1
|
Shanmugasundaram BU, Stanely SP, Ponnian SMP. Protocatechuic acid attenuates isoproterenol-induced heart failure by modulating cardiac oxidative stress, LDL-R/SREBP-2/PPAR-α, and Bax/Bcl-2 /Bcl-xL/Cyt.c/ Caspase - 9 and Caspase - 3 pathways. Eur J Pharmacol 2025; 998:177492. [PMID: 40057156 DOI: 10.1016/j.ejphar.2025.177492] [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: 11/25/2024] [Revised: 02/04/2025] [Accepted: 03/06/2025] [Indexed: 03/21/2025]
Abstract
Worldwide, heart failure (HF) is among the top causes of morbidity and mortality. Curative interventions using natural products for cardiovascular diseases have gained substantial attention worldwide. At present, there is little research on the protective mechanisms of protocatechuic acid in HF. Hence, we evaluated the protective mechanisms of protocatechuic acid on cardiac oxidative stress, low-density lipoprotein-receptor (LDL-R)/sterol regulatory element-binding protein-2 (SREBP-2)/peroxisome proliferator-activated receptor-α(PPAR-α) and B-cell lymphoma-2 associated-x (Bax)/B-cell lymphoma gene-2(Bcl-2)/B-cell lymphoma-extra large(Bcl-xL)/Cytochrome c (Cyt.c)/Caspase-3/Caspase -9 pathways in isoproterenol-induced HF rats. Rats were induced HF by isoproterenol (5 mg/kg body weight) daily for 7 days. Then, rats were treated with protocatechuic acid (16 mg/kg body weight) for four weeks. The serum HF diagnostic marker and heart lipid hydroperoxides were increased and systolic and diastolic pressure and heart antioxidants were reduced in the isoproterenol-induced HF rats. Moreover, serum and heart total cholesterol, triglycerides, and serum low-density lipoprotein-cholesterol and liver 3-hydroxy-3 -methylglutaryl coenzyme A reductase were elevated and serum high density lipoprotein-cholesterol was reduced in the HF rats. Furthermore, lipid peroxidation products in the heart lysosomal fraction and lysosomal enzymes in the serum and heart were increased.A reverse transcription-polymerase chain reaction study revealed altered expression of hepatic LDL-R, SREBP-2, and PPAR- α and myocardial expression of Bax, Bcl-2, Bcl-xL, Cyt.c, Caspases-3, and -9 genes. Nevertheless, HF rats treated with protocatechuic acid considerably attenuated all the hemodynamic, biochemical, molecular, transmission electron microscopic, and histological parameters investigated and protected HF rats, by its potent antioxidant, anti-hyperlipidemic and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Bhavadharseny Uma Shanmugasundaram
- Medicinal and Biomolecular Chemistry Laboratory, Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, 608 002, Tamil Nadu, India
| | - Shervin Prince Stanely
- Division of Biotechnology, Karunya Institute of Technology and Sciences, Coimbatore, 641 114, Tamil Nadu, India
| | - Stanely Mainzen Prince Ponnian
- Medicinal and Biomolecular Chemistry Laboratory, Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, 608 002, Tamil Nadu, India.
| |
Collapse
|
2
|
Han X, Hua Z, Chen H, Yang J. Cathepsins and age-related macular degeneration: A Mendelian randomization study unveiling causal relationships. Medicine (Baltimore) 2025; 104:e42357. [PMID: 40355192 PMCID: PMC12073853 DOI: 10.1097/md.0000000000042357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/18/2025] [Indexed: 05/14/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision impairment and blindness in older adults, profoundly affecting millions of individuals worldwide. Cathepsins are a crucial class of proteolytic enzymes that participates in multiple biological process. However, the role of cathepsins in AMD still remains unclear. This study aims to probe into the causal relationship between cathepsins and AMD using a 2-sample Mendelian randomization (MR). Instrumental variables associated with exposure (cathepsins) and the outcome (AMD) were sourced from published genome-wide association studies. To estimate the causal effects, methodologies such as inverse variance weighted, MR-Egger, and weighted median estimation (WM) were employed. Reverse MR and multivariate MR analyses were also performed. The elevated levels of cathepsin B significantly increased the risk of dry AMD, with an odds ratio (OR) of 1.068 (95% CI = 1.007-1.133) and a P-value of .029). Sensitivity analyses confirmed the robustness of these findings, with no evidence of heterogeneity or pleiotropy. Reverse MR analyses indicated that total AMD might elevate levels of cathepsin E (OR = 1.04, P = .029). Multivariate MR analysis showed significant associations between specific cathepsins and AMD subtypes, including cathepsin G and cathepsin O with significantly increasing risk. The study revealed a potential causal effect of cathepsin B on AMD, especially dry AMD. These findings provide potential therapeutic targets for AMD, and further research is needed to understand the underlying mechanisms.
Collapse
Affiliation(s)
- Xiaoyan Han
- Department of Ophthalmology and the Eye Institute, Eye and Ear, Nose, and Throat Hospital, Fudan University, Shanghai, PR China
- Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, PR China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, PR China
| | - Zhixiang Hua
- Department of Ophthalmology and the Eye Institute, Eye and Ear, Nose, and Throat Hospital, Fudan University, Shanghai, PR China
- Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, PR China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, PR China
| | - Han Chen
- Department of Ophthalmology and the Eye Institute, Eye and Ear, Nose, and Throat Hospital, Fudan University, Shanghai, PR China
- Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, PR China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, PR China
| | - Jin Yang
- Department of Ophthalmology and the Eye Institute, Eye and Ear, Nose, and Throat Hospital, Fudan University, Shanghai, PR China
- Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, PR China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, PR China
| |
Collapse
|
3
|
Lu L, Wang L, Yang M, Wang H. Role of METTL16 in PPARγ methylation and osteogenic differentiation. Cell Death Dis 2025; 16:271. [PMID: 40210616 PMCID: PMC11986173 DOI: 10.1038/s41419-025-07527-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 02/11/2025] [Accepted: 03/12/2025] [Indexed: 04/12/2025]
Abstract
Osteoporosis, a prevalent bone disease, is characterized by the deterioration of bone tissue microstructure and imbalanced osteogenesis. The regulatory role of PPARγ m6A methylation mediated by METTL16 remains poorly elucidated. This study utilized advanced single-cell RNA sequencing (scRNA-seq) and Bulk RNA-seq techniques to explore how METTL16 influences the osteogenic differentiation of Bone Marrow-Derived Mesenchymal Stem Cells (BMSCs) and its implication in osteoporosis. The research revealed that METTL16 enhances the suppression of osteogenic differentiation in BMSCs, while PPARγ is associated with BMSC ferroptosis. Mechanistically, METTL16 facilitates the m6A modification of PPARγ transcription, thereby promoting ferroptosis in BMSCs and impeding their osteogenic differentiation. The in vivo animal experiments confirmed the pivotal role of the METTL16-PPARγ axis in osteoporosis development in mice. These findings suggest that the regulation of PPARγ m6A methylation by METTL16, leading to ferroptosis, is a critical mechanism impacting BMSC osteogenic differentiation and the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Liangjie Lu
- Department of Orthopedics, Ningbo Medical Center Li Huili Hospital, Li Huili Hospital Affiliated to Ningbo University, Ningbo, China.
| | - Lijun Wang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Minjie Yang
- Department of Orthopaedics, Jiu jiang NO.1 People's Hospital, Jiu jiang, China
| | - Huihan Wang
- Department of Orthopaedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Shannon K, Weiss-Sadan T, Merquiol E, Dey G, Gilon T, Turk B, Blum G. Novel Nucleus-Oriented Quenched Activity-Based Probes Link Cathepsin Nuclear Localization with Mitosis. ACS Sens 2025; 10:1321-1333. [PMID: 39960252 PMCID: PMC11877631 DOI: 10.1021/acssensors.4c03217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
Cysteine cathepsins are important proteases that are highly upregulated in cancers and other diseases. While their reported location is mostly endolysosomal, some evidence shows their nuclear localization and involvement in the cell cycle. We aim to generate tools to investigate the involvement of cathepsins in the cell cycle progression. To investigate nuclear cathepsin activity, we designed nucleus-directed quenched activity-based probes (qABPs) by attaching cell-penetrating peptides (CPPs). qABPs are active-site-directed compounds that enable direct real-time monitoring of enzyme activity by the covalent linkage between the probe and the enzyme's active site. Biochemical evaluation of the CPP-qABPs showed potent and selective probes; cell fractionation, multimodal flow cytometry-imaging, and time-lapse movies demonstrated nuclear cathepsin activity in living cells. Interestingly, these probes reveal a spatiotemporal pattern, a surge of nuclear cathepsin just before mitosis, suggesting yet unrevealed roles of cathepsin in cell division. In summary, these nuclear-directed qABPs serve as unique scientific tools to unlock the hidden features of cysteine proteases and to understand their involvement in cell division and cancer.
Collapse
Affiliation(s)
- Karin
Reut Shannon
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, POB 12271, Jerusalem 9112001, Israel
| | - Tommy Weiss-Sadan
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, POB 12271, Jerusalem 9112001, Israel
| | - Emmanuelle Merquiol
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, POB 12271, Jerusalem 9112001, Israel
| | - Gourab Dey
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, POB 12271, Jerusalem 9112001, Israel
| | - Tamar Gilon
- Azrieli
College of Engineering, 26 Yaakov Shreibom Street, Jerusalem 9103501, Israel
| | - Boris Turk
- Department
of Biochemistry and Molecular Biology, J.
Stefan Institute, Jamova
39, SI-1000 Ljubljana, Slovenia
- Faculty
of Chemistry and Chemical Technology, University
of Ljubljana, Večna
Pot 113, SI-1000 Ljubljana, Slovenia
| | - Galia Blum
- The
Institute for Drug Research, The School of Pharmacy, The Faculty of
Medicine, The Hebrew University, POB 12271, Jerusalem 9112001, Israel
- The
Wohl Institute for Translational Medicine, Hadassah Hospital, Kalman
Ya’akov Man Street , Jerusalem 9112001, Israel
| |
Collapse
|
5
|
Conesa-Bakkali R, Morillo-Huesca M, Martínez-Fábregas J. Non-Canonical, Extralysosomal Activities of Lysosomal Peptidases in Physiological and Pathological Conditions: New Clinical Opportunities for Cancer Therapy. Cells 2025; 14:68. [PMID: 39851495 PMCID: PMC11763575 DOI: 10.3390/cells14020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Lysosomes are subcellular compartments characterised by an acidic pH, containing an ample variety of acid hydrolases involved in the recycling of biopolymers. Among these hydrolases, lysosomal proteases have merely been considered as end-destination proteases responsible for the digestion of waste proteins, trafficked to the lysosomal compartment through autophagy and endocytosis. However, recent reports have started to unravel specific roles for these proteases in the regulation of initially unexpected biological processes, both under physiological and pathological conditions. Furthermore, some lysosomal proteases are no longer restricted to the lysosomal compartment, as more novel non-canonical, extralysosomal targets are being identified. Currently, lysosomal proteases are accepted to play key functions in the extracellular milieu, attached to the plasma membrane and even in the cytosolic and nuclear compartments of the cell. Under physiological conditions, lysosomal proteases, through non-canonical, extralysosomal activities, have been linked to cell differentiation, regulation of gene expression, and cell division. Under pathological conditions, these proteases have been linked to cancer, mostly through their extralysosomal activities in the cytosol and nuclei of cells. In this review, we aim to provide a comprehensive summary of our current knowledge about the extralysosomal, non-canonical functions of lysosomal proteases, both under physiological and pathological conditions, with a particular interest in cancer, that could potentially offer new opportunities for clinical intervention.
