 is not a substrate that is cleaved by CTSL. Instead, CTSL is an upstream regulator that activates CUX1 transcription indirectly in a process that requires the proteolytic activity of CTSL. Our findings suggest that there is a transcription factor in between CTSL and CUX1, and cleavage of this factor by CTSL can activate CUX1 transcription, inducing endothelial senescence. Thus, our findings provide new insights into the signal transduction pathway that leads to atherosclerosis-associated cellular senescence.
Collapse
Affiliation(s)
- Yuwei Wu
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha, China
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Danli Jiang
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
- International Center for Aging and Cancer Hainan Medical University, Hainan, China
| | - Qing Liu
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Shaoyang Yan
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Tsinghua Medicine, Tsinghua University, Peking, China
| | - Xiuzhen Liu
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ting Wu
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha, China
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Wei Sun
- Department of Medicine, Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Gang Li
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Medicine, Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
10
|
Jin X, Yue X, Huang Z, Meng X, Xu S, Wu Y, Wan Y, Inoue A, Narisawa M, Hu L, Shi GP, Umegaki H, Murohara T, Lei Y, Kuzuya M, Cheng XW. Cathepsin K deficiency prevented stress-related thrombosis in a mouse FeCl 3 model. Cell Mol Life Sci 2024; 81:205. [PMID: 38703204 PMCID: PMC11069486 DOI: 10.1007/s00018-024-05240-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Exposure to chronic psychological stress (CPS) is a risk factor for thrombotic cardiocerebrovascular diseases (CCVDs). The expression and activity of the cysteine cathepsin K (CTSK) are upregulated in stressed cardiovascular tissues, and we investigated whether CTSK is involved in chronic stress-related thrombosis, focusing on stress serum-induced endothelial apoptosis. METHODS AND RESULTS Eight-week-old wild-type male mice (CTSK+/+) randomly divided to non-stress and 3-week restraint stress groups received a left carotid artery iron chloride3 (FeCl3)-induced thrombosis injury for biological and morphological evaluations at specific timepoints. On day 21 post-stress/injury, the stress had enhanced the arterial thrombi weights and lengths, in addition to harmful alterations of plasma ADAMTS13, von Willebrand factor, and plasminogen activation inhibitor-1, plus injured-artery endothelial loss and CTSK protein/mRNA expression. The stressed CTSK+/+ mice had increased levels of injured arterial cleaved Notch1, Hes1, cleaved caspase8, matrix metalloproteinase-9/-2, angiotensin type 1 receptor, galactin3, p16IN4A, p22phox, gp91phox, intracellular adhesion molecule-1, TNF-α, MCP-1, and TLR-4 proteins and/or genes. Pharmacological and genetic inhibitions of CTSK ameliorated the stress-induced thrombus formation and the observed molecular and morphological changes. In cultured HUVECs, CTSK overexpression and silencing respectively increased and mitigated stressed-serum- and H2O2-induced apoptosis associated with apoptosis-related protein changes. Recombinant human CTSK degraded γ-secretase substrate in a dose-dependent manor and activated Notch1 and Hes1 expression upregulation. CONCLUSIONS CTSK appeared to contribute to stress-related thrombosis in mice subjected to FeCl3 stress, possibly via the modulation of vascular inflammation, oxidative production and apoptosis, suggesting that CTSK could be an effective therapeutic target for CPS-related thrombotic events in patients with CCVDs.
Collapse
Affiliation(s)
- Xueying Jin
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Xueling Yue
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| | - Zhe Huang
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Xiangkun Meng
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Yuna Wu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
| | - Ying Wan
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Aiko Inoue
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Institute of Innovation for Future Society, Nagoya University, Nagoya, Aichi-Ken, 466-8550, Japan
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin, 541199, Guangxi, People's Republic of China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hiroyuki Umegaki
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Institute of Innovation for Future Society, Nagoya University, Nagoya, Aichi-Ken, 466-8550, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Yanna Lei
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
- Department of Intensive Care, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
| | - Masafumi Kuzuya
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Meitetsu Hospital, Nagoya, Aichi, 451-8511, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, Jilin, People's Republic of China.
| |
Collapse
|
11
|
Jansen I, Cahalane R, Hengst R, Akyildiz A, Farrell E, Gijsen F, Aikawa E, van der Heiden K, Wissing T. The interplay of collagen, macrophages, and microcalcification in atherosclerotic plaque cap rupture mechanics. Basic Res Cardiol 2024; 119:193-213. [PMID: 38329498 PMCID: PMC11008085 DOI: 10.1007/s00395-024-01033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
The rupture of an atherosclerotic plaque cap overlying a lipid pool and/or necrotic core can lead to thrombotic cardiovascular events. In essence, the rupture of the plaque cap is a mechanical event, which occurs when the local stress exceeds the local tissue strength. However, due to inter- and intra-cap heterogeneity, the resulting ultimate cap strength varies, causing proper assessment of the plaque at risk of rupture to be lacking. Important players involved in tissue strength include the load-bearing collagenous matrix, macrophages, as major promoters of extracellular matrix degradation, and microcalcifications, deposits that can exacerbate local stress, increasing tissue propensity for rupture. This review summarizes the role of these components individually in tissue mechanics, along with the interplay between them. We argue that to be able to improve risk assessment, a better understanding of the effect of these individual components, as well as their reciprocal relationships on cap mechanics, is required. Finally, we discuss potential future steps, including a holistic multidisciplinary approach, multifactorial 3D in vitro model systems, and advancements in imaging techniques. The obtained knowledge will ultimately serve as input to help diagnose, prevent, and treat atherosclerotic cap rupture.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Cahalane
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ranmadusha Hengst
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ali Akyildiz
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank Gijsen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
12
|
Ajani TA, Obikeze K, Magwebu ZE, Egieyeh S, Chauke CG. In-silico and in-vitro screening of Asiatic acid and Asiaticoside A against Cathepsin S enzyme. BMC Pharmacol Toxicol 2023; 24:67. [PMID: 38007481 PMCID: PMC10676574 DOI: 10.1186/s40360-023-00701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/31/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Atherosclerosis is a form of cardiovascular disease that affects the endothelium of the blood vessel. Series of events are involved in the pathophysiology of this disease which includes the breaking down of the connective tissue elastin and collagen responsible for the tensile strength of the arterial wall by proteolytic enzyme. One of these enzymes called Cathepsin S (CatS) is upregulated in the progression of the disease and its inhibition has been proposed to be a promising pharmacological target to improve the prognosis of the disease condition. Asiatic acid and asiaticoside A are both pentacyclic triterpenoids isolated from Centella asiatica. Their use in treating various cardiovascular diseases has been reported. METHODS In this study through in silico and in vitro methods, the pharmacokinetic properties, residue interaction, and inhibitory activities of these compounds were checked against the CatS enzyme. The SwissADME online package and the ToxTree 3.01 version of the offline software were used to determine the physicochemical properties of the compounds. RESULT Asiatic acid reported no violation of the Lipinski rule while asiaticoside A violated the rule with regards to its molecular structure and size. The molecular docking was done using Molecular Operating Environment (MOE) and the S-score of - 7.25988, - 7.08466, and - 4.147913 Kcal/mol were recorded for LY300328, asiaticoside A, and asiatic acid respectively. Asiaticoside A has a docking score value (- 7.08466Kcal/mol) close to the co-crystallize compound. Apart from the close docking score, the amino acid residue glycine69 and asparagine163 both interact with the co-crystallized compound and asiaticoside A. The in vitro result clearly shows the inhibitory effect of asiaticoside and asiatic acid. Asiaticoside A has an inhibitory value of about 40% and asiatic acid has an inhibitory value of about 20%. CONCLUSION This clearly shows that asiaticoside will be a better drug candidate than asiatic acid in inhibiting the CatS enzyme for the purpose of improving the outcome of atherosclerosis. However, certain modifications need to be made to the structural make-up of asiaticoside A to improve its pharmacokinetics properties.
Collapse
Affiliation(s)
| | - Kenechukwu Obikeze
- University of the Western Cape, School of Pharmacy, Bellville, South Africa
| | - Zandisiwe E Magwebu
- South African Medical Research Council, Primate Unit and Delft Animal Centre (PUDAC), Cape town, South Africa
| | - Samuel Egieyeh
- University of the Western Cape, School of Pharmacy, Bellville, South Africa
| | - Chesa G Chauke
- South African Medical Research Council, Primate Unit and Delft Animal Centre (PUDAC), Cape town, South Africa
| |
Collapse
|
13
|
Jamuna S, Ashokkumar R, Raja IS, Devaraj SN. Anti-Atherogenic Protection by Oligomeric Proanthocyanidins via Regulating Collagen Crosslinking Against CC Diet-Induced Atherosclerosis in Rats. Appl Biochem Biotechnol 2023; 195:4881-4892. [PMID: 37097399 DOI: 10.1007/s12010-023-04487-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
The synthesis of collagen and its turnover remained as critical determinants for the progression of atherosclerosis. During this condition, proteases secreted by SMCs and foam cells in the necrotic core degrade collagen. Growing evidences demonstrated that consumption of antioxidant rich diet is highly associated with a reduced risk of atherosclerosis. Oligomeric proanthocyanidins (OPC) have been proved to possess promising antioxidant, anti-inflammatory and cardioprotective activity, based on our previous studies. The present study aims to investigate the efficacy of OPC isolated from Crataegus oxyacantha berries as a natural collagen crosslinker and anti-atherogenic agent. Spectral studies like FTIR, ultraviolet and circular dichroism analysis confirmed the in vitro crosslinking ability of OPC with rat tail collagen when compared to the standard epigallocatechin gallate. The administration of cholesterol:cholic acid (CC) diet induces proteases-mediated collagen degradation that could result in plaque instability. Further, the CC diet fed rats showed significantly increased levels of total cholesterol and triacylglycerols which, in turn, increases the activities of collagen degrading proteases-MMPs (MMP 1, 2 and 9) and Cathepsin S and D. Upon OPC treatment, marked reduction in the lipid content, activation of proteases with concomitant increase in the mRNA levels of collagen Type I and Type III as similar to atorvastatin treatment were observed .Thus, OPC supplementation may contribute to the prevention of atherosclerotic plaque instability by acting as a natural crosslinker of collagen.