Collapse
Affiliation(s)
- Ryan Conesa-Bakkali
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Macarena Morillo-Huesca
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Jonathan Martínez-Fábregas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Biología, Universidad de Sevilla, Avenida Reina Mercedes, 41012 Sevilla, Spain
| |
Collapse
|
6
|
Lorentzen LG, Yeung K, Zitkeviciute A, Yang-Jensen KC, Eldrup N, Eiberg JP, Davies MJ. N-Terminal Proteomics Reveals Distinct Protein Degradation Patterns in Different Types of Human Atherosclerotic Plaques. J Proteome Res 2025; 24:144-157. [PMID: 39665830 DOI: 10.1021/acs.jproteome.4c00548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Atherosclerotic plaque rupture is a major cause of cardiovascular events. Plaque destabilization is associated with extracellular matrix (ECM) modification involving proteases which generate protein fragments with new N-termini. We hypothesized that rupture-prone plaques would contain elevated fragment levels, and their sequences would allow identification of active proteases and target proteins. Plaques from 21 patients who underwent surgery for symptomatic carotid artery stenosis were examined in an observational/cross-sectional study. Plaques were analyzed by liquid chromatography-mass spectrometry for the presence of N-terminal fragments. 33920 peptides were identified, with 17814 being N-terminal species. 5735 distinct N-terminal peptides were quantified and subjected to multidimensional scaling analysis and consensus clustering. These analyses indicated three clusters, which correlate with gross macroscopic plaque morphology (soft/mixed/hard), ultrasound classification (echolucent/echogenic), and the presence of hemorrhage/ulceration. Differences in the fragment complements are consistent with plaque-type-dependent turnover and degradation pathways. Identified peptides include signal and pro-peptides from synthesis and those from protein fragmentation. Sequence analysis indicates that targeted proteins include ECM species and responsible proteases (meprins, cathepsins, matrix metalloproteinases, elastase, and kallikreins). This study provides a large data set of peptide fragments and proteases present in plaques of differing stability. These species may have potential as biomarkers for improved atherosclerosis risk profiling.
Collapse
Affiliation(s)
- Lasse G Lorentzen
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
| | - Karin Yeung
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Auguste Zitkeviciute
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| | - Karen C Yang-Jensen
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nikolaj Eldrup
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jonas P Eiberg
- Department of Vascular Surgery, Heart Centre, University Hospital Copenhagen─Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
- Copenhagen Academy for Medical Education and Simulation, Capital Region of Denmark, Copenhagen 2100, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
7
|
Stewart MR, Quentel A, Manalo E, Montoya Mira J, Ranganathan S, Branchaud BP, Fischer JM, Tu E, Civitci F, Chiu YJ, Yildirim A. Profiling protease cleavage patterns in plasma for pancreatic cancer detection. Sci Rep 2024; 14:31809. [PMID: 39738320 PMCID: PMC11686259 DOI: 10.1038/s41598-024-83077-0] [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: 02/23/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025] Open
Abstract
Proteases are promising biomarkers for cancer early detection. Their enzymatic activity against peptide substrates allows for their straightforward detection using low-cost tests. However, the complexity of the human proteome makes it challenging to develop sensitive and selective tests against a specific protease biomarker. Here, we report a different approach by utilizing the total protease activity in plasma samples to detect pancreatic cancer. Instead of targeting a specific protease using a specific peptide substrate, we utilized an array of 360 FRET substrates to screen for cleavage patterns in plasma samples collected from screen negatives and pancreatitis or pancreatic ductal adenocarcinoma cancer (PDAC) patients. In this proof of concept study, we first screened all 360 substrates using a small cohort (n = 13) to identify the top 5 substrates that best separate different conditions. Then, we performed a validation study using a larger cohort (n = 86) and the selected substrates. There was a statistically significant increase in the total protease activity in PDAC samples compared to screen negative and pancreatitis samples. The selected substrates detected PDAC with an area under the curve (AUC) of 0.8. This work represents a novel strategy for identifying peptide substrates for the detection of PDAC and other cancers.
Collapse
Affiliation(s)
- Morgan R Stewart
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Arnaud Quentel
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Elise Manalo
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Jose Montoya Mira
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Srivathsan Ranganathan
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Bruce P Branchaud
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Jared M Fischer
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Eugene Tu
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Fehmi Civitci
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Yu-Jui Chiu
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Adem Yildirim
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA.
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA.
- Division of Oncological Sciences, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA.
| |
Collapse
|
8
|
Li Q, Zhou SR, Kim H, Wang H, Zhu JJ, Yang JK. Discovering novel Cathepsin L inhibitors from natural products using artificial intelligence. Comput Struct Biotechnol J 2024; 23:2606-2614. [PMID: 39006920 PMCID: PMC11245987 DOI: 10.1016/j.csbj.2024.06.009] [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: 02/05/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024] Open
Abstract
Cathepsin L (CTSL) is a promising therapeutic target for metabolic disorders. Current pharmacological interventions targeting CTSL have demonstrated potential in reducing body weight gain, serum insulin levels, and improving glucose tolerance. However, the clinical application of CTSL inhibitors remains limited. In this study, we used a combination of artificial intelligence and experimental methods to identify new CTSL inhibitors from natural products. Through a robust deep learning model and molecular docking, we screened 150 molecules from natural products for experimental validation. At a concentration of 100 µM, we found that 36 of them exhibited more than 50 % inhibition of CTSL. Notably, 13 molecules displayed over 90 % inhibition and exhibiting concentration-dependent effects. The molecular dynamics simulation on the two most potent inhibitors, Plumbagin and Beta-Lapachone, demonstrated stable interaction at the CTSL active site. Enzyme kinetics studies have shown that these inhibitors exert an uncompetitive inhibitory effect on CTSL. In conclusion, our research identifies Plumbagin and Beta-Lapachone as potential CTSL inhibitors, offering promising candidates for the treatment of metabolic disorders and illustrating the effectiveness of artificial intelligence in drug discovery.
Collapse
Affiliation(s)
- Qi Li
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Si-Rui Zhou
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Hanna Kim
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Hao Wang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Juan-Juan Zhu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
9
|
Momiyama Y, Kishimoto Y, Saita E, Ohmori R, Kondo K. Association between high plasma levels of legumain and cardiovascular events in patients undergoing coronary angiography. Heart Vessels 2024; 39:909-915. [PMID: 38289388 DOI: 10.1007/s00380-024-02373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 01/24/2024] [Indexed: 09/17/2024]
Abstract
Degradation of vascular extracellular matrix is important in atherosclerosis. Cysteine protease legumain is upregulated in atherosclerotic plaques. We recently reported that plasma legumain levels are high in patients with complex coronary lesions. This study investigated the association between legumain levels and cardiovascular events in 372 patients undergoing coronary angiography. Patients with acute coronary syndrome were excluded. Of the 372 patients, 225 had coronary artery disease (CAD). During a mean follow-up of 7.0 ± 4.3 years, cardiovascular events occured in 62 patients. Compared with 310 patients without events, 62 with events tended to have higher prevalence of complex lesions (15% vs. 10%). Notably, patients with events had higher legumain levels (median 5.51 vs. 4.90 ng/mL, P < 0.01) than those without events. A Kaplan-Meier analysis showed lower event-free survival in patients with legumain > 5.0 ng/mL than in those with ≤ 5.0 ng/mL (P < 0.01). In multivariate Cox regression analysis, legumain level was an independent predictor of cardiovascular events. The hazard ratio for legumain > 5.0 ng/mL for cardiovascular events was 2.18 (95%CI = 1.27-3.77, P < 0.01). Only among 225 patients with CAD, patients with events had higher legumain levels (5.49 vs. 4.73 ng/mL) than without events (P < 0.02). Legumain level was also a predictor of cardiovascular events in patients with CAD. Thus, high plasma legumain levels were associated with an increased risk of cardiovascular events in patients undergoing coronary angiography and those with stable CAD.
Collapse
Affiliation(s)
- Yukihiko Momiyama
- Department of Cardiology, NHO Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo, 152-8902, Japan.
| | - Yoshimi Kishimoto
- Department of Food Science and Human Nutrition, Setsunan University, Osaka, Japan
| | - Emi Saita
- Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
| | - Reiko Ohmori
- Faculty of Regional Design, Utsunomiya University, Tochigi, Japan
| | | |
Collapse
|
10
|
Chihab A, El Brahmi N, Hamdoun G, El Abbouchi A, Ghammaz H, Touil N, Bousmina M, El Fahime E, El Kazzouli S. Development of a new experimental NMR strategy for covalent cysteine protease inhibitors screening: toward enhanced drug discovery. RSC Adv 2024; 14:26829-26836. [PMID: 39184001 PMCID: PMC11342919 DOI: 10.1039/d4ra04938a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
In the development of antiviral drugs, proteases and polymerases are among the most important targets. Cysteine proteases, also known as thiol proteases, catalyze the degradation of proteins by cleaving peptide bonds using the nucleophilic thiol group of cysteine. As part of our research, we are examining how cysteine, an essential amino acid found in the active site of the main protease (Mpro) enzyme in SARS-CoV-2, interacts with electrophilic groups present in ethacrynic acid (EA) and compounds 4, 6, and 8 to form sulfur-carbon bonds. Nuclear magnetic resonance (NMR) spectroscopy was used to monitor the reaction rate between cysteine and Michael acceptors. We found that the inhibitory activity of these compounds towards Mpro is correlated to their chemical reactivity toward cysteine. This approach may serve as a valuable tool in drug development for detecting potential covalent inhibitors of Mpro and other cysteine proteases.