Collapse
Affiliation(s)
- Sankar Jamuna
- Affyclone Laboratories Pvt Ltd., 600044, Chrompet, Chennai, India
- Department of Biochemistry, University of Madras, Guindy campus, 600025, Chennai, India
| | - Rathinavel Ashokkumar
- Affyclone Laboratories Pvt Ltd., 600044, Chrompet, Chennai, India
- Department of Biochemistry, University of Madras, Guindy campus, 600025, Chennai, India
| | | | | |
Collapse
|
14
|
Zuo L, Dong Y, Pan Y, Yan H, Meng X, Li H, Zhao X, Wang Y, Wang Y, Liao X. Impact of Serum Cystatin C Level on Long-Term Cognitive Impairment After Acute Ischemic Stroke and Transient Ischemic Attack. Neuropsychiatr Dis Treat 2023; 19:1543-1554. [PMID: 37435549 PMCID: PMC10329915 DOI: 10.2147/ndt.s412825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/08/2023] [Indexed: 07/13/2023] Open
Abstract
Objective Cognitive impairment after stroke/transient ischemic attack (TIA) has a high prevalence. Cystatin C (CysC) has been found as a novel biomarker of neurodegenerative diseases, such as dementia and Alzheimer's disease. We aimed to explore the possible correlations of serum CysC level with cognitive impairment in patients who had mild ischemic stroke and TIA after 1 year. Methods We measured serum CysC level in 1025 participants with a minor ischemic stroke/TIA from enrolled from the Impairment of Cognition and Sleep (ICONS) study of the China National Stroke Registry-3 (CNSR-3). They were divided into four groups according to quartiles of baseline CysC levels. Patients' cognitive functions were assessed by Montreal Cognitive Assessment (MoCA)-Beijing at day 14 and at 1 year. Multiple logistic regression models were performed to evaluate the relationship between CysC and post-stroke cognitive impairment (PSCI) at 1-year follow-up. Results Cognitive impairment was defined as MoCA-Beijing ≤22. Most patients were in 60s (61.52±10.97 years old) with a median (interquartile range) National Institute of Health Stroke Scale(NIHSS) score of 3.00 (4.00) and greater than primary school level of education, and 743 participants (72.49%) were male. Among the 1025 participants, 331 participants (32.29%) patients suffered PSCI at 1-year follow-up. A U-shaped association was observed between CysC and 1-year PSCI [quartile (Q)1 vs Q3: adjusted odds ratio (aOR) 2.69, 95% CI 1.67-4.34, p < 0.0001; Q2 vs Q3: aOR 1.63, 95% CI 1.03-2.57, p = 0.0354; Q4 vs Q3: aOR 1.83, 95% CI 1.16-2.87, p = 0.009]. Moreover, the U-shaped trends were also found between CysC level and the subscores of attention, recall, abstraction and language in MoCA. Conclusion CysC showed a U-shaped correlation with 1-year overall cognitive function. It is probable that measurement of the serum CysC level would aid in the early diagnosis of PSCI.
Collapse
Affiliation(s)
- Lijun Zuo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yanhong Dong
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore, 117597, Singapore
| | - Yuesong Pan
- National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hongyi Yan
- National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xia Meng
- National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hao Li
- National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xingquan Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoling Liao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
15
|
Cheng XW, Narisawa M, Wang H, Piao L. Overview of multifunctional cysteinyl cathepsins in atherosclerosis-based cardiovascular disease: from insights into molecular functions to clinical implications. Cell Biosci 2023; 13:91. [PMID: 37202785 DOI: 10.1186/s13578-023-01040-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023] Open
Abstract
Cysteinyl cathepsins (CTSs) are widely known to have a proteolysis function that mediates recycling of unwanted proteins in endosomes and lysosomes, and investigation of CTSs has greatly improved with advances in live-imaging techniques both in vivo and in vitro, leading to three key findings. (1) CTSs are relocated from the lysosomes to other cellular spaces (i.e., cytosol, nucleus, nuclear membrane, plasma membrane, and extracellular milieu). (2) In addition to acidic cellular compartments, CTSs also exert biological activity in neutral environments. (3) CTSs also exert multiple nontraditional functions in, for example, extracellular matrix metabolism, cell signaling transduction, protein processing/trafficking, and cellular events. Various stimuli regulate the expression and activities of CTSs in vivo and vitro-e.g., inflammatory cytokines, oxidative stress, neurohormones, and growth factors. Accumulating evidence has confirmed the participation of CTSs in vascular diseases characterized by atherosclerosis, plaque rupture, thrombosis, calcification, aneurysm, restenosis/in-stent-restenosis, and neovasel formation. Circulating and tissue CTSs are promising as biomarkers and as a diagnostic imaging tool in patients with atherosclerosis-based cardiovascular disease (ACVD), and pharmacological interventions with their specific and non-specific inhibitors, and cardiovascular drugs might have potential for the therapeutic targeting of CTSs in animals. This review focuses on the update findings on CTS biology and the involvement of CTSs in the initiation and progression of ACVD and discusses the potential use of CTSs as biomarkers and small-molecule targets to prevent deleterious nontraditional functions in ACVD.
Collapse
Affiliation(s)
- Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China.
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, Jilin PR. 133000, China.
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4668550, Japan
| | - Hailong Wang
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China
| |
Collapse
|
16
|
S S, Camardo A, Dahal S, Ramamurthi A. Surface-Functionalized Stem Cell-Derived Extracellular Vesicles for Vascular Elastic Matrix Regenerative Repair. Mol Pharm 2023. [PMID: 37093652 DOI: 10.1021/acs.molpharmaceut.2c00769] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Extracellular vesicles (EVs) are nanosized vesicles that carry cell-specific biomolecular information. Our previous studies showed that adult human bone marrow mesenchymal stem cell (BM-MSC)-derived EVs provide antiproteolytic and proregenerative effects in cultures of smooth muscle cells (SMCs) derived from an elastase-infused rat abdominal aortic aneurysm (AAA) model, and this is promising toward their use as a therapeutic platform for naturally irreversible elastic matrix aberrations in the aortic wall. Since systemically administered EVs poorly home into sites of tissue injury, disease strategies to improve their affinity toward target tissues are of great significance for EV-based treatment strategies. Toward this goal, in this work, we developed a postisolation surface modification strategy to target MSC-derived EVs to the AAA wall. The EVs were surface-conjugated with a short, synthetic, azide-modified peptide sequence for targeted binding to cathepsin K (CatK), a cysteine protease overexpressed in the AAA wall. Conjugation was performed using a copper-free click chemistry method. We determined that such conjugation improved EV uptake into cultured aneurysmal SMCs in culture and their binding to the wall of matrix injured vessels ex vivo. The proregenerative and antiproteolytic effects of MSC-EVs on cultured rat aneurysmal SMCs were also unaffected following peptide conjugation. From this study, it appears that modification with short synthetic peptide sequences seems to be an effective strategy for improving the cell-specific uptake of EVs and may be effective in facilitating AAA-targeted therapy.
Collapse
Affiliation(s)
- Sajeesh S
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015-3027, United States
| | - Andrew Camardo
- Cleveland Clinic Foundation, 9620 Carnegie Ave. Cleveland, Ohio 44106, United States
| | - Shataakshi Dahal
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015-3027, United States
| | - Anand Ramamurthi
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015-3027, United States
| |
Collapse
|
17
|
Fei X, Pan L, Yuan W, Zhao Y, Jiang L, Huang Q, Wu Y, Ru G. Papain Exerts an Anti-atherosclerosis Effect with Suppressed MPA-mediated Foam Cell Formation by Regulating the MAPK and PI3K/Akt-NF-κB Pathways. Expert Opin Ther Targets 2023; 27:239-250. [PMID: 36947095 DOI: 10.1080/14728222.2023.2194531] [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: 03/23/2023]
Abstract
BACKGROUND Papain possesses a potential anti-atherosclerosis (AS) effect. This study aimed to explore the inhibitory effects of papain on the monocyte-platelet aggregates (MPAs)-mediated production of foam cells in vitro and AS in vivo. RESEARCH DESIGN AND METHODS THP-1 cells were induced or treated by platelet, papain, nuclear factor-κB (NF-κB, p65) inhibitor, or NF-κB activator. An AS rat model was established and treated with papain. The THP-1 cells, macrophages, and foam cells were detected, and CD36, CD11b and CCR2 (macrophages) and CD14 and CD41 (MPAs) were measured. The levels of inflammatory factors, lipoprotein, and mitogen-activated protein kinase (MAPK, p38) and phosphoInositide-3 Kinase (PI3K)/Akt(protein kinase B, PKB)-NF-κB pathways proteins were determined. Finally, injury of the thoracic aorta of AS rats was observed. RESULTS Papain reduced macrophage production, lipid accumulation, and foam cell formation in vitro and downregulated the expression of monocyte chemoattractant protein 1 (MCP-1), prostaglandin E2 (PGE2), and cyclooxygenase 2 (COX2), and that of p38, c-Jun N-terminal protein kinase (JNK), Akt, and p65. Moreover, the inhibitory effects of papain were reversed by the NF-κB activator. Similarly, papain alleviated aortic smooth muscle hyperplasia, lipid droplet accumulation, and collagen diffusion and inhibited the secretion of inflammatory factors and the expression of p38, JNK, Akt, and p65 in vivo. CONCLUSIONS Papain inhibited MPA-induced foam cell formation by inactivating the MAPK and PI3K/Akt-NF-κB pathways, thereby exerting an anti-AS effect.
Collapse
Affiliation(s)
- Xianming Fei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China 310014
| | - Lianlian Pan
- Department of Laboratory Medicine, Sanmen People's Hospital of Taizhou, Zhejiang, China 317100
| | - Wufen Yuan
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China 310014
| | - Yan Zhao
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China 310014
| | - Lei Jiang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China 310014
| | - Qinghua Huang
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China 310014
| | - Yan Wu
- Department of Laboratory Medicine, Lin'an First People's Hospital of Hangzhou, Hangzhou, Zhejiang, China 311300
| | - Guoqing Ru
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China 310014
| |
Collapse
|
18
|
Zheng B, Wang Y, Hu J, Bao Z, Wang M. Comparative analysis of two cathepsin L genes in Asiatic hard clam (Meretrix meretrix): Similar in sequence features, different in expression profiles. FISH & SHELLFISH IMMUNOLOGY 2023; 133:108527. [PMID: 36621705 DOI: 10.1016/j.fsi.2023.108527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
Cathepsin L is widely found in eukaryotes and prokaryotes, and it plays important roles in innate immunity. In the present study, we cloned two cathepsin L genes (designated as MmCTSL1 and MmCTSL2, respectively) from Asiatic hard clam (Meretrix meretrix). The complete sequence of MmCTSL1 cDNA contained a 5' untranslated region (UTR) of 31 bp, a 3' UTR of 228 bp with a poly (A) tail, and an open reading frame (ORF) of 1005 bp encoding 334 amino acids with predicted molecular weight of 37.5 kDa and theoretical isoelectric point of 5.27, and contained a signal peptide (from M1 to A16), a protease inhibitor I29 family domain (from W27 to F87), and a papain family cysteine protease domain (from L118 to T333). The complete sequence of MmCTSL2 cDNA contained a 5' UTR of 50 bp, a 3' UTR of 162 bp with a poly (A) tail, and an ORF of 996 bp encoding a polypeptide of 331 amino acids with predicted molecular weight of 36.8 kDa and theoretical isoelectric point of 7.07. It contained a signal peptide (from M1 to A16), a protease inhibitor I29 family domain (from W30 to F89), and a papain family cysteine protease domain (from L115 to T330). Real-time quantitative PCR analysis demonstrated that MmCTSL1 and MmCTSL2 were widely expressed in all the tested tissues, including adductor muscle, foot, gill, hemocytes, hepatopancreas and mantle, with the highest mRNA expression level in hepatopancreas and hemocytes, respectively. After Vibrio splendidus challenge, the mRNA expression levels of MmCTSL1 and MmCTSL2 in hemocytes and hepatopancreas were both significantly up-regulated with different expression profiles. In hemocytes, the expression levels of MmCTSL1 and MmCTSL2 reached their respective peaks (3.4-fold and 13.0-fold compared with the control, respectively) at 12 h after bacterial challenge, and MmCTSL2 responds earlier than MmCTSL1. In hepatopancreas, the expression levels of MmCTSL1 and MmCTSL2 reached their respective peaks at 6 h (9.0-fold compared with the control) and 24 h (2.8-fold compared with the control) after bacterial challenge, meaning that MmCTSL1 responds earlier than MmCTSL2. At the same time, whether in hepatopancreas or hemocytes, MmCTSL1 persist for a while after the bacterial challenge peak, while MmCTSL2 would quickly return to the initial level after the bacterial challenge peak. These results indicate that cathepsin L may be involved in the immune process of hard clam against V. splendidus with different potential roles.