Collapse
Affiliation(s)
| | | | | | | | - Hamza Ghammaz
- Centre National de la Recherche Scientifique et Technique (CNRST) Angle avenues des FAR et Allal El Fassi, Hay Ryad 10102 Rabat Morocco
| | - Nadia Touil
- Cell Culture Unit, Center of Virology, Infectious, and Tropical Diseases Mohammed V Military Hospital Rabat Morocco
| | | | - Elmostafa El Fahime
- Centre National de la Recherche Scientifique et Technique (CNRST) Angle avenues des FAR et Allal El Fassi, Hay Ryad 10102 Rabat Morocco
| | | |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Kalinin RE, Konopleva MG, Suchkov IA, Korotkova NV, Mzhavanadze ND. Interleukin-13: association with inflammation and cysteine proteolysis in varicose transformation of the vascular wall. KAZAN MEDICAL JOURNAL 2023; 104:896-906. [DOI: 10.17816/kmj430382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The present review considers current data on the structure, functions and role of interleukin-13 in the pathogenesis of vascular wall varicose transformation in terms of proteolysis and inflammatory response. It is known that interleukin-13 is able to interact with transforming growth factor-1 in diseases associated with fibrosis. The latter activates fibroblasts and excessive formation of the extracellular matrix, thereby inducing fibrosis of the vascular wall, which is one of the links in the pathogenesis of varicose veins. Also, to date, there is evidence of the interleukin-13 participation in the induction of certain proteolytic enzymes synthesis, such as matrix metalloproteinases. For the latter, participation in the transformation of the venous wall has been proven to date. The remodeling of the venous wall itself can lead to an increase in the expression of proteinases, providing a proteolytic mechanism for changing the structural organization of the venous wall in varicose veins of the lower extremities. At the same time, the involvement of lysosomal cysteine proteinases remains poorly understood. The expression and production of individual cathepsins are regulated by biologically active molecules: interleukin-1, interleukin-6, tumor necrosis factor , which are directly involved in inflammatory reactions in the wall of varicose veins. In particular, venous pathology develops in a vicious circle of inflammation with the formation of abnormal venous blood flow, chronic venous hypertension and dilation, and the recruitment of leukocytes. This leads to a further, deeper, remodeling of the walls and valves of the veins, an increase in blood pressure and the release of pro-inflammatory mediators chemokines and cytokines. In connection with the above, in order to understand the mechanisms of proteolysis in the vascular wall in varicose veins of the lower extremities, it is important to have an idea about the possible interactions of interleukin-13 with transforming growth factor-1, inflammatory cytokines, and cathepsins.
Collapse
|
14
|
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
|
15
|
Domingues N, Marques ARA, Calado RDA, Ferreira IS, Ramos C, Ramalho J, Soares MIL, Pereira T, Oliveira L, Vicente JR, Wong LH, Simões ICM, Pinho E Melo TMVD, Peden A, Almeida CG, Futter CE, Puertollano R, Vaz WLC, Vieira OV. Oxidized cholesteryl ester induces exocytosis of dysfunctional lysosomes in lipidotic macrophages. Traffic 2023; 24:284-307. [PMID: 37129279 DOI: 10.1111/tra.12888] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/29/2023] [Accepted: 04/20/2023] [Indexed: 05/03/2023]
Abstract
A key event in atherogenesis is the formation of lipid-loaded macrophages, lipidotic cells, which exhibit irreversible accumulation of undigested modified low-density lipoproteins (LDL) in lysosomes. This event culminates in the loss of cell homeostasis, inflammation, and cell death. Nevertheless, the exact chemical etiology of atherogenesis and the molecular and cellular mechanisms responsible for the impairment of lysosome function in plaque macrophages are still unknown. Here, we demonstrate that macrophages exposed to cholesteryl hemiazelate (ChA), one of the most prevalent products of LDL-derived cholesteryl ester oxidation, exhibit enlarged peripheral dysfunctional lysosomes full of undigested ChA and neutral lipids. Both lysosome area and accumulation of neutral lipids are partially irreversible. Interestingly, the dysfunctional peripheral lysosomes are more prone to fuse with the plasma membrane, secreting their undigested luminal content into the extracellular milieu with potential consequences for the pathology. We further demonstrate that this phenotype is mechanistically linked to the nuclear translocation of the MiT/TFE family of transcription factors. The induction of lysosome biogenesis by ChA appears to partially protect macrophages from lipid-induced cytotoxicity. In sum, our data show that ChA is involved in the etiology of lysosome dysfunction and promotes the exocytosis of these organelles. This latter event is a new mechanism that may be important in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Neuza Domingues
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - André R A Marques
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Rita Diogo Almeida Calado
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Inês S Ferreira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Cristiano Ramos
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - José Ramalho
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Maria I L Soares
- CQC and Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Telmo Pereira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Luís Oliveira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - José R Vicente
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Louise H Wong
- Department of Cell Biology, UCL Institute of Ophthalmology, London, UK
| | - Inês C M Simões
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | | | - Andrew Peden
- Department of Biomedical Science & Center for Membrane Interactions and Dynamics, University of Sheffield, UK
| | - Cláudia Guimas Almeida
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Clare E Futter
- Department of Cell Biology, UCL Institute of Ophthalmology, London, UK
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Winchil L C Vaz
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Otília V Vieira
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
16
|
Chan AS, Wu S, Vernon ST, Tang O, Figtree GA, Liu T, Yang JY, Patrick E. Overcoming cohort heterogeneity for the prediction of subclinical cardiovascular disease risk. iScience 2023; 26:106633. [PMID: 37192969 PMCID: PMC10182278 DOI: 10.1016/j.isci.2023.106633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/03/2023] [Accepted: 04/04/2023] [Indexed: 05/18/2023] Open
Abstract
Cardiovascular disease remains a leading cause of mortality with an estimated half a billion people affected in 2019. However, detecting signals between specific pathophysiology and coronary plaque phenotypes using complex multi-omic discovery datasets remains challenging due to the diversity of individuals and their risk factors. Given the complex cohort heterogeneity present in those with coronary artery disease (CAD), we illustrate several different methods, both knowledge-guided and data-driven approaches, for identifying subcohorts of individuals with subclinical CAD and distinct metabolomic signatures. We then demonstrate that utilizing these subcohorts can improve the prediction of subclinical CAD and can facilitate the discovery of novel biomarkers of subclinical disease. Analyses acknowledging cohort heterogeneity through identifying and utilizing these subcohorts may be able to advance our understanding of CVD and provide more effective preventative treatments to reduce the burden of this disease in individuals and in society as a whole.
Collapse
Affiliation(s)
- Adam S. Chan
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, Australia
| | - Songhua Wu
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Stephen T. Vernon
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Owen Tang
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Tongliang Liu
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, Australia
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Jean Y.H. Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, Australia
| | - Ellis Patrick
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, Australia
- Westmead Medical Institute, Sydney, NSW, Australia
| |
Collapse
|
17
|
Milenkovic D, Rodriguez‐Mateos A, Lucosz M, Istas G, Declerck K, Sansone R, Deenen R, Köhrer K, Corral‐Jara KF, Altschmied J, Haendeler J, Kelm M, Berghe WV, Heiss C. Flavanol Consumption in Healthy Men Preserves Integrity of Immunological-Endothelial Barrier Cell Functions: Nutri(epi)genomic Analysis. Mol Nutr Food Res 2022; 66:e2100991. [PMID: 35094491 PMCID: PMC9787825 DOI: 10.1002/mnfr.202100991] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/16/2022] [Indexed: 12/30/2022]
Abstract
SCOPE While cocoa flavanol (CF) consumption improves cardiovascular risk biomarkers, molecular mechanisms underlying their protective effects are not understood. OBJECTIVE To investigate nutri(epi)genomic effects of CF and identify regulatory networks potential mediating vascular health benefits. METHODS AND RESULTS Twenty healthy middle-aged men consume CF (bi-daily 450 mg) or control drinks for 1 month. Microarray analysis identifies 2235 differentially expressed genes (DEG) involved in processes regulating immune response, cell adhesion, or cytoskeleton organization. Distinct patterns of DEG correlate with CF-related changes in endothelial function, arterial stiffness, and blood pressure. DEG profile negatively correlates with expression profiles of cardiovascular disease patients. CF modulated DNA methylation profile of genes implicates in cell adhesion, actin cytoskeleton organization, or cell signaling. In silico docking analyses indicate that CF metabolites have the potential of binding to cell signaling proteins and transcription factors. Incubation of plasma obtained after CF consumption decrease monocyte to endothelial adhesion and dose-dependently increase nitric oxide-dependent chemotaxis of circulating angiogenic cells further validating the biological functions of CF metabolites. CONCLUSION In healthy humans, CF consumption may mediate vascular protective effects by modulating gene expression and DNA methylation towards a cardiovascular protective effect, in agreement with clinical results, by preserving integrity of immunological-endothelial barrier functions.
Collapse
Affiliation(s)
- Dragan Milenkovic
- Department of NutritionUniversity of California DavisDavisCA95616USA
- INRAEUNHUniversité Clermont AuvergneClermont‐FerrandF‐63000France
| | - Ana Rodriguez‐Mateos
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Department of Nutritional SciencesSchool of Life Course and Population SciencesFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Margarete Lucosz
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Geoffrey Istas
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Department of Nutritional SciencesSchool of Life Course and Population SciencesFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Ken Declerck
- PPESDepartment of Biomedical SciencesUniversity of Antwerp (UA)WilrijkBelgium
| | - Roberto Sansone
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - René Deenen
- Biological and Medical Research Center (BMFZ)Heinrich Heine UniversityDüsseldorfGermany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ)Heinrich Heine UniversityDüsseldorfGermany
| | | | - Joachim Altschmied
- Environmentally‐induced Cardiovascular DegenerationClinical Chemistry and Laboratory DiagnosticsMedical FacultyUniversity Hospital and Heinrich‐Heine UniversityDüsseldorfGermany
- IUF‐Leibniz Research Institute for Environmental MedicineDüsseldorfGermany
| | - Judith Haendeler
- Environmentally‐induced Cardiovascular DegenerationClinical Chemistry and Laboratory DiagnosticsMedical FacultyUniversity Hospital and Heinrich‐Heine UniversityDüsseldorfGermany
| | - Malte Kelm
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Wim Vanden Berghe
- PPESDepartment of Biomedical SciencesUniversity of Antwerp (UA)WilrijkBelgium
| | - Christian Heiss
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Clinical Medicine SectionDepartment of Clinical and Experimental MedicineFaculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
- Department of Vascular MedicineSurrey and Sussex NHS Healthcare TrustEast Surrey HospitalRedhillUK
| |
Collapse
|
18
|
Structure determinants defining the specificity of papain-like cysteine proteases. Comput Struct Biotechnol J 2022; 20:6552-6569. [DOI: 10.1016/j.csbj.2022.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
|
19
|
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: 0.3] [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
|
20
|
Avgustinovich DF, Tenditnik MV, Bondar NP, Marenina MK, Zhanaeva SY, Lvova MN, Katokhin AV, Pavlov KS, Evseenko VI, Tolstikova TG. Behavioral effects and inflammatory markers in the brain and periphery after repeated social defeat stress burdened by Opisthorchis felineus infection in mice. Physiol Behav 2022; 252:113846. [PMID: 35594930 DOI: 10.1016/j.physbeh.2022.113846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 10/18/2022]
Abstract
The combination of 4-week repeated social defeat stress (RSDS) and Opisthorchis felineus infection was modeled in C57BL/6 mice. Various parameters were compared between three experimental groups of male mice (SS: mice subjected to RSDS, OF: mice infected with O. felineus, and OF + SS: mice subjected to both adverse factors) and behavior-tested and intact (INT) controls. The combination caused liver hypertrophy and increased the blood level of proinflammatory cytokine interleukin 6 and proteolytic activity of cathepsin B in the hippocampus. Meanwhile, hypertrophy of the spleen and of adrenal glands was noticeable. Anxious behavior in the elevated plus-maze test was predominantly due to the infection, with synergistic effects of an interaction of the two adverse factors on multiple parameters in OF + SS mice. Depression-like behavior in the forced swimming test was caused only by RSDS and was equally pronounced in SS mice and OF + SS mice. Helminths attenuated the activities of cathepsin B in the liver and hypothalamus (which were high in SS mice) and increased cathepsin L activity in the liver. The highest blood level of corticosterone was seen in SS mice but was decreased to control levels by the trematode infection. OF mice had the lowest level of corticosterone, comparable to that in INT mice. Thus, the first data were obtained on the ability of O. felineus helminths-even at the immature stage-to modulate the effects of RSDS, thereby affecting functional connections of the host, namely "helminths → liver↔brain axis."