Collapse
Affiliation(s)
- Bo Zheng
- MOE Key Laboratory of Marine Genetics and Breeding (Qingdao 266003), and Key Laboratory of Tropical Aquatic Germplasm of Hainan Province of Sanya Oceanographic Institute (Sanya 572024), Ocean University of China, China
| | - Yan Wang
- MOE Key Laboratory of Marine Genetics and Breeding (Qingdao 266003), and Key Laboratory of Tropical Aquatic Germplasm of Hainan Province of Sanya Oceanographic Institute (Sanya 572024), Ocean University of China, China; Laboratory for Marine Fisheries Science and Food Production Processes, and Center for Marine Molecular Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - Jingjie Hu
- MOE Key Laboratory of Marine Genetics and Breeding (Qingdao 266003), and Key Laboratory of Tropical Aquatic Germplasm of Hainan Province of Sanya Oceanographic Institute (Sanya 572024), Ocean University of China, China; Laboratory for Marine Fisheries Science and Food Production Processes, and Center for Marine Molecular Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Hainan Yazhou Bay Seed Laboratory, Sanya, 572024, China
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding (Qingdao 266003), and Key Laboratory of Tropical Aquatic Germplasm of Hainan Province of Sanya Oceanographic Institute (Sanya 572024), Ocean University of China, China; Laboratory for Marine Fisheries Science and Food Production Processes, and Center for Marine Molecular Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Hainan Yazhou Bay Seed Laboratory, Sanya, 572024, China
| | - Mengqiang Wang
- MOE Key Laboratory of Marine Genetics and Breeding (Qingdao 266003), and Key Laboratory of Tropical Aquatic Germplasm of Hainan Province of Sanya Oceanographic Institute (Sanya 572024), Ocean University of China, China; Laboratory for Marine Fisheries Science and Food Production Processes, and Center for Marine Molecular Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Hainan Yazhou Bay Seed Laboratory, Sanya, 572024, China.
| |
Collapse
|
19
|
Tian Y, Liu S, Zhang Y, Yang J, Guo P, Zhang H, Yu X, Zou T. Immune infiltration and immunophenotyping in atrial fibrillation. Aging (Albany NY) 2023; 15:213-229. [PMID: 36602538 PMCID: PMC9876632 DOI: 10.18632/aging.204470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023]
Abstract
Atrial fibrillation (AF) is a relatively common arrhythmia in clinical practice. Although significant progress has been achieved in the treatment of AF and its associated complications, research on AF prevention lags behind, mainly due to the lack of a deep understanding of AF pathogenesis. In recent years, as our knowledge has grown, the role of the inflammatory/immune response in the occurrence and progression of AF has gradually gained attention. In this paper, based on existing gene expression data in the Gene Expression Omnibus database, a detailed description of immune infiltration status in AF is presented using a series of analytical methods, including differential analysis, Gene Ontology categorization, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, and weighted gene coexpression network analysis, and analysis tools such as CIBERSORTx and Cytoscape. Several new AF/immune infiltrations-related signature genes were identified, and the AF/immune infiltration pathology was classified based on these immune signature genes, thus providing novel insights into the pathogenesis of AF based on the inflammatory response.
Collapse
Affiliation(s)
- Yuqing Tian
- Department of Cardiology, Affiliated Hospital of Panzhihua University, Panzhihua 617000, Sichuan, P.R. China
- Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Shiying Liu
- Department of Plastic Surgery, Affiliated Hospital of Panzhihua University, Panzhihua 617000, Sichuan, P.R. China
| | - Yanan Zhang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China
| | - Jiefu Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| | - Peiyao Guo
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
- Peking University Fifth School of Clinical Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Hongchao Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| | - Tong Zou
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| |
Collapse
|
20
|
Biasizzo M, Javoršek U, Vidak E, Zarić M, Turk B. Cysteine cathepsins: A long and winding road towards clinics. Mol Aspects Med 2022; 88:101150. [PMID: 36283280 DOI: 10.1016/j.mam.2022.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022]
Abstract
Biomedical research often focuses on properties that differentiate between diseased and healthy tissue; one of the current focuses is elevated expression and altered localisation of proteases. Among these proteases, dysregulation of cysteine cathepsins can frequently be observed in inflammation-associated diseases, which tips the functional balance from normal physiological to pathological manifestations. Their overexpression and secretion regularly exhibit a strong correlation with the development and progression of such diseases, making them attractive pharmacological targets. But beyond their mostly detrimental role in inflammation-associated diseases, cysteine cathepsins are physiologically highly important enzymes involved in various biological processes crucial for maintaining homeostasis and responding to different stimuli. Consequently, several challenges have emerged during the efforts made to translate basic research data into clinical applications. In this review, we present both physiological and pathological roles of cysteine cathepsins and discuss the clinical potential of cysteine cathepsin-targeting strategies for disease management and diagnosis.
Collapse
Affiliation(s)
- Monika Biasizzo
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Miki Zarić
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
21
|
Jiang L, Xu C, Zhao Y, Huang Q, Yuan W, Wu Y, Fei X. Papain ameliorates monocyte-platelet aggregate formation-mediated inflammatory responses in monocytes by upregulating miRNA-146a transcription. PLoS One 2022; 17:e0278059. [PMID: 36409752 PMCID: PMC9678272 DOI: 10.1371/journal.pone.0278059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/09/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND MicroRNA-146a (miRNA-146a) is a nuclear factor κB (NF-κB)-inducible and inflammation-sensitive miRNA, while papain elicits anti-inflammatory effects by inhibiting monocyte-platelet aggregate (MPA)-mediated NF-κB pathway activation in monocytes. This study aimed to demonstrate the underlying effects of papain on MPA formation-initiated miRNA-146a expression and subsequent action in monocytes. METHODS THP-1 cells were exposed to papain, miRNA-146a mimic and inhibitor, NF-κB inhibitor (BAY11-7082), and platelets. Flow cytometry was used to measure the MPA formation-initiated monocyte activation. Levels of miRNA-146a, cyclooxygenase 2 (COX-2) mRNA and protein, and monocyte chemoattractant protein 1 (MCP-1) were analyzed in monocytes by RT-PCR, western blot, and ELISA. RESULTS The NF-κB inhibitor and miRNA-146a mimics upregulated miRNA-146a expression but suppressed subsequent monocyte activation and expression of COX-2 and MCP-1. Following exposure to papain, the enhanced miRNA-146a transcription induced by MPA-formation was found along with significant inhibition of monocyte activation in a dose-dependent manner. However, the inhibitory tendency was significantly reversed by miRNA-146a inhibitors. Expression of COX-2 mRNA and protein, as well as MCP-1, was inhibited in monocytes by papain, whereas miRNA-146a inhibitors promoted COX-2 and MCP-1 expression. CONCLUSION Our findings suggest that papain can inhibit MPA formation-mediated expression of inflammatory mediators in activated monocytes by upregulating miRNA-146a transcription.
Collapse
Affiliation(s)
- Lei Jiang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chan Xu
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yan Zhao
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qinghua Huang
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wufeng Yuan
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Wu
- Department of Laboratory Medicine, Lin’an First People’s Hospital of Hangzhou, Hangzhou, Zhejiang, China
| | - Xianming Fei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Mijanović O, Jakovleva A, Branković A, Zdravkova K, Pualic M, Belozerskaya TA, Nikitkina AI, Parodi A, Zamyatnin AA. Cathepsin K in Pathological Conditions and New Therapeutic and Diagnostic Perspectives. Int J Mol Sci 2022; 23:ijms232213762. [PMID: 36430239 PMCID: PMC9698382 DOI: 10.3390/ijms232213762] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022] Open
Abstract
Cathepsin K (CatK) is a part of the family of cysteine proteases involved in many important processes, including the degradation activity of collagen 1 and elastin in bone resorption. Changes in levels of CatK are associated with various pathological conditions, primarily related to bone and cartilage degradation, such as pycnodysostosis (associated with CatK deficiency), osteoporosis, and osteoarthritis (associated with CatK overexpression). Recently, the increased secretion of CatK is being highly correlated to vascular inflammation, hypersensitivity pneumonitis, Wegener granulomatosis, berylliosis, tuberculosis, as well as with tumor progression. Due to the wide spectrum of diseases in which CatK is involved, the design and validation of active site-specific inhibitors has been a subject of keen interest in pharmaceutical companies in recent decades. In this review, we summarized the molecular background of CatK and its involvement in various diseases, as well as its clinical significance for diagnosis and therapy.
Collapse
Affiliation(s)
- Olja Mijanović
- Dia-M, LCC, 7 b.3 Magadanskaya Str., 129345 Moscow, Russia
- The Human Pathology Department, Sechenov First Moscow State University, 119991 Moscow, Russia
| | | | - Ana Branković
- Department of Forensics Engineering, University of Criminal Investigation and Police Studies, Cara Dusana 196, 11000 Belgrade, Serbia
| | - Kristina Zdravkova
- AD Alkaloid Skopje, Boulevar Alexander the Great 12, 1000 Skopje, North Macedonia
| | - Milena Pualic
- Institute Cardiovascular Diseases Dedinje, Heroja Milana Tepica 1, 11000 Belgrade, Serbia
| | - Tatiana A. Belozerskaya
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia
| | - Angelina I. Nikitkina
- ArhiMed Clinique for New Medical Technologies, Vavilova St. 68/2, 119261 Moscow, Russia
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Andrey A. Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7X, UK
- Correspondence: ; Tel.: +7-9261180220
| |
Collapse
|
23
|
Su J, Cheng J, Hu Y, Yu Q, Li Z, Li J, Zheng N, Zhang Z, Yang J, Li X, Zhang Z, Wang Y, Zhu K, Du W, Chen X. Transfer RNA-derived small RNAs and their potential roles in the therapeutic heterogeneity of sacubitril/valsartan in heart failure patients after acute myocardial infarction. Front Cardiovasc Med 2022; 9:961700. [PMID: 36247465 PMCID: PMC9558900 DOI: 10.3389/fcvm.2022.961700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/14/2022] [Indexed: 12/05/2022] Open
Abstract
BackgroundIt has been reported that sacubitril/valsartan can improve cardiac function in acute myocardial infarction (AMI) patients complicated by heart failure (HF). However, a number of patients cannot be treated successfully; this phenomenon is called sacubitril/valsartan resistance (SVR), and the mechanisms remain unclear.MethodsIn our present research, the expression profiles of transfer RNA (tRNA)-derived small RNAs (tsRNAs) in SVR along with no sacubitril/valsartan resistance (NSVR) patients were determined by RNA sequencing. Through bioinformatics, quantitative real-time PCR (qRT-PCR), and cell-based experiments, we identified SVR-related tsRNAs and confirmed their diagnostic value, predicted their targeted genes, and explored the enriched signal pathways as well as regulatory roles of tsRNAs in SVR.ResultsOur research indicated that 36 tsRNAs were upregulated and that 21 tsRNAs were downregulated in SVR. Among these tsRNAs, the expression of tRF-59:76-Tyr-GTA-2-M3 and tRF-60:76-Val-AAC-1-M5 was upregulated, while the expression of tRF-1:29-Gly-GCC-1 was downregulated in the group of SVR. Receiver operating characteristic (ROC) curve analysis demonstrated that these three tsRNAs were potential biomarkers of the therapeutic heterogeneity of sacubitril/valsartan. Moreover, tRF-60:76-Val-AAC-1-M5 might target Tnfrsf10b and Bcl2l1 to influence the observed therapeutic heterogeneity through the lipid and atherosclerosis signaling pathways.ConclusionHence, tsRNA might play a vital role in SVR. These discoveries provide new insights for the mechanistic investigation of responsiveness to sacubitril/valsartan.
Collapse
Affiliation(s)
- Jia Su
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
| | - Ji Cheng
- Department of Emergency, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Yingchu Hu
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
| | - Qinglin Yu
- Department of Traditional Chinese Internal Medicine, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
| | - Zhenwei Li
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
| | - Jiyi Li
- Department of Cardiology, Yuyao People’s Hospital of Zhejiang Province, Yuyao, Zhejiang, China
| | - Nan Zheng
- Department of Cardiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Zhaoxia Zhang
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
| | - Jin Yang
- Department of Geriatrics, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
| | - Xiaojing Li
- Department of Geriatrics, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
| | - Zeqin Zhang
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
| | - Yong Wang
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
| | - Keqi Zhu
- Department of Traditional Chinese Internal Medicine, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- *Correspondence: Keqi Zhu,
| | - Weiping Du
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
- Weiping Du,
| | - Xiaomin Chen
- Department of Cardiology, Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
- Xiaomin Chen,
| |
Collapse
|
24
|
Association between triglyceride glucose index, coronary artery calcification and multivessel coronary disease in Chinese patients with acute coronary syndrome. Cardiovasc Diabetol 2022; 21:187. [PMID: 36114495 PMCID: PMC9482257 DOI: 10.1186/s12933-022-01615-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Background Multivessel coronary disease (MVCD) is the common type of coronary artery disease in acute coronary syndrome (ACS). Coronary artery calcification (CAC) has been confirmed the strong predictor of major adverse cardiovascular events (MACEs). Several studies have validated that triglyceride glucose (TyG) index can reflect the degree of coronary calcification or predict MACEs. However, no evidence to date has elucidated and compared the predictive intensity of TyG index or/and coronary artery calcification score (CACS) on multi-vascular disease and MACEs in ACS patients. Methods A total of 935 patients, diagnosed with ACS and experienced coronary computed tomography angiography (CCTA) from August 2015 to March 2022 in the Second Hospital of Shandong University, were selected for retrospective analysis. The subjects were divided into TyG index quartile 1–4 groups (Q1-Q4 groups), non-multivessel coronary disease (non-MVCD) and multivessel coronary disease (MVCD) groups, respectively. The general data, past medical or medication history, laboratory indicators, cardiac color Doppler ultrasound, CACS, and TyG indexes were respectively compared among these groups. The ROC curve preliminarily calculated and analyzed the diagnostic value of TyG index, CACS, and the combination of the two indicators for MVCD. Univariate and multivariate logistic regression analysis discriminated the independent hazard factors for forecasting MVCD. Results Compared with the lower TyG index and non-MVCD groups, the higher TyG index and MVCD groups had higher values of age, smoking history, waist circumference, systolic blood pressure, low-density lipoprotein cholesterol(LDL-C), fasting blood glucose and glycosylated hemoglobin, and CACS, but lower values of high-density lipoprotein cholesterol(HDL-C) (all P < 0.01). Coronary artery calcification is more common in the left anterior descending artery. Compared with non-MVCD, each unit increase in TyG index was associated with a 1.213-fold increased risk of MVCD. Logistic regression analysis adjusted for potential confounders indicated that TyG index is an independent risk factor for MVCD. With the increase of TyG index, the incidence of MACEs, apart from all-cause death, cardiac death, unexpected re-hospitalization of heart failure, recurrent ACS or unplanned revascularization, and non-fatal stroke in coronary artery increased (P log-rank < 0.001). Conclusion TyG index could completely substitute for CACS as a reliable, practical, and independent indicator for predicting the severity and prognosis of MVCD in patients with ACS.
Collapse
|
25
|
Xu J, Zhong Y, Yin H, Linneman J, Luo Y, Xia S, Xia Q, Yang L, Huang X, Kang K, Wang J, Niu Y, Li L, Gou D. Methylation-mediated silencing of PTPRD induces pulmonary hypertension by promoting pulmonary arterial smooth muscle cell migration via the PDGFRB/PLCγ1 axis. J Hypertens 2022; 40:1795-1807. [PMID: 35848503 PMCID: PMC9451921 DOI: 10.1097/hjh.0000000000003220] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 05/15/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Pulmonary hypertension is a lethal disease characterized by pulmonary vascular remodeling and is mediated by abnormal proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs). Platelet-derived growth factor BB (PDGF-BB) is the most potent mitogen for PASMCs and is involved in vascular remodeling in pulmonary hypertension development. Therefore, the objective of our study is to identify novel mechanisms underlying vascular remodeling in pulmonary hypertension. METHODS We explored the effects and mechanisms of PTPRD downregulation in PASMCs and PTPRD knockdown rats in pulmonary hypertension induced by hypoxia. RESULTS We demonstrated that PTPRD is dramatically downregulated in PDGF-BB-treated PASMCs, pulmonary arteries from pulmonary hypertension rats, and blood and pulmonary arteries from lung specimens of patients with hypoxic pulmonary arterial hypertension (HPAH) and idiopathic PAH (iPAH). Subsequently, we found that PTPRD was downregulated by promoter methylation via DNMT1. Moreover, we found that PTPRD knockdown altered cell morphology and migration in PASMCs via modulating focal adhesion and cell cytoskeleton. We have demonstrated that the increase in cell migration is mediated by the PDGFRB/PLCγ1 pathway. Furthermore, under hypoxic condition, we observed significant pulmonary arterial remodeling and exacerbation of pulmonary hypertension in heterozygous PTPRD knock-out rats compared with the wild-type group. We also demonstrated that HET group treated with chronic hypoxia have higher expression and activity of PLCγ1 in the pulmonary arteries compared with wild-type group. CONCLUSION We propose that PTPRD likely plays an important role in the process of pulmonary vascular remodeling and development of pulmonary hypertension in vivo .
Collapse
Affiliation(s)
- Junhua Xu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Yanfeng Zhong
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Haoyang Yin
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - John Linneman
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yixuan Luo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Sijian Xia
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Qinyi Xia
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Xingtao Huang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Kang Kang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| |
Collapse
|
26
|
Bahar ME, Hwang JS, Ahmed M, Lai TH, Pham TM, Elashkar O, Akter KM, Kim DH, Yang J, Kim DR. Targeting Autophagy for Developing New Therapeutic Strategy in Intervertebral Disc Degeneration. Antioxidants (Basel) 2022; 11:antiox11081571. [PMID: 36009290 PMCID: PMC9405341 DOI: 10.3390/antiox11081571] [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: 07/18/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 12/25/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent cause of low back pain. IVDD is characterized by abnormal expression of extracellular matrix components such as collagen and aggrecan. In addition, it results in dysfunctional growth, senescence, and death of intervertebral cells. The biological pathways involved in the development and progression of IVDD are not fully understood. Therefore, a better understanding of the molecular mechanisms underlying IVDD could aid in the development of strategies for prevention and treatment. Autophagy is a cellular process that removes damaged proteins and dysfunctional organelles, and its dysfunction is linked to a variety of diseases, including IVDD and osteoarthritis. In this review, we describe recent research findings on the role of autophagy in IVDD pathogenesis and highlight autophagy-targeting molecules which can be exploited to treat IVDD. Many studies exhibit that autophagy protects against and postpones disc degeneration. Further research is needed to determine whether autophagy is required for cell integrity in intervertebral discs and to establish autophagy as a viable therapeutic target for IVDD.
Collapse
Affiliation(s)
- Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
| | - Trang Minh Pham
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
| | - Omar Elashkar
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
| | - Kazi-Marjahan Akter
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, GyeongNam, Korea
| | - Dong-Hee Kim
- Department of Orthopaedic Surgery, Institute of Health Sciences, Gyeongsang National University Hospital and Gyeongsang National University College of Medicine, Jinju 52727, GyeongNam, Korea
| | - Jinsung Yang
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, GyeongNam, Korea
- Correspondence: ; Tel.: +82-55-772-8054
| |
Collapse
|
27
|
Effects of Serum LDL-C, CysC, and D-D in Patients with Coronary Atherosclerotic Heart Disease. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:5771960. [PMID: 35800677 PMCID: PMC9256368 DOI: 10.1155/2022/5771960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/31/2022] [Indexed: 01/09/2023]
Abstract
Objective To investigate the effects of low-density lipoprotein cholesterol (LDL-C) and serum cystatin C (CysC) combined with D-dimer (D-D) on patients with coronary atherosclerotic heart disease (CHD). Methods 90 patients with CHD who were admitted to our hospital and diagnosed by coronary angiography (CAG) from February 2020 to June 2021 were selected as the study subjects. 90 patients were grouped according to different types and branches of coronary lesions, and 30 patients with outpatient health check-ups at the same period were selected as the control group, and the differences in serum LDL-C, CysC, and D-D levels between the groups were compared. The logistic regression model was built to explore risk factors affecting the occurrence of CHD. Also, receiver operating characteristic (ROC) curves were drawn to analyze the diagnostic value of LDL-C, CysC, and D-D in CHD. Results In the comparison of LDL-C, CysC, and D-D levels, CHD group > control group (P < 0.05); stable angina (SAP) group > unstable angina (UAP) group > acute myocardial infarction (AMI) group (P < 0.05); three-branch group > two-branch group > single-branch group (P < 0.05). The logistic regression model showed that high expression levels of LDL-C, CysC, and D-D, male gender, and combined hypertension were risk factors for CHD. The area under the curve (AUC) of the combination of LDL-C, CysC, and D-D was 0.868, and the sensitivity and specificity were 88.89% and 73.33%, respectively, which are higher than those in single diagnosis (P < 0.05). Conclusions LDL-C, CysC, and D-D are highly expressed in CHD samples, and the combination of the three is beneficial to enhance the diagnostic accuracy of clinical CHD.
Collapse
|
28
|
Montavon B, Winter LE, Gan Q, Arasteh A, Montaño AM. Mucopolysaccharidosis Type IVA: Extracellular Matrix Biomarkers in Cardiovascular Disease. Front Cardiovasc Med 2022; 9:829111. [PMID: 35620518 PMCID: PMC9127057 DOI: 10.3389/fcvm.2022.829111] [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: 12/04/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease (CVD) in Mucopolysaccharidosis Type IVA (Morquio A), signified by valvular disease and cardiac hypertrophy, is the second leading cause of death and remains untouched by current therapies. Enzyme replacement therapy (ERT) is the gold-standard treatment for MPS disorders including Morquio A. Early administration of ERT improves outcomes of patients from childhood to adulthood while posing new challenges including prognosis of CVD and ERT's negligible effect on cardiovascular health. Thus, having accurate biomarkers for CVD could be critical. Here we show that cathepsin S (CTSS) and elastin (ELN) can be used as biomarkers of extracellular matrix remodeling in Morquio A disease. We found in a cohort of 54 treatment naïve Morquio A patients and 74 normal controls that CTSS shows promising attributes as a biomarker in young Morquio A children. On the other hand, ELN shows promising attributes as a biomarker in adolescent and adult Morquio A. Plasma/urine keratan sulfate (KS), and urinary glycosaminoglycan (GAG) levels were significantly higher in Morquio A patients (p < 0.001) which decreased with age of patients. CTSS levels did not correlate with patients' phenotypic severity but differed significantly between patients (median range 5.45-8.52 ng/mL) and normal controls (median range 9.61-15.9 ng/mL; p < 0.001). We also studied α -2-macroglobulin (A2M), C-reactive protein (CRP), and circulating vascular cell adhesion molecule-1 (sVCAM-1) in a subset of samples to understand the relation between ECM biomarkers and the severity of CVD in Morquio A patients. Our experiments revealed that CRP and sVCAM-1 levels were lower in Morquio A patients compared to normal controls. We also observed a strong inverse correlation between urine/plasma KS and CRP (p = 0.013 and p = 0.022, respectively) in Morquio A patients as well as a moderate correlation between sVCAM-1 and CTSS in Morquio A patients at all ages (p = 0.03). As the first study to date investigating CTSS and ELN levels in Morquio A patients and in the normal population, our results establish a starting point for more elaborate studies in larger populations to understand how CTSS and ELN levels correlate with Morquio A severity.