Collapse
Affiliation(s)
- Damira F Avgustinovich
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva, 10, Novosibirsk 630090, Russia; Group of Mechanochemistry of Organic Substances, Institute of Solid State Chemistry and Mechanochemistry, SB RAS, Novosibirsk, Russia.
| | - Mikhail V Tenditnik
- Laboratory of Experimental Models of Neurodegenerative Processes, Scientific-Research Institute of Neurosciences and Medicine, SB RAS, Novosibirsk, Russia
| | - Natalia P Bondar
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva, 10, Novosibirsk 630090, Russia; Novosibirsk State University, Novosibirsk, Russia
| | - Mariya K Marenina
- Department of Medicinal Chemistry, Laboratory of Pharmacological Research, N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, SB RAS, Novosibirsk, Russia
| | - Svetlana Ya Zhanaeva
- Department of Psychoneuroimmunology, Scientific-Research Institute of Neurosciences and Medicine, SB RAS, Novosibirsk, Russia
| | - Maria N Lvova
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva, 10, Novosibirsk 630090, Russia
| | - Alexey V Katokhin
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva, 10, Novosibirsk 630090, Russia
| | - Konstantin S Pavlov
- Laboratory of Experimental Models of Neurodegenerative Processes, Scientific-Research Institute of Neurosciences and Medicine, SB RAS, Novosibirsk, Russia
| | - Veronica I Evseenko
- Group of Mechanochemistry of Organic Substances, Institute of Solid State Chemistry and Mechanochemistry, SB RAS, Novosibirsk, Russia
| | - Tatiana G Tolstikova
- Department of Medicinal Chemistry, Laboratory of Pharmacological Research, N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, SB RAS, Novosibirsk, Russia
| |
Collapse
|
21
|
Heib M, Weiß J, Saggau C, Hoyer J, Fuchslocher Chico J, Voigt S, Adam D. Ars moriendi: Proteases as sculptors of cellular suicide. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119191. [PMID: 34973300 DOI: 10.1016/j.bbamcr.2021.119191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
The Ars moriendi, which translates to "The Art of Dying," encompasses two Latin texts that gave advice on how to die well and without fear according to the Christian precepts of the late Middle Ages. Given that ten to hundred billion cells die in our bodies every day, it is obvious that the concept of a well and orderly ("regulated") death is also paramount at the cellular level. In apoptosis, as the most well-studied form of regulated cell death, proteases of the caspase family are the central mediators. However, caspases are not the only proteases that act as sculptors of cellular suicide, and therefore, we here provide an overview of the impact of proteases in apoptosis and other forms of regulated cell death.
Collapse
Affiliation(s)
- Michelle Heib
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Michaelisstr. 5, 24105 Kiel, Germany
| | - Jonas Weiß
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Michaelisstr. 5, 24105 Kiel, Germany
| | - Carina Saggau
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Michaelisstr. 5, 24105 Kiel, Germany
| | - Justus Hoyer
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Michaelisstr. 5, 24105 Kiel, Germany
| | | | - Susann Voigt
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Michaelisstr. 5, 24105 Kiel, Germany
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Michaelisstr. 5, 24105 Kiel, Germany.
| |
Collapse
|
22
|
Zhou Y, Ng DY, Richards AM, Wang P. Loss of full-length pumilio 1 abrogates miRNA-221-induced gene p27 silencing-mediated cell proliferation in the heart. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 27:456-470. [PMID: 35036057 PMCID: PMC8728526 DOI: 10.1016/j.omtn.2021.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 12/09/2021] [Indexed: 12/02/2022]
Abstract
Upregulated expression of microRNA (miR)-221 is associated with downregulation of p27 and subsequent increased cell proliferation in a variety of human cancers. It is unknown whether miR-221 mimics could trigger neoplastic cellular proliferation. In vitro, we demonstrated miR-221 significantly downregulates the expression of P27 and increases proliferation of H9c2 and cardiac fibroblasts. The knockdown of PUM1 but not PUM2 abolished such effects by miR-221, as verified by RT-qPCR and western blot, direct binding of p27 3′ UTR by luciferase reporter assay and cell proliferation by Ki67. In vivo expression of P27 in the rat liver, heart, kidney, spleen, and muscle were not affected by miR-221 at 1 and 4 mg/kg and concurrently full-length (FL) PUM1 (140 kDa) was not detected. Instead, isoforms of 105 and 90 kDa were observed and generated through alternative RNA slicing verified by cDNA cloning and sequencing and cathepsin K cleavage confirmed by studies with the inhibitor odanacatib. This is the first study to address the possible pro-proliferative effects of miR-221 mimic therapeutics in cardiovascular applications. Loss of FL PUM1 expression is a key factor abrogating miR-221-mediated p27 regulation, although other concurrent mechanisms cannot be excluded. Our findings provide essential insights into the context-dependent nature of miRNA functionality.
Collapse
|
23
|
Pan L, Bai P, Weng X, Liu J, Chen Y, Chen S, Ma X, Hu K, Sun A, Ge J. Legumain Is an Endogenous Modulator of Integrin αvβ3 Triggering Vascular Degeneration, Dissection, and Rupture. Circulation 2022; 145:659-674. [PMID: 35100526 DOI: 10.1161/circulationaha.121.056640] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND The development of thoracic aortic dissection (TAD) is closely related to extracellular matrix degradation and vascular smooth muscle cell (VSMC) transformation from contractile to synthetic type. LGMN (legumain) degrades extracellular matrix components directly or by activating downstream signals. The role of LGMN in VSMC differentiation and the occurrence of TAD remains elusive. METHODS Microarray datasets concerning vascular dissection or aneurysm were downloaded from the Gene Expression Omnibus database to screen differentially expressed genes. Four-week-old male Lgmn knockout mice (Lgmn-/-), macrophage-specific Lgmn knockout mice (LgmnF/F;LysMCre), and RR-11a-treated C57BL/6 mice were given BAPN (β-aminopropionitrile monofumarate; 1 g/kg/d) in drinking water for 4 weeks for TAD modeling. RNA sequencing analysis was performed to recapitulate transcriptome profile changes. Cell interaction was examined in macrophage and VSMC coculture system. The reciprocity of macrophage-derived LGMN with integrin αvβ3 in VSMCs was tested by coimmunoprecipitation assay and colocalization analyses. RESULTS Microarray datasets from the Gene Expression Omnibus database indicated upregulated LGMN in aorta from patients with TAD and mice with angiotensin II-induced AAA. Elevated LGMN was evidenced in aorta and sera from patients with TAD and mice with BAPN-induced TAD. BAPN-induced TAD progression was significantly ameliorated in Lgmn-deficient or inhibited mice. Macrophage-specific deletion of Lgmn alleviated BAPN-induced extracellular matrix degradation. Unbiased profiler polymerase chain reaction array and Gene Ontology analysis displayed that LGMN regulated VSMC phenotype transformation. Macrophage-specific deletion of Lgmn ameliorated VSMC phenotypic switch in BAPN-treated mice. Macrophage-derived LGMN inhibited VSMC differentiation in vitro as assessed by macrophages and the VSMC coculture system. Macrophage-derived LGMN bound to integrin αvβ3 in VSMCs and blocked integrin αvβ3, thereby attenuating Rho GTPase activation, downregulating VSMC differentiation markers and eventually exacerbating TAD development. ROCK (Rho kinase) inhibitor Y-27632 reversed the protective role of LGMN depletion in vascular dissection. CONCLUSIONS LGMN signaling may be a novel target for the prevention and treatment of TAD.
Collapse
Affiliation(s)
- Lihong Pan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Peiyuan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Jin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (Y.C.)
| | - Siqin Chen
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Xiurui Ma
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.)
| | - Aijun Sun
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| |
Collapse
|
24
|
The Proteolysis of ECM in Intervertebral Disc Degeneration. Int J Mol Sci 2022; 23:ijms23031715. [PMID: 35163637 PMCID: PMC8835917 DOI: 10.3390/ijms23031715] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc (IVD) degeneration (IDD) is a pathological process that commonly occurs throughout the human life span and is a major cause of lower back pain. Better elucidation of the molecular mechanisms involved in disc degeneration could provide a theoretical basis for the development of lumbar disc intervention strategies. In recent years, extracellular matrix (ECM) homeostasis has received much attention due to its relevance to the mechanical properties of IVDs. ECM proteolysis mediated by a variety of proteases is involved in the pathological process of disc degeneration. Here, we discuss in detail the relationship between the IVD as well as the ECM and the role of ECM proteolysis in the degenerative process of the IVD. Targeting ECM proteolysis-associated proteases may be an effective means of intervention in IDD.
Collapse
|
25
|
Sheikh AM, Wada Y, Tabassum S, Inagaki S, Mitaki S, Yano S, Nagai A. Aggregation of Cystatin C Changes Its Inhibitory Functions on Protease Activities and Amyloid β Fibril Formation. Int J Mol Sci 2021; 22:ijms22189682. [PMID: 34575849 PMCID: PMC8465189 DOI: 10.3390/ijms22189682] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
Cystatin C (CST3) is an endogenous cysteine protease inhibitor, which is implicated in cerebral amyloid angiopathy (CAA). In CAA, CST3 is found to be aggregated. The purpose of this study is to investigate whether this aggregation could alter the activity of the protein relevant to the molecular pathology of CAA. A system of CST3 protein aggregation was established, and the aggregated protein was characterized. The results showed that CST3 aggregated both at 80 °C without agitation, and at 37 °C with agitation in a time-dependent manner. However, the levels of aggregation were high and appeared earlier at 80 °C. Dot-blot immunoassay for oligomers revealed that CST3 could make oligomeric aggregates at the 37 °C condition. Electron microscopy showed that CST3 could make short fibrillary aggregates at 37 °C. Cathepsin B activity assay demonstrated that aggregated CST3 inhibited the enzyme activity less efficiently at pH 5.5. At 7.4 pH, it lost the inhibitory properties almost completely. In addition, aggregated CST3 did not inhibit Aβ1-40 fibril formation, rather, it slightly increased it. CST3 immunocytochemistry showed that the protein was positive both in monomeric and aggregated CST3-treated neuronal culture. However, His6 immunocytochemistry revealed that the internalization of exogenous recombinant CST3 by an astrocytoma cell culture was higher when the protein was aggregated compared to its monomeric form. Finally, MTT cell viability assay showed that the aggregated form of CST3 was more toxic than the monomeric form. Thus, our results suggest that aggregation may result in a loss-of-function phenotype of CST3, which is toxic and responsible for cellular degeneration.
Collapse
Affiliation(s)
- Abdullah Md. Sheikh
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Yasuko Wada
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
| | - Shatera Tabassum
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Satoshi Inagaki
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
| | - Shingo Mitaki
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
| | - Shozo Yano
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Atsushi Nagai
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya Cho, Izumo 693-8501, Japan; (Y.W.); (S.I.); (S.M.)