Collapse
Affiliation(s)
- Brittany Montavon
- Department of Pediatrics, School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Linda E. Winter
- Department of Pediatrics, School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Qi Gan
- Department of Pediatrics, School of Medicine, Saint Louis University, St. Louis, MO, United States
| | | | - Adriana M. Montaño
- Department of Pediatrics, School of Medicine, Saint Louis University, St. Louis, MO, United States
- Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
29
|
BELCARO G, CESARONE MR, DUGALL M, CORSI M, HOSOI M, BAVERA PM, COTELLESE R, FERAGALLI B, IPPOLITO E. Effects of the collagen modulator Centellicum® and spinal elongation exercises on subclinical abdominal aneurysmal dilatation. ITALIAN JOURNAL OF VASCULAR AND ENDOVASCULAR SURGERY 2022. [DOI: 10.23736/s1824-4777.22.01537-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
30
|
Blann AD, Brown JE, Heitmar R. Angiogenesis, Metabolism, Endothelial and Platelet Markers in Diabetes and Cardiovascular Disease. Br J Biomed Sci 2022; 79:10313. [PMID: 35996503 PMCID: PMC9302542 DOI: 10.3389/bjbs.2022.10313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/15/2022] [Indexed: 12/31/2022]
Abstract
Introduction: Diabetes is a leading risk factor for cardiovascular disease (CVD), the pathophysiology of both being linked to metabolic, endothelial, renal, angiogenic and platelet abnormalities. We hypothesised that abnormalities in these systems are more adverse in those whose CVD is compounded by diabetes, compared to those with diabetes or CVD alone. Materials and methods: Serum or plasma from 66 patients with diabetes alone, 76 with CVD alone, and 70 with both diabetes and CVD i.e. diabetic cardiovascular disease, was probed for markers of angiogenesis [angiopoietin 1 and 2, vascular endothelial growth factor (VEGF) and endoglin], metabolic [soluble receptor for advanced glycation products (sRAGE), leptin, lipocalin-2, interleukin-8, and cystatin-C], the endothelium (von Willebrand factor, endothelial microparticles and soluble E selectin)], and the platelet (platelet microparticles and soluble P selectin) by ELISA, Luminex or flow cytometry. Results: VEGF (p = 0.04), von Willebrand factor (p = 0.001) and endothelial microparticles (p = 0.042) were all higher in diabetic cardiovascular disease than in diabetes alone and cardiovascular disease alone. Soluble E selectin was higher in diabetic cardiovascular disease than in diabetes alone (p = 0.045), whilst cystatin-C (p = 0.004) and soluble P selectin (p < 0.001) were higher in diabetes and diabetic cardiovascular disease than in cardiovascular disease alone. There were no differences in angiopoietin 1 or 2, endoglin, sRAGE, leptin, lipocalin-2, or interleukin-8. Conclusion: Angiopoietin 1 or 2, endoglin, sRAGE, leptin, lipocalin-2, interleukin-8, and cystatin-c cannot differentiate diabetes from cardiovascular disease, or both conditions combined. Our data point to a more adverse endothelial (von Willebrand factor, endothelial microparticles), and angiogenic profile (VEGF) in those with diabetic cardiovascular disease, supporting the view that this group should be targeted more aggressively.
Collapse
Affiliation(s)
- A. D. Blann
- School of Applied Sciences, Huddersfield University, Huddersfield, United Kingdom
- *Correspondence: A. D. Blann,
| | - J. E. Brown
- Department of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - R. Heitmar
- School of Applied Sciences, Huddersfield University, Huddersfield, United Kingdom
| |
Collapse
|
31
|
West M, Kirby A, Stewart RA, Blankenberg S, Sullivan D, White HD, Hunt D, Marschner I, Janus E, Kritharides L, Watts GF, Simes J, Tonkin AM. Circulating Cystatin C Is an Independent Risk Marker for Cardiovascular Outcomes, Development of Renal Impairment, and Long-Term Mortality in Patients With Stable Coronary Heart Disease: The LIPID Study. J Am Heart Assoc 2022; 11:e020745. [PMID: 35179040 PMCID: PMC9075058 DOI: 10.1161/jaha.121.020745] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background Elevated plasma cystatin C levels reflect reduced renal function and increased cardiovascular risk. Less is known about whether the increased risk persists long‐term or is independent of renal function and other important biomarkers. Methods and Results Cystatin C and other biomarkers were measured at baseline (in 7863 patients) and 1 year later (in 6106 patients) in participants in the LIPID (Long‐Term Intervention with Pravastatin in Ischemic Disease) study, who had a previous acute coronary syndrome. Outcomes were ascertained during the study (median follow‐up, 6 years) and long‐term (median follow‐up, 16 years). Glomerular filtration rate (GFR) was estimated using Chronic Kidney Disease Epidemiology Collaboration equations (first GFR‐creatinine, then GFR‐creatinine‐cystatin C). Over 6 years, in fully adjusted multivariable time‐to‐event models, with respect to the primary end point of coronary heart disease mortality or nonfatal myocardial infarction, for comparison of Quartile 4 versus 1 of baseline cystatin C, the hazard ratio was 1.37 (95% CI, 1.07–1.74; P=0.01), and for major cardiovascular events was 1.47 (95% CI, 1.19–1.82; P<0.001). Over 16 years, the association of baseline cystatin C with coronary heart disease, cardiovascular, and all‐cause mortality persisted (each P<0.001) and remained significant after adjustment for estimated GFR‐creatinine‐cystatin C. Cystatin C also predicted the development of chronic kidney disease for 6 years (odds ratio, 6.61; 95% CI, 4.28–10.20) independently of estimated GFR‐creatinine and other risk factors. However, this association was no longer significant after adjustment for estimated GFR‐creatinine‐cystatin C. Conclusions Cystatin C independently predicted major cardiovascular events, development of chronic kidney disease, and cardiovascular and all‐cause mortality. Prediction of long‐term mortality was independent of improved estimation of GFR. Registration URL: https://anzctr.org.au; Unique identifier: ACTRN12616000535471.
Collapse
Affiliation(s)
- Malcolm West
- Department of MedicineUniversity of QueenslandBrisbaneAustralia
| | - Adrienne Kirby
- National Health and Medical Research Council Clinical Trials CentreUniversity of SydneySydneyAustralia
| | - Ralph A. Stewart
- Green Lane Cardiovascular ServiceAuckland City HospitalUniversity of AucklandAucklandNew Zealand
| | | | - David Sullivan
- Department of Chemical PathologyRoyal Prince Alfred HospitalSydneyAustralia
| | - Harvey D. White
- Green Lane Cardiovascular ServiceAuckland City HospitalUniversity of AucklandAucklandNew Zealand
| | - David Hunt
- Cardiology DepartmentRoyal Melbourne HospitalMelbourneAustralia
| | - Ian Marschner
- National Health and Medical Research Council Clinical Trials CentreUniversity of SydneySydneyAustralia
| | - Edward Janus
- Department of MedicineWestern Health Chronic Disease AllianceWestern HealthMelbourne Medical SchoolUniversity of MelbourneMelbourneAustralia
| | - Leonard Kritharides
- Department of CardiologyConcord Repatriation General HospitalSydney Local Health DistrictSydneyAustralia
- ANZAC Medical Research InstituteFaculty of MedicineUniversity of SydneySydneyAustralia
| | - Gerald F. Watts
- School of MedicineFaculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
| | - John Simes
- National Health and Medical Research Council Clinical Trials CentreUniversity of SydneySydneyAustralia
| | - Andrew M. Tonkin
- School of Public Health and Preventive MedicineMonash UniversityPerthAustralia
| | | |
Collapse
|
32
|
Davidson WS, Shah AS, Sexmith H, Gordon SM. The HDL Proteome Watch: Compilation of studies leads to new insights on HDL function. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159072. [PMID: 34800735 PMCID: PMC8715479 DOI: 10.1016/j.bbalip.2021.159072] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW High density lipoproteins (HDL) are a heterogeneous family of particles that contain distinct complements of proteins that define their function. Thus, it is important to accurately and sensitively identify proteins associated with HDL. Here we highlight the HDL Proteome Watch Database which tracks proteomics studies from different laboratories across the world. RECENT FINDINGS In 45 published reports, almost 1000 individual proteins have been detected in preparations of HDL. Of these, 251 have been identified in at least three different laboratories. The known functions of these consensus HDL proteins go well beyond traditionally recognized roles in lipid transport with many proteins pointing to HDL functions in innate immunity, inflammation, cell adhesion, hemostasis and protease regulation, and even vitamin and metal binding. SUMMARY The HDL proteome derived across multiple studies using various methodologies provides confidence in protein identifications that can offer interesting new insights into HDL function. We also point out significant issues that will require additional study going forward.
Collapse
Affiliation(s)
- W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, United States of America.
| | - Amy S Shah
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, OH 45229, United States of America.
| | - Hannah Sexmith
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, OH 45229, United States of America.
| | - Scott M Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America.
| |
Collapse
|
33
|
Cystatin C and cystatin SN as possible soluble tumor markers in malignant uveal melanoma. Radiol Oncol 2021; 56:83-91. [PMID: 34957724 PMCID: PMC8884861 DOI: 10.2478/raon-2021-0049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/21/2021] [Indexed: 11/22/2022] Open
Abstract
Background The aim of the study was to determine the concentration of endogenous cystatin C and cystatin SN, as potential tumor biomarkers, in the serum and biological fluids of the eye in both healthy controls and patients with uveal melanoma. Patients and methods The concentration of both cystatins was determined in the intraocular fluid (IOF), tear fluid, and serum of patients with uveal melanoma and compared to baseline measurements in IOF, tears, serum, cerebral spinal fluid, saliva and urine of healthy controls. Results The concentration of cystatin C in all the biological matrices obtained from healthy controls significantly exceeded the concentration of cystatin SN and was independent of gender. Cystatin C concentrations in the tear fluid of patients with uveal melanoma (both the eye with the malignancy, as well as the contralateral, non-affected eye), were significantly greater than cystatin C concentrations in the tear fluid of healthy controls and was independent of tumor size. The concentration of cystatin SN in IOF of patients with uveal melanoma was significantly less than the corresponding concentration of cystatin SN in healthy controls. Conclusions The ratio of cystatins (CysC:CysSN) in both the serum and tear fluid, as well as the concentration of cystatin SN in IOF, would appear to strongly suggest the presence of uveal melanoma. It is further suggested that multiple diagnostic criteria be utilized if a patient is suspected of having uveal melanoma, such as determination of the cystatin C and cystatin SN concentrations in serum, tears, and IOF, ocular fundus and ultrasound imaging, and biopsy with histopathological evaluation.
Collapse
|
34
|
Yang H, Li H, Chen W, Mei Z, Yuan Y, Wang X, Chu L, Xu Y, Sun Y, Li D, Gao H, Zhan B, Li H, Yang X. Therapeutic Effect of Schistosoma japonicum Cystatin on Atherosclerotic Renal Damage. Front Cell Dev Biol 2021; 9:760980. [PMID: 34901005 PMCID: PMC8656285 DOI: 10.3389/fcell.2021.760980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/11/2021] [Indexed: 01/15/2023] Open
Abstract
Atherosclerosis is a chronic inflammation of the arterial vessel wall driven by lipid metabolism disorders. Although helminthic infection and their derivatives have been identified to attenuate the chronic inflammatory diseases, the immunomodulatory effect of recombinant Schistosoma japonicum cystatin (rSj-Cys) on metabolic diseases and atherosclerosis has not been reported. In this study, we investigated the therapeutic efficacy of rSj-Cys on atherosclerotic renal damage and explored the related immunological mechanism. The results demonstrated that treatment with rSj-Cys significantly reduced body weight gain, hyperlipidemia, and atherosclerosis induced by the high-fat diet in apoE–/– mice. The treatment of rSj-Cys also significantly improved kidney functions through promoting macrophage polarization from M1 to M2, therefore inhibiting M1 macrophage–induced inflammation. The possible mechanism underlying the regulatory effect of rSj-Cys on reducing atherosclerosis and atherosclerotic renal damage is that rSj-Cys stimulates regulatory T cell and M2 macrophage polarization that produce regulatory cytokines, such as interleukin 10 and transforming growth factor β. The therapeutic effect of rSj-Cys on atherosclerotic renal damage is possibly through inhibiting the activation of TLR2/Myd88 signaling pathway. The results in this study provide evidence for the first time that Schistosoma-derived cystatin could be developed as a therapeutic agent to treat lipid metabolism disorder and atherosclerosis that threats million lives around the world.
Collapse
Affiliation(s)
- Huijuan Yang
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Hongqi Li
- Department of Gerontology, Anhui Provincial Hospital, First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Weidong Chen
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijie Mei
- Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Liang Chu
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yu Xu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Yan Sun
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Dingru Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Hongyu Gao
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Basic Medical College of Bengbu Medical College, Bengbu, China
| |
Collapse
|
35
|
Cathepsin C Is Involved in Macrophage M1 Polarization via p38/MAPK Pathway in Sudden Cardiac Death. Cardiovasc Ther 2021; 2021:6139732. [PMID: 34737793 PMCID: PMC8536465 DOI: 10.1155/2021/6139732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/27/2021] [Indexed: 12/28/2022] Open
Abstract
This study was aimed at identifying molecular markers associated with the pathogenesis of sudden cardiac death (SCD). It provides a proteomic analysis of human left anterior descending coronary artery from subjects diagnosed with SCD through histological examination and cases of nondisease accidental deaths through autopsy. A total of 2784 proteins were obtained from label-free quantitative proteomic analysis. This included a total of 265 differential proteins which were involved in SCD-related processes, such as inflammation, muscle system process regulation, metal ion transport, and lysosomal pathway. Western blotting was carried out to measure the expressions of cathepsin C (CTSC), focal adhesion kinase (FAK), p-FAK, and proteins related to the p38/MAPK signaling pathway, whereas immunohistochemistry was performed to determine the localization and expression of CTSC, TNF-α, and CD206 in arterial tissues. It was found that CTSC were the most expressed proteins with a significant upward trend in SCD cases. Besides, CTSC regulated macrophage polarization to M1 through the FAK-induced p38/MAPK signaling pathway. This promoted the release of inflammatory factors and eventually increased the inflammatory response. In conclusion, this study implies that CTSC may be one of the key molecular targets for promoting macrophage M1 polarization in SCD, which may provide new therapeutic insights into the treatment of inflammatory diseases.
Collapse
|
36
|
Zhu J, Yang G. H 2S signaling and extracellular matrix remodeling in cardiovascular diseases: A tale of tense relationship. Nitric Oxide 2021; 116:14-26. [PMID: 34428564 DOI: 10.1016/j.niox.2021.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network that not only provides mechanical support but also transduces essential molecular signals in organ functions. ECM is constantly remodeled to control tissue homeostasis, responsible for cell adhesion, cell migration, cell-to-cell communication, and cell differentiation, etc. The dysregulation of ECM components contributes to various diseases, including cardiovascular diseases, fibrosis, cancer, and neurodegenerative diseases, etc. Aberrant ECM remodeling is initiated by various stress, such as oxidative stress, inflammation, ischemia, and mechanical stress, etc. Hydrogen sulfide (H2S) is a gasotransmitter that exhibits a wide variety of cytoprotective and physiological functions through its anti-oxidative and anti-inflammatory actions. Amounting research shows that H2S can attenuate aberrant ECM remodeling. In this review, we discussed the implications and mechanisms of H2S in the regulation of ECM remodeling in cardiovascular diseases, and highlighted the potential of H2S in the prevention and treatment of cardiovascular diseases through attenuating adverse ECM remodeling.
Collapse
Affiliation(s)
- Jiechun Zhu
- School of Biological, Chemical & Forensic Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Guangdong Yang
- School of Biological, Chemical & Forensic Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada.
| |
Collapse
|
37
|
Iwata H, Osborn EA, Ughi GJ, Murakami K, Goettsch C, Hutcheson JD, Mauskapf A, Mattson PC, Libby P, Singh SA, Matamalas J, Aikawa E, Tearney GJ, Aikawa M, Jaffer FA. Highly Selective PPARα (Peroxisome Proliferator-Activated Receptor α) Agonist Pemafibrate Inhibits Stent Inflammation and Restenosis Assessed by Multimodality Molecular-Microstructural Imaging. J Am Heart Assoc 2021; 10:e020834. [PMID: 34632804 PMCID: PMC8751880 DOI: 10.1161/jaha.121.020834] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND New pharmacological approaches are needed to prevent stent restenosis. This study tested the hypothesis that pemafibrate, a novel clinical selective PPARα (peroxisome proliferator‐activated receptor α) agonist, suppresses coronary stent‐induced arterial inflammation and neointimal hyperplasia. METHODS AND RESULTS Yorkshire pigs randomly received either oral pemafibrate (30 mg/day; n=6) or control vehicle (n=7) for 7 days, followed by coronary arterial implantation of 3.5 × 12 mm bare metal stents (2–4 per animal; 44 stents total). On day 7, intracoronary molecular‐structural near‐infrared fluorescence and optical coherence tomography imaging was performed to assess the arterial inflammatory response, demonstrating that pemafibrate reduced stent‐induced inflammatory protease activity (near‐infrared fluorescence target‐to‐background ratio: pemafibrate, median [25th‐75th percentile]: 2.8 [2.5–3.3] versus control, 4.1 [3.3–4.3], P=0.02). At day 28, animals underwent repeat near‐infrared fluorescence–optical coherence tomography imaging and were euthanized, and coronary stent tissue molecular and histological analyses. Day 28 optical coherence tomography imaging showed that pemafibrate significantly reduced stent neointima volume (pemafibrate, 43.1 [33.7–54.1] mm3 versus control, 54.2 [41.2–81.1] mm3; P=0.03). In addition, pemafibrate suppressed day 28 stent‐induced cellular inflammation and neointima expression of the inflammatory mediators TNF‐α (tumor necrosis factor‐α) and MMP‐9 (matrix metalloproteinase 9) and enhanced the smooth muscle differentiation markers calponin and smoothelin. In vitro assays indicated that the STAT3 (signal transducer and activator of transcription 3)–myocardin axes mediated the inhibitory effects of pemafibrate on smooth muscle cell proliferation. CONCLUSIONS Pemafibrate reduces preclinical coronary stent inflammation and neointimal hyperplasia following bare metal stent deployment. These results motivate further trials evaluating pemafibrate as a new strategy to prevent clinical stent restenosis.
Collapse
Affiliation(s)
- Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA.,Department of Cardiovascular Biology and Medicine Juntendo University Graduate School of Medicine Tokyo Japan
| | - Eric A Osborn
- Cardiovascular Research CenterCardiology DivisionMassachusetts General HospitalHarvard Medical School Boston MA.,Cardiology Division Beth Israel Deaconess Medical CenterHarvard Medical School Boston MA
| | - Giovanni J Ughi
- Wellman Center for Photomedicine Massachusetts General HospitalHarvard Medical School Boston MA
| | - Kentaro Murakami
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Claudia Goettsch
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Adam Mauskapf
- Cardiovascular Research CenterCardiology DivisionMassachusetts General HospitalHarvard Medical School Boston MA
| | - Peter C Mattson
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Peter Libby
- Center for Excellence in Vascular Biology Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Joan Matamalas
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA.,Center for Excellence in Vascular Biology Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA.,Department of Human Pathology I.M. Sechenov First Moscow State Medical University of the Ministry of Health Moscow Russian Federation
| | - Guillermo J Tearney
- Wellman Center for Photomedicine Massachusetts General HospitalHarvard Medical School Boston MA.,Department of Pathology Massachusetts General HospitalHarvard Medical School Boston MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA.,Center for Excellence in Vascular Biology Cardiovascular Division Brigham and Women's Hospital Harvard Medical School Boston MA.,Channing Division of Network Medicine Brigham and Women's HospitalHarvard Medical School Boston MA
| | - Farouc A Jaffer
- Cardiovascular Research CenterCardiology DivisionMassachusetts General HospitalHarvard Medical School Boston MA.,Wellman Center for Photomedicine Massachusetts General HospitalHarvard Medical School Boston MA
| |
Collapse
|
38
|
Ding Y, Liu L, Chen Z, Li H, Pan Y, Wang J, Meng X, Lin J, Jing J, Xie X, Xiang X, Wang Y. Serum Cystatin C Predicts Stroke Clinical Outcomes at 1 Year Independent of Renal Function. Front Neurol 2021; 12:676872. [PMID: 34456843 PMCID: PMC8385197 DOI: 10.3389/fneur.2021.676872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/25/2021] [Indexed: 11/17/2022] Open
Abstract
Objective: Serum cystatin C (CysC) is a sensitive marker of renal function to predict cardiovascular diseases. We aimed to investigate the predictive value of CysC for clinical outcomes independent of renal function in patients with acute ischemic stroke (AIS). Methods: We measured serum CysC levels in 10,256 AIS patients from Third China National Stroke Registry (CNSR-III). The primary outcome was a combination of all-cause mortality and major disability (modified Rankin scale score, 3–6). Secondary outcomes included stroke recurrence and combined vascular events at 1 year. Outcomes were analyzed using logistic regression and Cox proportional hazards models, respectively. Results: The median CysC of included patients was 0.95 mg/l (interquartile range, 0.83–1.10 mg/l). A U-shaped association was observed between CysC and primary outcome (all-cause mortality or major disability) [quartile (Q)1 vs. Q2: adjusted odds ratio (aOR) 1.29, 95% CI 1.06–1.58, p = 0.012; Q3 vs. Q2: aOR 1.12, 95% CI 0.93–1.35, p = 0.242; Q4 vs. Q2: aOR 1.35, 95% CI 1.10–1.65, p = 0.004]. A similar trend also existed in “preserved renal function” patients. Adding CysC to a model containing conventional risk factors improved the model performance with integrated discrimination improvement (IDI) of 0.13% (p = 0.016) and net reclassification index (NRI) of 13.10% (p <0.001) for primary outcome. No significant association was observed for stroke recurrence or combined vascular event rate in different CysC quartiles. Conclusions: CysC showed a U-shaped correlation with 1-year stroke clinical outcome, suggesting that serum CysC may not only be a simple candidate marker of renal function.