- Correspondence: ; Tel./Fax: +81-0853-20-2198
| |
Collapse
|
26
|
Hansen L, Brasch HD, Paterson E, Patel J, Bockett N, Davis PF, Tan ST. Expression of Cathepsins B, D, and G in Extracranial Arterio-Venous Malformation. Front Surg 2021; 8:676871. [PMID: 34409065 PMCID: PMC8367294 DOI: 10.3389/fsurg.2021.676871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/30/2021] [Indexed: 12/16/2022] Open
Abstract
Objectives: We have previously identified a population of cells that expressed stemness-associated markers in extracranial arterio-venous malformation (AVM) and demonstrated expression of cathepsins B, D, and G on embryonic stem cell (ESC)-like populations in other vascular anomalies. This study investigated the expression of cathepsins B, D, and G, and their localization in relation to this primitive population in extracranial AVM. Methods: Immunohistochemical staining was performed on AVM tissue samples from 13 patients to demonstrate expression of cathepsins B, D, and G. Western blotting was performed on four AVM tissue samples and three AVM-derived primary cell lines to confirm protein expression of cathepsins B and D proteins. RT-qPCR was performed on three AVM-derived primary cell lines to demonstrate transcript expression of cathepsins B, D, and G. Enzymatic activity assays were performed on three AVM-derived primary cell lines to investigate if cathepsins B and D were active. Localization of the cathepsins was investigated using immunofluorescence dual-staining of the cathepsins with the ESC markers OCT4 and SOX2, and mast cells marker chymase on two of the 13 AVM tissue samples. Results: Immunohistochemical staining demonstrated expression of cathepsins B, D, and G in all 13 AVM tissue samples. Western blotting showed expression of cathepsins B and D proteins in all four AVM tissue samples and all three AVM-derived primary cell lines. RT-qPCR demonstrated transcripts of cathepsins B, D, and G in all three AVM-derived primary cell lines. Enzymatic activity assays showed that cathepsins B and D were active. Immunofluorescence staining showed expression of cathepsins B and D on the OCT4+/SOX2+ endothelium and media of the lesional vessels and cells within the stroma in AVM nidus. Cathepsin G was expressed on the chymase+ phenotypic mast cells. Conclusions: This study demonstrated the novel finding of the expression of cathepsins B, D, and G in AVM. Cathepsins B and D were expressed by the primitive population, and cathepsin G was localized to mast cells, within the AVM nidus.
Collapse
Affiliation(s)
- Lauren Hansen
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Helen D Brasch
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Erin Paterson
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Josie Patel
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | | | - Paul F Davis
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand.,Centre for the Study and Treatment of Vascular Birthmarks, Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Lower Hutt, New Zealand.,Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Abstract
Cysteine cathepsins are proteases critical in physiopathological processes and show potential as targets or biomarkers for diseases and medical conditions. The 11 members of the cathepsin family are redundant in some cases but remarkably independent of others, demanding the development of both pan-cathepsin targeting tools as well as probes that are selective for specific cathepsins with little off-target activity. This review addresses the diverse design strategies that have been employed to accomplish this tailored selectivity among cysteine cathepsin targets and the imaging modalities incorporated. The power of these diverse tools is contextualized by briefly highlighting the nature of a few prominent cysteine cathepsins, their involvement in select diseases, and the application of cathepsin imaging probes in research spanning basic biochemical studies to clinical applications.
Collapse
Affiliation(s)
- Kelton A Schleyer
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| |
Collapse
|
28
|
Hussein NA, Malla S, Pasternak MA, Terrero D, Brown NG, Ashby CR, Assaraf YG, Chen ZS, Tiwari AK. The role of endolysosomal trafficking in anticancer drug resistance. Drug Resist Updat 2021; 57:100769. [PMID: 34217999 DOI: 10.1016/j.drup.2021.100769] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/10/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
Multidrug resistance (MDR) remains a major obstacle towards curative treatment of cancer. Despite considerable progress in delineating the basis of intrinsic and acquired MDR, the underlying molecular mechanisms remain to be elucidated. Emerging evidences suggest that dysregulation in endolysosomal compartments is involved in mediating MDR through multiple mechanisms, such as alterations in endosomes, lysosomes and autophagosomes, that traffic and biodegrade the molecular cargo through macropinocytosis, autophagy and endocytosis. For example, altered lysosomal pH, in combination with transcription factor EB (TFEB)-mediated lysosomal biogenesis, increases the sequestration of hydrophobic anti-cancer drugs that are weak bases, thereby producing an insufficient and off-target accumulation of anti-cancer drugs in MDR cancer cells. Thus, the use of well-tolerated, alkalinizing compounds that selectively block Vacuolar H⁺-ATPase (V-ATPase) may be an important strategy to overcome MDR in cancer cells and increase chemotherapeutic efficacy. Other mechanisms of endolysosomal-mediated drug resistance include increases in the expression of lysosomal proteases and cathepsins that are involved in mediating carcinogenesis and chemoresistance. Therefore, blocking the trafficking and maturation of lysosomal proteases or direct inhibition of cathepsin activity in the cytosol may represent novel therapeutic modalities to overcome MDR. Furthermore, endolysosomal compartments involved in catabolic pathways, such as macropinocytosis and autophagy, are also shown to be involved in the development of MDR. Here, we review the role of endolysosomal trafficking in MDR development and discuss how targeting endolysosomal pathways could emerge as a new therapeutic strategy to overcome chemoresistance in cancer.
Collapse
Affiliation(s)
- Noor A Hussein
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Mariah A Pasternak
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - David Terrero
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Noah G Brown
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY, USA.
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA; Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, 43614, OH, USA.
| |
Collapse
|
29
|
Jan M, Cueto R, Jiang X, Lu L, Sardy J, Xiong X, Yu JE, Pham H, Khan M, Qin X, Ji Y, Yang XF, Wang H. Molecular processes mediating hyperhomocysteinemia-induced metabolic reprogramming, redox regulation and growth inhibition in endothelial cells. Redox Biol 2021; 45:102018. [PMID: 34140262 PMCID: PMC8282538 DOI: 10.1016/j.redox.2021.102018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 01/04/2023] Open
Abstract
Hyperhomocysteinemia (HHcy) is an established and potent independent risk factor for degenerative diseases, including cardiovascular disease (CVD), Alzheimer disease, type II diabetes mellitus, and chronic kidney disease. HHcy has been shown to inhibit proliferation and promote inflammatory responses in endothelial cells (EC), and impair endothelial function, a hallmark for vascular injury. However, metabolic processes and molecular mechanisms mediating HHcy-induced endothelial injury remains to be elucidated. This study examined the effects of HHcy on the expression of microRNA (miRNA) and mRNA in human aortic EC treated with a pathophysiologically relevant concentration of homocysteine (Hcy 500 μM). We performed a set of extensive bioinformatics analyses to identify HHcy-altered metabolic and molecular processes. The global functional implications and molecular network were determined by Gene Set Enrichment Analysis (GSEA) followed by Cytoscape analysis. We identified 244 significantly differentially expressed (SDE) mRNA, their relevant functional pathways, and 45 SDE miRNA. HHcy-altered SDE inversely correlated miRNA-mRNA pairs (45 induced/14 reduced mRNA) were discovered and applied to network construction using an experimentally verified database. We established a hypothetical model to describe the biochemical and molecular network with these specified miRNA/mRNA axes, finding: 1) HHcy causes metabolic reprogramming by increasing glucose uptake and oxidation, by glycogen debranching and NAD+/CoA synthesis, and by stimulating mitochondrial reactive oxygen species production via NNT/IDH2 suppression-induced NAD+/NADP-NADPH/NADP+ metabolism disruption; 2) HHcy activates inflammatory responses by activating inflammasome-pyroptosis mainly through ↓miR193b→↑CASP-9 signaling and by inducing IL-1β and adhesion molecules through the ↓miR29c→↑NEDD9 and the ↓miR1256→↑ICAM-1 axes, as well as GPCR and interferon α/β signaling; 3) HHcy promotes cell degradation by the activation of lysosome autophagy and ubiquitin proteasome systems; 4) HHcy causes cell cycle arrest at G1/S and S/G2 transitions, suppresses spindle checkpoint complex and cytokinetic abscission, and suppresses proliferation through ↓miRNA335/↑VASH1 and other axes. These findings are in accordance with our previous studies and add a wealth of heretofore-unexplored molecular and metabolic mechanisms underlying HHcy-induced endothelial injury. This is the first study to consider the effects of HHcy on both global mRNA and miRNA expression changes for mechanism identification. Molecular axes and biochemical processes identified in this study are useful not only for the understanding of mechanisms underlying HHcy-induced endothelial injury, but also for discovering therapeutic targets for CVD in general. Identified multiple HHcy-altered metabolic and molecular processes potentially responsible for HHcy-induced endothelial injury via examining global mRNA/miRNA expression changes in Hcy-treated EC and performing comprehensive bioinformatic studies. HHcy may activate glucose uptake signaling via the ↓miR148b→↑SLC2A axis. HHcy may induce glucose oxidation signaling by switching pyruvate metabolism from lactate synthesis to mitochondrial oxidation via expression changes of ↑MPC1 & ↓LDHB. HHcy may disrupt redox homeostasis mostly by suppressing NNT/IDH2-related mt-NADPH/mt-NAD+ signaling. HHcy may increase FA β-oxidation, glutamine, TCA cycle and OXPHOS signaling. HHcy may activate inflammatory signaling via the ↓miR29c→↑NEDD9 and the ↓miR1256→↑ICAM-1 axes. HHcy may activate inflammasome/pyroptosis-related signaling by the ↓miR137→↑TLR3, the ↓miR574→↑TRAF5, and the ↓miR193b→↑CASP-9 axes, and induce IL1α/β and CASP-10/7. HHcy may induce inflammation signaling via GPCR activation through the ↓miRNA335→↑CXCR4/↑GNA14 axes. HHcy may activate molecular degradation process signaling through the ↓miRNA335→↑ASAH1/↑ABCB9 axes. HHcy may suppress cell cycle and proliferation through the miR491→↓HMGA2→↓CCNA2/CCNB2, the ↓miR335→↑VASH1, the ↓miR181a→↑PHLDA1, the miR6045→↓CENPH, the miR22→↓PRR11/↓BRCA2, and the miR605/miR497/miR514a→CEP55 axes
Collapse
Affiliation(s)
- Michael Jan
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States; Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, United States
| | - Ramon Cueto
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Liu Lu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Jason Sardy
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Xinyu Xiong
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Justine E Yu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Hung Pham
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Xuebing Qin
- Tulane National Primate Research Center, School of Medicine, Tulane University, Covington, LA, United States
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States; Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States; Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
30
|
Trillhaase A, Schmidt B, Märtens M, Haferkamp U, Erdmann J, Aherrahrou Z. The CAD risk locus 9p21 increases the risk of vascular calcification in an iPSC-derived VSMC model. Stem Cell Res Ther 2021; 12:166. [PMID: 33676559 PMCID: PMC7936418 DOI: 10.1186/s13287-021-02229-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is the leading cause of death worldwide. Chromosome locus 9p21 was the first to be associated with increased risk of CAD and coronary artery calcification (CAC). Vascular calcification increases the risk for CAD. Vascular smooth muscle cells (VSMCs) are one of the major cell types involved in the development of vascular calcification. METHODS So far, mainly animal models or primary SMCs have been used to model human vascular calcification. In this study, a human in vitro assay using iPSC-derived VSMCs was developed to examine vascular calcification. Human iPSCs were derived from a healthy non-risk (NR) and risk (R) donor carrying SNPs in the 9p21 locus. Additionally, 9p21 locus knockouts of each donor iPSC line (NR and R) were used. Following differentiation, the iPSC-derived VSMCs were characterized based on cell type, proliferation, and migration rate, along with calcium phosphate (CaP) deposits. CaP deposits were confirmed using Calcein and Alizarin Red S staining and then quantified. RESULTS The data demonstrated significantly more proliferation, migration, and CaP deposition in VSMCs derived from the R and both KO iPSC lines than in those derived from the NR line. Molecular analyses confirmed upregulation of calcification markers. These results are consistent with recent data demonstrating increased calcification when the 9p21 murine ortholog is knocked-out. CONCLUSION Therefore, in conclusion, genetic variation or deletion of the CAD risk locus leads to an increased risk of vascular calcification. This in vitro human iPSC model of calcification could be used to develop new drug screening strategies to combat CAC.