Collapse
Affiliation(s)
- Yarong Ding
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Liping Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Zimo Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Hao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Junfeng Wang
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Jinxi Lin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Xuewei Xie
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Xianglong Xiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|
39
|
Zheng X, She HD, Zhang QX, Si T, Wu KS, Xiao YX. Cystatin C predicts the risk of incident cerebrovascular disease in the elderly: A meta-analysis on survival date studies. Medicine (Baltimore) 2021; 100:e26617. [PMID: 34260548 PMCID: PMC8284707 DOI: 10.1097/md.0000000000026617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 05/26/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Stroke is the third leading cause of global year of life lost in all-age and second-ranked cause of disability adjusted life years in middle-aged and elder population. Therefore, it is critical to study the relationship between vascular-related risk factors and cerebrovascular diseases. Several cross-sectional studies have shown that Cystatin C (Cys C) is an independent risk factor for cerebrovascular diseases and levels of Cys C are significantly higher in stroke patients than in healthy individuals. In this meta-analysis, we introduce a Cox proportional hazards model to evaluate the causality between Cys C and the risk of cerebrovascular accident in the elderly. METHODS We searched PubMed, EMBASE, the Web of Science, and the Cochrane Library from 1985 to 2019 for studies on the relationship between serum Cys C and incidence stroke with Cox proportional hazards models. We conducted a subgroup analysis of the selected studies to determine a connection between atherosclerosis and stroke. Finally, 7 research studies, including 26,768 patients without a history of cerebrovascular, were studied. RESULTS After comparing the maximum and minimum Cys C levels, the hazard ratio for all types of stroke, including ischemic and hemorrhagic stroke, was 1.18 (95% confidence interval 1.04-1.31) with moderate heterogeneity (I2 = 43.0%; P = .119) in a fixed-effect model after pooled adjustment for other potential risk factors. In the subgroup analysis, the hazard ratio and 95% confidence interval for Cys C stratified by atherosclerosis was 1.85 (0.97-2.72). As shown in Egger linear regression test, there was no distinct publication bias (P = .153). CONCLUSION Increased serum Cys C is significantly associated with future stroke events in the elderly, especially in patients with carotid atherosclerosis. Thus, serum levels of Cys C could serve as a predicted biomarker for stroke attack.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Hong-da She
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Qiao-xin Zhang
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Tong Si
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing
| | - Ku-sheng Wu
- Department of Public and preventive medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Ying-xiu Xiao
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| |
Collapse
|
40
|
Zhang S, Li P, Xin M, Jin X, Zhao L, Nan Y, Cheng XW. Dipeptidyl peptidase-4 inhibition prevents lung injury in mice under chronic stress via the modulation of oxidative stress and inflammation. Exp Anim 2021; 70:541-552. [PMID: 34219073 PMCID: PMC8614009 DOI: 10.1538/expanim.21-0067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure to chronic psychosocial stress is a risk factor for various pulmonary diseases. In view of the essential role of dipeptidyl peptidase 4 (DPP4) in animal and human lung pathobiology, we investigated the role of DPP4 in stress-related lung injury in mice. Eight-week-old male mice were randomly divided into a non-stress group and a 2-week immobilization stress group. Non-stress control mice were left undisturbed. The mice subjected to immobilized stress were randomly assigned to the vehicle or the DPP4 inhibitor anagliptin for 2 weeks. Chronic stress reduced subcutaneous and inguinal adipose volumes and increased blood DPP4 levels. The stressed mice showed increased levels in the lungs of genes and/or proteins related to oxidative stress (p67phox, p47phox, p22phox and gp91phox), inflammation (monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, and intracellular adhesion molecule-1), apoptosis (caspase-3, -8, -9), senescence (p16INK4A, p21, and p53) and proteolysis (matrix metalloproteinase-2 to -9, cathepsin S/K, and tissue inhibitor of matrix metalloproteinase-1 and -2), and reduced levels of eNOS, Sirt1, and Bcl-2 proteins; and these effects were reversed by genetic and pharmacological inhibitions of DPP4. We then exposed human umbilical vein endothelial cells in vitro to hydrogen peroxide; anagliptin treatment was also observed to mitigate oxidative and inflammatory molecules in this setting. Anagliptin can improve lung injury in stressed mice, possibly by mitigating vascular inflammation, oxidative stress production, and proteolysis. DPP4 may become a new therapeutic target for chronic psychological stress-related lung disease in humans and animals.
Collapse
Affiliation(s)
- Shengming Zhang
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Ping Li
- State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union, Medical College
| | - Minglong Xin
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Xianglan Jin
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Longguo Zhao
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Yongshan Nan
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Xian Wu Cheng
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| |
Collapse
|
41
|
Patoulias D, Stavropoulos K, Imprialos K, Athyros V, Grassos H, Doumas M, Faselis C. Inflammatory Markers in Cardiovascular Disease; Lessons Learned and Future Perspectives. Curr Vasc Pharmacol 2021; 19:323-342. [PMID: 32188386 DOI: 10.2174/1570161118666200318104434] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) still remains the leading cause of morbidity and mortality worldwide. It is now established that inflammation plays a crucial role in atherosclerosis and atherothrombosis, and thus, it is closely linked to cardiovascular disease. OBJECTIVE The aim of the present review is to summarize and critically appraise the most relevant evidence regarding the potential use of inflammatory markers in the field of CVD. METHODS We conducted a comprehensive research of the relevant literature, searching MEDLINE from its inception until November 2018, primarily for meta-analyses, randomized controlled trials and observational studies. RESULTS Established markers of inflammation, mainly C-reactive protein, have yielded significant results both for primary and secondary prevention of CVD. Newer markers, such as lipoprotein-associated phospholipase A2, lectin-like oxidized low-density lipoprotein receptor-1, cytokines, myeloperoxidase, cell adhesion molecules, matrix metalloproteinases, and the CD40/CD40 ligand system, have been largely evaluated in human studies, enrolling both individuals from the general population and patients with established CVD. Some markers have yielded conflicting results; however, others are now recognized not only as promising biomarkers of CVD, but also as potential therapeutic targets, establishing the role of anti-inflammatory and pleiotropic drugs in CVD. CONCLUSION There is significant evidence regarding the role of consolidated and novel inflammatory markers in the field of diagnosis and prognosis of CVD. However, multimarker model assessment, validation of cut-off values and cost-effectiveness analyses are required in order for those markers to be integrated into daily clinical practice.
Collapse
Affiliation(s)
- Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Konstantinos Imprialos
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Vasilios Athyros
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Michael Doumas
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Charles Faselis
- VA Medical Center, and George Washington University, Washington, DC 20422, United States
| |
Collapse
|
42
|
Deficiency of cysteinyl cathepsin K suppresses the development of experimental intimal hyperplasia in response to chronic stress. J Hypertens 2021; 38:1514-1524. [PMID: 32205563 DOI: 10.1097/hjh.0000000000002424] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chronic psychological stress (CPS) is linked to cardiovascular disease initiation and progression. Given that cysteinyl cathepsin K (CatK) participates in vascular remodeling and atherosclerotic plaque growth in several animal models, we investigated the role of CatK in the development of experimental neointimal hyperplasia in response to chronic stress. METHODS AND RESULTS At first, male wild-type (CatK) mice that underwent carotid ligation injury were subjected to chronic immobilization stress. On postoperative and stressed day 14, the results demonstrated that stress accelerated injury-induced neointima hyperplasia. On day 4, stressed mice showed following: increased levels of monocyte chemoattractant protein-1, gp91phox, toll-like receptor-2 (TLR2), TLR4, and CatK mRNAs or/and proteins, oxidative stress production, aorta-derived smooth muscle cell (SMC) migration, and macrophage infiltration as well as targeted intracellular proliferating-related molecules. Stressed mice showed increased matrix metalloproteinase-2 (MMP-2) and MMP-9 mRNA expressions and activities and elastin disruption in the injured carotid arteries. Second, CatK and CatK deficiency (CatK) mice received ligation injury and stress to explore the role of CatK. The stress-induced harmful changes were prevented by CatK. Finally, CatK mice that had undergone ligation surgery were randomly assigned to one of two groups and administered vehicle or CatK inhibitor for 14 days. Pharmacological CatK intervention produced a vascular benefit. CONCLUSION These data indicate that CatK deletion protects against the development of experimental neointimal hyperplasia via the attenuation of inflammatory overaction, oxidative stress production, and VSMC proliferation, suggesting that CatK is a novel therapeutic target for the management of CPS-related restenosis after intravascular intervention therapies.
Collapse
|
43
|
Piao L, Li Y, Narisawa M, Shen X, Cheng XW. Role of Dipeptidyl Peptidase-4 in Atherosclerotic Cardiovascular Disease in Humans and Animals with Chronic Stress. Int Heart J 2021; 62:470-478. [PMID: 33994495 DOI: 10.1536/ihj.20-181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Exposure to psychosocial stress is a risk factor for cardiovascular disease, including vascular atherosclerosis-based cardiovascular disease (ACVD). Dipeptidyl peptidase-4 (DPP-4) is a complex enzyme that acts as a membrane-anchored cell surface exopeptidase. DPP-4 is upregulated in metabolic and inflammatory cardiovascular disorders. DPP-4 exhibits many physiological and pharmacological functions by regulating its extremely abundant substrates, such as glucagon-like peptide-1 (GLP-1). Over the last 10 years, emerging data have demonstrated unexpected roles of DPP-4 in extracellular and intracellular signaling, immune activation, inflammation, oxidative stress production, cell apoptosis, insulin resistance, and lipid metabolism. This mini-review focuses on recent novel findings in this field, highlighting a DPP-4-mediated regulation of GLP-1-dependent and -independent signaling pathways as a potential therapeutic molecular target in treatments of chronic psychological stress-related ACVD in humans and animals.