Collapse
Affiliation(s)
- Anja Trillhaase
- Institute for Cardiogenetics, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Luebeck, Germany.,University Heart Centre Luebeck, 23562, Luebeck, Germany
| | - Beatrice Schmidt
- Institute for Cardiogenetics, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Luebeck, Germany.,University Heart Centre Luebeck, 23562, Luebeck, Germany
| | - Marlon Märtens
- Institute for Cardiogenetics, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Luebeck, Germany.,University Heart Centre Luebeck, 23562, Luebeck, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), 22525, Hamburg, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Luebeck, Germany.,University Heart Centre Luebeck, 23562, Luebeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Luebeck, Germany. .,University Heart Centre Luebeck, 23562, Luebeck, Germany.
| |
Collapse
|
31
|
Liu Y, Liu N, Liu Q. Constructing a ceRNA-immunoregulatory network associated with the development and prognosis of human atherosclerosis through weighted gene co-expression network analysis. Aging (Albany NY) 2021; 13:3080-3100. [PMID: 33460396 PMCID: PMC7880393 DOI: 10.18632/aging.202486] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022]
Abstract
There is now overwhelming experimental and clinical evidence that atherosclerosis (AS) is a chronic inflammatory disease. The recent discovery of a new group of mediators known as competing endogenous RNA (ceRNA) offers a unique opportunity for investigating immunoregulation in AS. In this study, we used gene expression profiles from GEO database to construct a lncRNA-miRNA-mRNA ceRNA network during AS plaque development through weighted gene co-expression network analysis (WGCNA). GO annotation and pathway enrichment analysis suggested that the ceRNA network was mainly involved in the immune response. CIBERSORT and GSVA were used to calculate the immune cell infiltration score and identified macrophage as hub immunocyte in plaque development. A macrophage related ceRNA subnetwork was constructed through correlation analysis. Samples from Biobank of Karolinska Endarterectomy (BiKE) were used to identify prognostic factors from the subnetwork and yielded 7 hub factors that can predict ischemic events including macrophage GSVA score and expression value of AL138756.1, CTSB, MAFB, LYN, GRK3, and BID. A nomogram based on the key factors was established. GSEA identified that the PD1 signaling pathway was negatively associated with these prognostic factors which may explain the cardiovascular side effect of immune checkpoint therapy in anti-tumor treatment.
Collapse
Affiliation(s)
- Yaozhong Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Na Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
32
|
Jing Y, Shi J, Lu B, Zhang W, Yang Y, Wen J, Hu R, Yang Z, Wang X. Association of Circulating Cathepsin S and Cardiovascular Disease Among Patients With Type 2 Diabetes: A Cross-Sectional Community-Based Study. Front Endocrinol (Lausanne) 2021; 12:615913. [PMID: 33746900 PMCID: PMC7973458 DOI: 10.3389/fendo.2021.615913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/01/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cathepsin S, as an adipokine, was reported to play a critical role in various disease, including atherosclerosis and diabetes. The present study aims to elucidate the relationship between circulating cathepsin S and cardiovascular disease (CVD) in patients with type 2 diabetes. METHODS A total of 339 type 2 diabetes individuals were enrolled in this cross-sectional community-based study. Basic information, medical and laboratory data were collected. Serum cathepsin S levels were assessed by ELISA. RESULTS Compared to the CVD (-) group, levels of serum cathepsin S were significantly higher in the CVD (+) group, with the median 23.68 ng/ml (18.54-28.02) and 26.81 ng/ml (21.19-37.69) respectively (P < 0.001). Moreover, patients with acute coronary syndrome (ACS) had substantially higher levels of serum cathepsin S than those with stable angina pectoris (SAP), with the median 34.65 ng/ml (24.33-42.83) and 25.52 ng/ml (20.53-31.47) respectively (P < 0.01). The spearman correlation analysis showed that circulating cathepsin S was correlated with several cardiovascular risk factors. The univariate and multivariate logistic regression analysis revealed that circulating cathepsin S was an independent risk factor for CVD (all P < 0.001) after adjustment for potential confounders. Restricted cubic spline analysis showed circulating cathepsin S had a linearity association with CVD. In addition, receiver operating characteristic (ROC) curve analysis demonstrated that the area under curve (AUC) values of cathepsin S was 0.80 (95% CI: 0.75-0.84, P < 0.001), with the optimal cutoff value of cathepsin 26.28 ng/ml. CONCLUSION Circulating cathepsin S was significantly higher in the CVD (+) group than that in the CVD (-) one among type 2 diabetes. The increased serum cathepsin S levels were associated with increased risks of CVD, even after adjusting for potential confounders. Thus, cathepsin S might be a potential diagnostic biomarker for CVD.
Collapse
Affiliation(s)
- Yu Jing
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Shi
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bin Lu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiwei Zhang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yehong Yang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Wen
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Renming Hu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Yang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Zhen Yang, ; Xuanchun Wang,
| | - Xuanchun Wang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Zhen Yang, ; Xuanchun Wang,
| |
Collapse
|
33
|
Osborn EA, Albaghdadi M, Libby P, Jaffer FA. Molecular Imaging of Atherosclerosis. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
34
|
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: 0.4] [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
|
35
|
Legumain is a predictor of all-cause mortality and potential therapeutic target in acute myocardial infarction. Cell Death Dis 2020; 11:1014. [PMID: 33243972 PMCID: PMC7691341 DOI: 10.1038/s41419-020-03211-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022]
Abstract
The prognostic impact of extracellular matrix (ECM) modulation and its regulatory mechanism post-acute myocardial infarction (AMI), require further clarification. Herein, we explore the predictive role of legumain—which showed the ability in ECM degradation—in an AMI patient cohort and investigate the underlying mechanisms. A total of 212 AMI patients and 323 healthy controls were enrolled in the study. Moreover, AMI was induced in mice by permanent ligation of the left anterior descending artery and fibroblasts were adopted for mechanism analysis. Based on the cut-off value for the receiver-operating characteristics curve, AMI patients were stratified into low (n = 168) and high (n = 44) plasma legumain concentration (PLG) groups. However, PLG was significantly higher in AMI patients than that in the healthy controls (median 5.9 μg/L [interquartile range: 4.2–9.3 μg/L] vs. median 4.4 μg/L [interquartile range: 3.2–6.1 μg/L], P < 0.001). All-cause mortality was significantly higher in the high PLG group compared to that in the low PLG group (median follow-up period, 39.2 months; 31.8% vs. 12.5%; P = 0.002). Multivariate Cox regression analysis showed that high PLG was associated with increased all-cause mortality after adjusting for clinical confounders (HR = 3.1, 95% confidence interval (CI) = 1.4–7.0, P = 0.005). In accordance with the clinical observations, legumain concentration was also increased in peripheral blood, and infarcted cardiac tissue from experimental AMI mice. Pharmacological blockade of legumain with RR-11a, improved cardiac function, decreased cardiac rupture rate, and attenuated left chamber dilation and wall thinning post-AMI. Hence, plasma legumain concentration is of prognostic value in AMI patients. Moreover, legumain aggravates cardiac remodelling through promoting ECM degradation which occurs, at least partially, via activation of the MMP-2 pathway.
Collapse
|
36
|
Koike K, Bando K, Ando J, Yamakoshi H, Terayama N, Dodo K, Smith NI, Sodeoka M, Fujita K. Quantitative Drug Dynamics Visualized by Alkyne-Tagged Plasmonic-Enhanced Raman Microscopy. ACS NANO 2020; 14:15032-15041. [PMID: 33079538 DOI: 10.1021/acsnano.0c05010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Visualizing live-cell uptake of small-molecule drugs is paramount for drug development and pharmaceutical sciences. Bioorthogonal imaging with click chemistry has made significant contributions to the field, visualizing small molecules in cells. Furthermore, recent developments in Raman microscopy, including stimulated Raman scattering (SRS) microscopy, have realized direct visualization of alkyne-tagged small-molecule drugs in live cells. However, Raman and SRS microscopy still suffer from limited detection sensitivity with low concentration molecules for observing temporal dynamics of drug uptake. Here, we demonstrate the combination of alkyne-tag and surface-enhanced Raman scattering (SERS) microscopy for the real-time monitoring of drug uptake in live cells. Gold nanoparticles are introduced into lysosomes of live cells by endocytosis and work as SERS probes. Raman signals of alkynes can be boosted by enhanced electric fields generated by plasmon resonance of gold nanoparticles when alkyne-tagged small molecules are colocalized with the nanoparticles. With time-lapse 3D SERS imaging, this technique allows us to investigate drug uptake by live cells with different chemical and physical conditions. We also perform quantitative evaluation of the uptake speed at the single-cell level using digital SERS counting under different quantities of drug molecules and temperature conditions. Our results illustrate that alkyne-tag SERS microscopy has a potential to be an alternative bioorthogonal imaging technique to investigate temporal dynamics of small-molecule uptake of live cells for pharmaceutical research.
Collapse
Affiliation(s)
- Kota Koike
- Department of Applied Physics, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuki Bando
- Department of Applied Physics, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jun Ando
- Department of Applied Physics, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroyuki Yamakoshi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Naoki Terayama
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Kosuke Dodo
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Nicholas Isaac Smith
- Immunology Frontier Research Center, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mikiko Sodeoka
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Katsumasa Fujita
- Department of Applied Physics, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
37
|
Quesnel A, Karagiannis GS, Filippou PS. Extracellular proteolysis in glioblastoma progression and therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874:188428. [PMID: 32956761 DOI: 10.1016/j.bbcan.2020.188428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Gliomas encompass highly invasive primary central nervous system (CNS) tumours of glial cell origin with an often-poor clinical prognosis. Of all gliomas, glioblastoma is the most aggressive form of primary brain cancer. Current treatments in glioblastoma are insufficient due to the invasive nature of brain tumour cells, which typically results in local tumour recurrence following treatment. The latter represents the most important cause of mortality in glioblastoma and underscores the necessity for an in-depth understanding of the underlying mechanisms. Interestingly, increased synthesis and secretion of several proteolytic enzymes within the tumour microenvironment, such as matrix metalloproteinases, lysosomal proteases, cathepsins and kallikreins for extracellular-matrix component degradation may play a major role in the aforementioned glioblastoma invasion mechanisms. These proteolytic networks are key players in establishing and maintaining a tumour microenvironment that promotes tumour cell survival, proliferation, and migration. Indeed, the targeted inhibition of these proteolytic enzymes has been a promisingly useful therapeutic strategy for glioblastoma management in both preclinical and clinical development. We hereby summarize current advances on the biology of the glioblastoma tumour microenvironment, with a particular emphasis on the role of proteolytic enzyme families in glioblastoma invasion and progression, as well as on their subsequent prognostic value as biomarkers and their therapeutic targeting in the era of precision medicine.
Collapse
Affiliation(s)
- Agathe Quesnel
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA; Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Panagiota S Filippou
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom.
| |
Collapse
|
38
|
Adams LC, Brangsch J, Reimann C, Kaufmann JO, Buchholz R, Karst U, Botnar RM, Hamm B, Makowski MR. Simultaneous molecular MRI of extracellular matrix collagen and inflammatory activity to predict abdominal aortic aneurysm rupture. Sci Rep 2020; 10:15206. [PMID: 32939002 PMCID: PMC7494914 DOI: 10.1038/s41598-020-71817-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/18/2020] [Indexed: 12/23/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease with an up to 80% mortality in case of rupture. Current biomarkers fail to account for size-independent risk of rupture. By combining the information of different molecular probes, multi-target molecular MRI holds the potential to enable individual characterization of AAA. In this experimental study, we aimed to examine the feasibility of simultaneous imaging of extracellular collagen and inflammation for size-independent prediction of risk of rupture in murine AAA. The study design consisted of: (1) A outcome-based longitudinal study with imaging performed once after one week with follow-up and death as the end-point for assessment of rupture risk. (2) A week-by-week study for the characterization of AAA development with imaging after 1, 2, 3 and 4 weeks. For both studies, the animals were administered a type 1 collagen-targeted gadolinium-based probe (surrogate marker for extracellular matrix (ECM) remodeling) and an iron oxide-based probe (surrogate marker for inflammatory activity), in one imaging session. In vivo measurements of collagen and iron oxide probes showed a significant correlation with ex vivo histology (p < 0.001) and also corresponded well to inductively-coupled plasma-mass spectrometry and laser-ablation inductively-coupled plasma mass spectrometry. Combined evaluation of collagen-related ECM remodeling and inflammatory activity was the most accurate predictor for AAA rupture (sensitivity 80%, specificity 100%, area under the curve 0.85), being superior to information from the individual probes alone. Our study supports the feasibility of a simultaneous assessment of collagen-related extracellular matrix remodeling and inflammatory activity in a murine model of AAA.
Collapse
Affiliation(s)
- Lisa C Adams
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Julia Brangsch
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.,Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Königsweg 67, Building 21, 14163, Berlin, Germany
| | - Carolin Reimann
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.,Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Königsweg 67, Building 21, 14163, Berlin, Germany
| | - Jan O Kaufmann
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.,Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing (BAM), Richard-Willstätter-Str. 11, 12489, Berlin, Germany.,Department of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
| | - Rebecca Buchholz
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 30, 48149, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 30, 48149, Münster, Germany
| | - Rene M Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital Westminster Bridge Road, London, SE1 7EH, UK.,Wellcome Trust/EPSRC Centre for Medical Engineering, King's College London, London, UK.,BHF Centre of Excellence, King's College London, Denmark Hill Campus, 125 Coldharbour Lane, London, SE5 9NU, UK.,Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bernd Hamm
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus R Makowski
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.,School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital Westminster Bridge Road, London, SE1 7EH, UK.,Wellcome Trust/EPSRC Centre for Medical Engineering, King's College London, London, UK.,School of Medicine, Department of Diagnostic and Interventional Radiology, Technical University of Munich, 81675, Munich, Germany
| |
Collapse
|
39
|
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: 258] [Impact Index Per Article: 51.6] [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
|
40
|
Lei Y, Ehle B, Kumar SV, Müller S, Moll S, Malone AF, Humphreys BD, Andrassy J, Anders HJ. Cathepsin S and Protease-Activated Receptor-2 Drive Alloimmunity and Immune Regulation in Kidney Allograft Rejection. Front Cell Dev Biol 2020; 8:398. [PMID: 32582696 PMCID: PMC7290053 DOI: 10.3389/fcell.2020.00398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/29/2020] [Indexed: 01/08/2023] Open
Abstract
Alloantigen presentation is an essential process in acute allorejection. In this context, we speculated on a pathogenic role of cathepsin S (Cat-S), a cysteine protease known to promote antigenic peptide loading into MHC class II and to activate protease-activated receptor (PAR)-2 on intrarenal microvascular endothelial and tubular epithelial cells. Single-cell RNA sequencing and immunostaining of human kidney allografts confirmed Cat-S expression in intrarenal mononuclear phagocytes. In vitro, Cat-S inhibition suppressed CD4 + T cell lymphocyte activation in a mixed lymphocyte assay. In vivo, we employed a mouse model of kidney transplantation that showed preemptive Cat-S inhibition significantly protected allografts from tubulitis and intimal arteritis. To determine the contribution of PAR-2 activation, first, Balb/c donor kidneys were transplanted into Balb/c recipient mice without signs of rejection at day 10. In contrast, kidneys from C57BL/6J donor mice revealed severe intimal arteritis, tubulitis, interstitial inflammation, and glomerulitis. Kidneys from Par2-deficient C57BL/6J mice revealed partial protection from tubulitis and lower intrarenal expression levels for Fasl, Tnfa, Ccl5, and Ccr5. Together, we conclude that Cat-S and PAR-2 contribute to immune dysregulation and kidney allograft rejection, possibly involving Cat-S-mediated activation of PAR-2 on recipient parenchymal cells in the allograft.
Collapse
Affiliation(s)
- Yutian Lei
- Division of Nephrology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | - Benjamin Ehle
- Division for General, Visceral, Transplant, Vascular and Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Santhosh V Kumar
- Division of Nephrology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | - Susanne Müller
- Department of Pathology, University of Munich, Munich, Germany
| | - Solange Moll
- Institute of Clinical Pathology, University Hospital Geneva, Geneva, Switzerland
| | - Andrew F Malone
- Division of Nephrology, Department of Medicine, Washington University in Saint Louis School of Medicine, St. Louis, MO, United States
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in Saint Louis School of Medicine, St. Louis, MO, United States.,Department of Developmental Biology, Washington University in Saint Louis School of Medicine, St. Louis, MO, United States
| | - Joachim Andrassy
- Division for General, Visceral, Transplant, Vascular and Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
41
|
Yu C, Wan Y, Piao L, Wu Cheng X. Can cysteinyl cathepsin activity control diet-induced NAFLD? IJC HEART & VASCULATURE 2020; 28:100516. [PMID: 32373709 PMCID: PMC7195526 DOI: 10.1016/j.ijcha.2020.100516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/04/2020] [Accepted: 04/09/2020] [Indexed: 11/25/2022]
Key Words
- CSTB, cathepsin B
- CTSB+/+, wild-type
- CTSB−/−, cathepsin B deficiency
- CTSs, cathepsins
- FABP4, fatty acid binding protein 4
- FPC, fructose-palmitate-cholesterol
- LDL, low density lipoprotein
- MMP, metalloproteinase
- NAFLD, Non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- Scad, short-chain acyl dehydrogenase-1
Collapse
Affiliation(s)
| | | | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, China
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, China
| |
Collapse
|
42
|
Trivedi PC, Bartlett JJ, Pulinilkunnil T. Lysosomal Biology and Function: Modern View of Cellular Debris Bin. Cells 2020; 9:cells9051131. [PMID: 32375321 PMCID: PMC7290337 DOI: 10.3390/cells9051131] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
Lysosomes are the main proteolytic compartments of mammalian cells comprising of a battery of hydrolases. Lysosomes dispose and recycle extracellular or intracellular macromolecules by fusing with endosomes or autophagosomes through specific waste clearance processes such as chaperone-mediated autophagy or microautophagy. The proteolytic end product is transported out of lysosomes via transporters or vesicular membrane trafficking. Recent studies have demonstrated lysosomes as a signaling node which sense, adapt and respond to changes in substrate metabolism to maintain cellular function. Lysosomal dysfunction not only influence pathways mediating membrane trafficking that culminate in the lysosome but also govern metabolic and signaling processes regulating protein sorting and targeting. In this review, we describe the current knowledge of lysosome in influencing sorting and nutrient signaling. We further present a mechanistic overview of intra-lysosomal processes, along with extra-lysosomal processes, governing lysosomal fusion and fission, exocytosis, positioning and membrane contact site formation. This review compiles existing knowledge in the field of lysosomal biology by describing various lysosomal events necessary to maintain cellular homeostasis facilitating development of therapies maintaining lysosomal function.
Collapse
Affiliation(s)
- Purvi C. Trivedi
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4H7, Canada; (P.C.T.); (J.J.B.)
- Dalhousie Medicine New Brunswick, Saint John, NB E2L 4L5, Canada
| | - Jordan J. Bartlett
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4H7, Canada; (P.C.T.); (J.J.B.)
- Dalhousie Medicine New Brunswick, Saint John, NB E2L 4L5, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4H7, Canada; (P.C.T.); (J.J.B.)
- Dalhousie Medicine New Brunswick, Saint John, NB E2L 4L5, Canada
- Correspondence: ; Tel.: +1-(506)-636-6973
| |
Collapse
|
43
|
Cardoso Dos Santos M, Algar WR, Medintz IL, Hildebrandt N. Quantum dots for Förster Resonance Energy Transfer (FRET). Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115819] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
44
|
Osawa Y, Semba RD, Fantoni G, Candia J, Biancotto A, Tanaka T, Bandinelli S, Ferrucci L. Plasma proteomic signature of the risk of developing mobility disability: A 9-year follow-up. Aging Cell 2020; 19:e13132. [PMID: 32157804 PMCID: PMC7189986 DOI: 10.1111/acel.13132] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Mobility disability is a powerful indicator of poor health in older adults. The biological and pathophysiological mechanism underlying the development of mobility disability remains unknown. This study conducted a data-driven discovery phase investigation to identify plasma proteins that predict the incidence of mobility disability in community-dwelling older adults without mobility disability at baseline. METHODS We investigated 660 women and men, aged 71.9 ± 6.0 (60-94) years, who participated in the Invecchiare in Chianti, "Aging in the Chianti Area" study and completed the 400-m walk at fast pace (400-m walk) at enrollment. Median follow-up time was 8.57 [interquartile, 3.20-9.08] years. SOMAscan technology was used to measure 1,301 plasma proteins at enrollment. The incident of mobility disability was defined as inability to complete the 400-m walk. Protein-specific Cox proportional hazard model was adjusted for sex, age, and other important covariates. RESULTS Plasma levels of 75 proteins predicted mobility disability (p < .05). Significant proteins were enriched for the KEGG "PI3K-Akt signaling," "phagosomes," and "cytokine-cytokine receptor interaction" pathways. After multiple comparison adjustment, plasma cathepsin S (CTSS; hazard ratio [HR] 1.33, 95% CI: 1.17, 1.51, q = 0.007), growth/differentiation factor 15 (GDF15; HR: 1.45, 95% CI: 1.23, 1.72, q = 0.007), and thrombospondin-2 (THBS2; HR: 1.44, 95% CI: 1.22, 1.69, q = 0.007) remained significantly associated with high risk of losing mobility. CONCLUSION CTSS, GDF15, and THBS2 are novel blood biomarkers associated with new mobility disability in community-dwelling individuals. Overall, our analysis suggests that cellular senescence and inflammation should be targeted for prevention of mobility disability.