Collapse
Affiliation(s)
- Limei Piao
- Department of Cardiology, Yanbian University Hospital
| | - Yanglong Li
- Department of Cardiology, Yanbian University Hospital
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Xionghu Shen
- Department of Oncology, Yanbian University Hospital
| | - Xian Wu Cheng
- Department of Cardiology, Yanbian University Hospital
| |
Collapse
|
44
|
Bollavaram K, Leeman TH, Lee MW, Kulkarni A, Upshaw SG, Yang J, Song H, Platt MO. Multiple sites on SARS-CoV-2 spike protein are susceptible to proteolysis by cathepsins B, K, L, S, and V. Protein Sci 2021; 30:1131-1143. [PMID: 33786919 PMCID: PMC8138523 DOI: 10.1002/pro.4073] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/15/2021] [Accepted: 03/24/2021] [Indexed: 12/23/2022]
Abstract
SARS‐CoV‐2 is the coronavirus responsible for the COVID‐19 pandemic. Proteases are central to the infection process of SARS‐CoV‐2. Cleavage of the spike protein on the virus's capsid causes the conformational change that leads to membrane fusion and viral entry into the target cell. Since inhibition of one protease, even the dominant protease like TMPRSS2, may not be sufficient to block SARS‐CoV‐2 entry into cells, other proteases that may play an activating role and hydrolyze the spike protein must be identified. We identified amino acid sequences in all regions of spike protein, including the S1/S2 region critical for activation and viral entry, that are susceptible to cleavage by furin and cathepsins B, K, L, S, and V using PACMANS, a computational platform that identifies and ranks preferred sites of proteolytic cleavage on substrates, and verified with molecular docking analysis and immunoblotting to determine if binding of these proteases can occur on the spike protein that were identified as possible cleavage sites. Together, this study highlights cathepsins B, K, L, S, and V for consideration in SARS‐CoV‐2 infection and presents methodologies by which other proteases can be screened to determine a role in viral entry. This highlights additional proteases to be considered in COVID‐19 studies, particularly regarding exacerbated damage in inflammatory preconditions where these proteases are generally upregulated. PDB Code(s): 6VYB, 4Z2A, 5F02, 4P6E, 5TUN, 2IPP and 3H6S;
Collapse
Affiliation(s)
- Keval Bollavaram
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Tiffanie H Leeman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Maggie W Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Akhil Kulkarni
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Sophia G Upshaw
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Jiabei Yang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA.,Biomedical Engineering, Peking University, Beijing, China
| | - Hannah Song
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| | - Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia, USA
| |
Collapse
|
45
|
Kim KM, Lee JY, Jeon BH, Quan KT, Na M, Nam KW, Chae S. Extract of Curcuma zedoaria R. prevents atherosclerosis in apolipoprotein E-deficient mice. Nutr Res Pract 2021; 15:319-328. [PMID: 34093973 PMCID: PMC8155225 DOI: 10.4162/nrp.2021.15.3.319] [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: 05/04/2020] [Revised: 07/03/2020] [Accepted: 12/20/2020] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/OBJECTIVES Curcuma zedoaria R. (Zingiberaceae) has been used to treat headache, fever, and hypertension-related symptoms in Asian countries, including Korea, China, and Japan. We investigated whether dietary intake of a C. zedoaria extract (CzE) affected atherosclerosis in vivo. MATERIALS/METHODS Apolipoprotein E-deficient (ApoE−/−) mice (n = 32) were fed a normal diet (ND), a high-cholesterol diet (HCD), an HCD containing CzE (100 mg/kg/day), or an HCD containing simvastatin (10 mg/kg/day) for 12 weeks. The anti-atherosclerotic effects were evaluated by observing changes in fatty streak lesions, immunohistochemical analysis, ex vivo fluorescence imaging, lipid profiles, and western blot analysis. RESULTS The CzE-fed group showed a 41.6% reduction of atherosclerosis. Furthermore, CzE significantly reduced the levels of serum triglyceride, high-density lipoprotein, the chemokine (C-X3-C-motif) ligand 1, the adhesion molecules vascular cell adhesion molecule-1, intracellular adhesion molecule-1, and E-selectin; down-regulation of tumor necrosis factor-α, interleukin-6, high mobility group box-1, and cathepsin levels in the aortic sinuses and aortas of ApoE−/− mice were also observed. CONCLUSIONS The results suggest that the inclusion of a water extract of C. zedoaria in a HCD is closely correlated with reducing the risk of vascular inflammatory diseases in an ApoE mouse model.
Collapse
Affiliation(s)
- Ki Mo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.,Department of Korean Life Science and Technology, University of Science and Technology, Daejeon 34113, Korea
| | - Joo Young Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Byeong Hwa Jeon
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Khong Trong Quan
- Department of Pharmacognosy, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea
| | - MinKyun Na
- Department of Pharmacognosy, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea
| | - Kung-Woo Nam
- Department of Life Science and Biotechnology, Soonchunhyang University, Asan 31538, Korea
| | - Sungwook Chae
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.,Department of Korean Life Science and Technology, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
46
|
Yu C, Wan Y, Xu W, Jin X, Zhang S, Xin M, Jiang H, Cheng X. Increased Circulating Cathepsin L in Patients with Coronary Artery Disease. Int Heart J 2020; 62:9-15. [PMID: 33390563 DOI: 10.1536/ihj.20-182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cathepsin L (CatL) is a potent collagenase involved in atherosclerotic vascular remodeling and dysfunction in animals and humans. This study investigated the hypothesis that plasma CatL is associated with the prevalence of coronary artery disease (CAD). Between February May 2011 and January 2013, 206 consecutive subjects were enrolled from among patients who underwent coronary angiography and percutaneous coronary intervention treatment. Age-matched subjects (n = 215) served as controls. Plasma CatL and high-sensitive C-reactive protein (hs-CRP) and high-density lipoprotein cholesterol were measured. The patients with CAD had significantly higher plasma CatL levels compared to the controls (1.4 ± 0.4 versus 0.4 ± 0.2 ng/mL, P < 0.001), and the patients with acute coronary syndrome had significantly higher plasma CatL levels compared to those with stable angina pectoris (1.7 ± 0.7 versus 0.8 ± 0.4 ng/mL, P < 0.01). Linear regression analysis showed that overall, the plasma CatL levels were inversely correlated with the high-density lipoprotein levels (r = -0.32, P < 0.01) and positively with hs-CRP levels (r = 0.35, P < 0.01). Multiple logistic regression analyses shows that cathepsin L levels were independent predictors of CAD (add ratio, 1.8; 95% CI, 1.2 to 2.1; P < 0.01). These data demonstrated that increased levels of plasma CatL are closely associated with the presence of CAD and that circulating CatL serves as a useful biomarker for CAD.
Collapse
Affiliation(s)
- Chenglin Yu
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Ying Wan
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Wenhu Xu
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Xiongjie Jin
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Shengming Zhang
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Minglong Xin
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Haiying Jiang
- Department of Physiology and Pathophysiology, Jiaxing University Medical College
| | - Xianwu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital.,Department of Physiology and Pathophysiology, Jiaxing University Medical College
| |
Collapse
|
47
|
Yetkin E, Cuglan B, Turhan H, Yalta K. Where cystatin C acts: inside or outside of the plaque. Neurol Sci 2020; 41:3765-3766. [DOI: 10.1007/s10072-020-04538-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/21/2020] [Indexed: 10/24/2022]
|
48
|
Karakus A, Tütüncü A, Çamcı S, Uğuz B, Özmen G, Arı H, Demir M. A New Perspective for Isolated Coronary Artery Ectasia: Cystatin C. Cureus 2020; 12:e11053. [PMID: 33224650 PMCID: PMC7676437 DOI: 10.7759/cureus.11053] [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] [Indexed: 11/05/2022] Open
Abstract
Introduction The pathophysiology of isolated coronary artery ectasia (iCAE) has not been clearly identified, although multiple abnormalities, including arteritis, endothelial dysfunction, and vascular destruction, have been reported. In this study, we aimed to analyze serum cystatin C concentrations in patients with iCAE and controls. Methods Forty-seven patients with iCAE (mean age: 55.9 ± 11.5) and 32 individuals with normal coronary angiography (mean age: 57.8.1 ± 9.6) were included in the study. Plasma cystatin C levels were measured by using the principle of particle-enhanced turbidimetric immunoassay (PETIA). Results Serum cystatin C concentrations were significantly lower in patients with iCAE compared with the control group (0.98 ± 0.17 mg/L versus 1.17 ± 2.6 mg/L, p-value = 0.001). A significantly positive relationship was found between serum cystatin C levels and creatinine and high-sensitivity C-reactive protein (hs-CRP) levels in both groups (r-value = 0.288, p-value = 0.005, r-value = 0.143, p-value = 0.007, respectively). In multivariate logistic regression analysis, serum cystatin C level found to be a significant predictor for the presence of iCAE (OR: 0.837, CI: 95% (0.341 - 1.637), p-value = 0.013). Receiver operating characteristic (ROC) analysis determined that a cystatin C value < 1.02 mg/L had a sensitivity of 56% and a specificity of 78% for the prediction of ectasia. Conclusion We conclude that cystatin C independently can be a useful predictor for the presence of iCAE.
Collapse
|
49
|
Cathepsin K Deficiency Impaired Ischemia-Induced Neovascularization in Aged Mice. Stem Cells Int 2020; 2020:6938620. [PMID: 32676120 PMCID: PMC7346230 DOI: 10.1155/2020/6938620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022] Open
Abstract
Background Aging is a major risk factor for cardiovascular disease. Cysteine protease cathepsin K (CatK) has been implicated in the process of angiogenesis, but the exact roles of individual CatK in vessel formation during aging are poorly understood. Methods and Results To study the putative role of CatK in ischemia-induced angiogenesis, we applied a hindlimb ischemia model to aged wild-type (CatK+/+) and CatK-deficient (CatK−/−) mice. A serial laser Doppler blood-flow analysis revealed that the recovery of the ischemic/normal blood-flow ratio in the aged CatK−/−mice was impaired throughout the follow-up period. On postoperative day 14, CatK deficiency had also impaired capillary formation. CatK deficiency reduced the levels of cleaved Notch1, phospho-Akt, and/or vascular endothelial growth factor (VEGF) proteins in the ischemic muscles and bone marrow-derived c-Kit+ cells. A flow cytometry analysis revealed that CatK deficiency reduced the numbers of endothelial progenitor cell (EPC)-like CD31+/c-Kit+ cells in the peripheral blood as well as the ischemic vasculature. In vitro experiments, CatK−/− impaired bone-derived c-Kit+ cellular functions (migration, invasion, proliferation, and tubulogenesis) in aged mice. Our findings demonstrated that aging impaired the ischemia-induced angiogenesis associated with the reductions of the production and mobilization of CD31+/c-Kit+ cells in mice. Conclusions These findings established that the impairment of ischemia-induced neovascularization in aged CatK−/− mice is due, at least in part, to the reduction of EPC mobilization and the homing of the cells into vasculature that is associated with the impairment of Notch1 signaling activation at advanced ages.
Collapse
|
50
|
Yadati T, Houben T, Bitorina A, Shiri-Sverdlov R. The Ins and Outs of Cathepsins: Physiological Function and Role in Disease Management. Cells 2020; 9:cells9071679. [PMID: 32668602 PMCID: PMC7407943 DOI: 10.3390/cells9071679] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/14/2022] Open
Abstract
Cathepsins are the most abundant lysosomal proteases that are mainly found in acidic endo/lysosomal compartments where they play a vital role in intracellular protein degradation, energy metabolism, and immune responses among a host of other functions. The discovery that cathepsins are secreted and remain functionally active outside of the lysosome has caused a paradigm shift. Contemporary research has unraveled many versatile functions of cathepsins in extralysosomal locations including cytosol and extracellular space. Nevertheless, extracellular cathepsins are majorly upregulated in pathological states and are implicated in a wide range of diseases including cancer and cardiovascular diseases. Taking advantage of the differential expression of the cathepsins during pathological conditions, much research is focused on using cathepsins as diagnostic markers and therapeutic targets. A tailored therapeutic approach using selective cathepsin inhibitors is constantly emerging to be safe and efficient. Moreover, recent development of proteomic-based approaches for the identification of novel physiological substrates offers a major opportunity to understand the mechanism of cathepsin action. In this review, we summarize the available evidence regarding the role of cathepsins in health and disease, discuss their potential as biomarkers of disease progression, and shed light on the potential of extracellular cathepsin inhibitors as safe therapeutic tools.
Collapse
|