Collapse
Affiliation(s)
- Yusuke Osawa
- Longitudinal Study SectionTranslational Gerontology BranchNational Institute on AgingNational Institutes of HealthBaltimoreMDUSA
| | - Richard D. Semba
- Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Giovanna Fantoni
- Clinical Research CoreNational Institute on AgingNational Institutes of HealthBaltimoreMDUSA
| | - Julián Candia
- Laboratory of Human CarcinogenesisCenter for Cancer ResearchNational Cancer InstituteNIHBethesdaMDUSA
| | - Angélique Biancotto
- Precision Immunology, Immunology and Inflammation Research Therapeutic AreaSanofiCambridgeMAUSA
| | - Toshiko Tanaka
- Longitudinal Study SectionTranslational Gerontology BranchNational Institute on AgingNational Institutes of HealthBaltimoreMDUSA
| | | | - Luigi Ferrucci
- Longitudinal Study SectionTranslational Gerontology BranchNational Institute on AgingNational Institutes of HealthBaltimoreMDUSA
| |
Collapse
|
45
|
Lai CH, Chang JY, Wang KC, Lee FT, Wu HL, Cheng TL. Pharmacological Inhibition of Cathepsin S Suppresses Abdominal Aortic Aneurysm in Mice. Eur J Vasc Endovasc Surg 2020; 59:990-999. [PMID: 32033870 DOI: 10.1016/j.ejvs.2020.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/30/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Evidence suggests that cathepsin S (CTSS), a potent mammalian elastase, participates in abdominal aortic aneurysm (AAA) formation. This study examines the hypothesis that pharmacological inhibition of CTSS with an α-ketoamide based compound 6r might suppress AAA in mice. METHODS Experimental study of the CaCl2 induced AAA model in B6 mice and angiotensin II (AngII) infused AAA model in ApoE-/- mice. The effects of intraperitoneal administration of 6r (25 mg/kg) and vehicle every three days since one day after AAA induction were evaluated at 28 days using CaCl2 induced (n = 12 per group) and AngII infused (n = 8 per group) models. Additionally, the effects of post-treatment with 6r and vehicle from seven days or 14 days after AAA induction were evaluated at 28 days using the CaCl2 induced model (n = 6 per group). Aortic samples were harvested for histological and biochemical analyses, including cathepsin levels, Verhoeff Van Gieson staining, TUNEL assay, and immunostaining for macrophages. RESULTS In the CaCl2 induced model, treatment with 6r suppressed aortic dilatation observed in vehicle treated controls (median: 0.58 vs. 0.92 mm; p < .001), along with reduced CTSS and cathepsin K (CTSK) levels (both p < .001), preserved elastin integrity (p < .001), fewer medial apoptotic cells (p = .012) and less macrophage infiltration (p = .041). In the AngII infused model, the aortic diameter was smaller in 6r treated mice than in vehicle treated controls (median: 0.95 vs. 1.84 mm; p = .047). The levels of CTSS (p < .001) and CTSK (p = .033) and the numbers of elastin breaks (p < .001), medial apoptotic cells (p < .001) and infiltrating macrophages (p = .030) were attenuated under 6r treatment. Finally, post-treatment with 6r from seven days (p = .046) or 14 days (p = .012) after AAA induction limited CaCl2 induced AAA. CONCLUSION Pharmacological inhibition of CTSS by 6r suppresses AAA formation in mice. Also, post-treatment with 6r retards mouse AAA progression. These findings provide proof of concept validation for CTSS as a potential therapeutic target in AAA.
Collapse
Affiliation(s)
- Chao-Han Lai
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan
| | - Jang-Yang Chang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Kuan-Chieh Wang
- Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Fang-Tzu Lee
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hua-Lin Wu
- Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Lin Cheng
- Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopaedic Research Centre, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
46
|
Lysosomal Abnormalities in Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21030811. [PMID: 32012649 PMCID: PMC7036830 DOI: 10.3390/ijms21030811] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/25/2020] [Indexed: 12/12/2022] Open
Abstract
The lysosome, a key organelle for cellular clearance, is associated with a wide variety of pathological conditions in humans. Lysosome function and its related pathways are particularly important for maintaining the health of the cardiovascular system. In this review, we highlighted studies that have improved our understanding of the connection between lysosome function and cardiovascular diseases with an emphasis on a recent breakthrough that characterized a unique autophagosome-lysosome fusion mechanism employed by cardiomyocytes through a lysosomal membrane protein LAMP-2B. This finding may impact the development of future therapeutic applications.
Collapse
|
47
|
Dai R, Wu Z, Chu HY, Lu J, Lyu A, Liu J, Zhang G. Cathepsin K: The Action in and Beyond Bone. Front Cell Dev Biol 2020; 8:433. [PMID: 32582709 PMCID: PMC7287012 DOI: 10.3389/fcell.2020.00433] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/08/2020] [Indexed: 02/05/2023] Open
Abstract
Cathepsin K (CatK) is one of the most potent proteases in lysosomal cysteine proteases family, of which main function is to mediate bone resorption. Currently, CatK is among the most attractive targets for anti-osteoporosis drug development. Although many pharmaceutical companies are working on the development of selective inhibitors for CatK, there is no FDA approved drug till now. Odanacatib (ODN) developed by Merck & Co. is the only CatK inhibitor candidate which demonstrated high therapeutic efficacy in patients with postmenopausal osteoporosis in Phase III clinical trials. Unfortunately, the development of ODN was finally terminated due to the cardio-cerebrovascular adverse effects. Therefore, it arouses concerns on the undesirable CatK inhibition in non-bone sites. It is known that CatK has far-reaching actions throughout various organs besides bone. Many studies have also demonstrated the involvement of CatK in various diseases beyond the musculoskeletal system. This review not only summarized the functional roles of CatK in bone and beyond bone, but also discussed the potential relevance of the CatK action beyond bone to the adverse effects of inhibiting CatK in non-bone sites.
Collapse
Affiliation(s)
- Rongchen Dai
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Zeting Wu
- International Medical Service Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Hang Yin Chu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Jun Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
- *Correspondence: Jin Liu,
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
- Ge Zhang,
| |
Collapse
|
48
|
Umei TC, Kishimoto Y, Aoyama M, Saita E, Niki H, Ikegami Y, Ohmori R, Kondo K, Momiyama Y. High Plasma Levels of Legumain in Patients with Complex Coronary Lesions. J Atheroscler Thromb 2019; 27:711-717. [PMID: 31735728 PMCID: PMC7406406 DOI: 10.5551/jat.52027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aim: The degradation of the vascular extracellular matrix is important for atherosclerosis. The cysteine protease legumain was shown to be upregulated in atherosclerotic plaques, especially unstable plaques. However, no study has reported blood legumain levels in patients with coronary artery disease (CAD). Methods: We investigated plasma legumain and C-reactive protein (CRP) levels in 372 patients undergoing elective coronary angiography. Results: CAD was found in 225 patients. Compared with patients without CAD, those with CAD had higher CRP levels (median 0.60 [0.32, 1.53] vs. 0.46 [0.22, 0.89] mg/L, P < 0.001), but no difference was found in legumain levels between patients with and without CAD (median 5.08 [3.87, 6.82] vs. 4.99 [3.84, 6.88] ng/mL). A stepwise increase in CRP was found depending on the number of > 50% stenotic vessels: 0.55 mg/L in 1-vessel, 0.71 mg/L in 2-vessel, and 0.86 mg/L in 3-vessel diseases (P < 0.001). However, legumain did not differ among 1-, 2-, and 3-vessel diseases (5.20, 4.93, and 5.01 ng/mL, respectively). Of 225 patients with CAD, 40 (18%) had complex lesions. No difference was found in CRP levels between patients with CAD with and without complex lesions (0.60 [0.34, 1.53] vs. 0.60 [0.32, 1.51] mg/L). Notably, legumain levels were higher in patients with CAD with complex lesions than without such lesions (6.05 [4.64, 8.64] vs. 4.93 [3.76, 6.52] ng/mL, P < 0.01). In multivariate analysis, legumain levels were not a factor for CAD, but were a factor for complex lesions. The odds ratio for complex lesions was 2.45 (95% CI = 1.26–4.79) for legumain > 5.5 ng/mL. Conclusion: Plasma legumain levels were associated with the presence of complex coronary lesions.
Collapse
Affiliation(s)
- Tomohiko C Umei
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Yoshimi Kishimoto
- Endowed Research Department "Food for Health", Ochanomizu University
| | - Masayuki Aoyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Emi Saita
- Endowed Research Department "Food for Health", Ochanomizu University
| | - Hanako Niki
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Yukinori Ikegami
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Reiko Ohmori
- Faculty of Regional Design, Utsunomiya University
| | - Kazuo Kondo
- Endowed Research Department "Food for Health", Ochanomizu University.,Institute of Life Innovation Studies, Toyo University
| | - Yukihiko Momiyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| |
Collapse
|
49
|
Pande M, Srivastava R. Molecular and clinical insights into protein misfolding and associated amyloidosis. Eur J Med Chem 2019; 184:111753. [PMID: 31622853 DOI: 10.1016/j.ejmech.2019.111753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 09/24/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022]
Abstract
The misfolding of normally soluble proteins causes their aggregation and deposition in the tissues which disrupts the normal structure and function of the corresponding organs. The proteins with high β-sheet contents are more prone to form amyloids as they exhibit high propensity of self-aggregation. The self aggregated misfolded proteins act as template for further aggregation that leads to formation of protofilaments and eventually amyloid fibrils. More than 30 different types of proteins are known to be associated with amyloidosis related diseases. Several aspects of the amyloidogenic behavior of proteins remain elusive. The exact reason that causes misfolding of the protein and its association into amyloid fibrils is not known. These misfolded intermediates surpass the over engaged quality control system of the cell which clears the misfolded intermediates. This promotes the self-aggregation, accumulation and deposition of these misfolded species in the form of amyloids in the different parts of the body. The amyloid deposition can be localized as in Alzheimer disease or systemic as reported in most of the amyloidosis. The amyloidosis can be of acquired type or familial. The current review aims at bringing together recent updates and comprehensive information about protein amyloidosis and associated diseases at one place.
Collapse
Affiliation(s)
- Monu Pande
- Department of Biochemistry, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India
| | - Ragini Srivastava
- Department of Biochemistry, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
50
|
Mechanistic insights into the inhibition mechanism of cysteine cathepsins by chalcone-based inhibitors—a QM cluster model approach. Struct Chem 2019. [DOI: 10.1007/s11224-018-1273-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|