1
|
Chen JH, Chen JY, Chen YC, Li WC. Sex difference in the association between creatinine-to-cystatin C ratio and metabolic syndrome among Chinese adults. Front Endocrinol (Lausanne) 2024; 15:1389295. [PMID: 39205686 PMCID: PMC11349525 DOI: 10.3389/fendo.2024.1389295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
Background Metabolic syndrome (MetS), characterized by central obesity, insulin resistance, dyslipidemia, and hypertension, affects 20-25% of the global population. The creatinine-to-cystatin C ratio (CCR) is an indicator of skeletal muscle mass. While CCR may play a role in MetS development, sex differences in these associations are not fully understood. Therefore, this study aimed to investigate how CCR levels are associated with MetS in a Chinese adult population, focusing on possible sex disparities. Method We conducted a retrospective cross-sectional analysis of 9,376 adults from Xiamen Chang Gung Hospital between 2014 to 2016. We examined the relationship between CCR and MetS, adjusting for cardiometabolic risk factors. Results The prevalence of MetS was 24.7% in males and 18.0% in females. Interestingly, we observed significant sex differences in the association between CCR quartiles and MetS. Females in the lowest CCR quartile had a significantly higher risk of MetS (odds ratio=1.84). Receiver operating characteristic curve analysis revealed acceptable diagnostic power of CCR for MetS in females (area under the curve=0.65) but not in males. Conclusion Our findings suggest that CCR is an independent risk factor for MetS in females, highlighting the importance of sex-specific assessments when evaluating MetS risk.
Collapse
Affiliation(s)
- Jo-Hsuan Chen
- Department of Family Medicine, Chang-Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jau-Yuan Chen
- Department of Family Medicine, Chang-Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Health Management, Xiamen Chang-Gung Hospital, Xiamen, China
| | - Yi-Chuan Chen
- Department of Family Medicine, Chang-Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Wen-Cheng Li
- Department of Family Medicine, Chang-Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
2
|
Wang HL, Narisawa M, Wu P, Meng X, Cheng XW. The many roles of cathepsins in restenosis. Heliyon 2024; 10:e24720. [PMID: 38333869 PMCID: PMC10850908 DOI: 10.1016/j.heliyon.2024.e24720] [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: 09/01/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Drug-eluting stents (DES) and dual antiplatelet regimens have significantly improved the clinical management of ischemic heart disease; however, the drugs loaded with DES in clinical practice are mostly paclitaxel or rapamycin derivatives, which target symptoms of post implantation proliferation and inflammation, leading to delayed re-endothelialization and neo-atherosclerosis. Along with the treatments already in place, there is a need for novel strategies to lessen the negative clinical outcomes of DES delays as well as a need for greater understanding of their pathobiological mechanisms. This review concentrates on the function of cathepsins (Cats) in the inflammatory response and granulation tissue formation that follow Cat-induced damage to the vasculature scaffold, as well as the functions of Cats in intimal hyperplasia, which is characterized by the migration and proliferation of smooth muscle cells, and endothelial denudation, re-endothelialization, and/or neo-endothelialization. Additionally, Cats can alter essential neointima formation and immune response inside scaffolds, and if Cats are properly controlled in vivo, they may improve scaffold biocompatibility. This unique profile of functions could lead to an original concept for a cathepsin-based coronary intervention treatment as an adjunct to stent placement.
Collapse
Affiliation(s)
- Hai Long Wang
- Department of Adult Intensive Care Unit, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4668550, Japan
| | - Pan Wu
- Department of Adult Intensive Care Unit, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiangkun Meng
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, PR China
| |
Collapse
|
3
|
Ajani TA, Obikeze K, Magwebu ZE, Egieyeh S, Chauke CG. In-silico and in-vitro screening of Asiatic acid and Asiaticoside A against Cathepsin S enzyme. BMC Pharmacol Toxicol 2023; 24:67. [PMID: 38007481 PMCID: PMC10676574 DOI: 10.1186/s40360-023-00701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/31/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Atherosclerosis is a form of cardiovascular disease that affects the endothelium of the blood vessel. Series of events are involved in the pathophysiology of this disease which includes the breaking down of the connective tissue elastin and collagen responsible for the tensile strength of the arterial wall by proteolytic enzyme. One of these enzymes called Cathepsin S (CatS) is upregulated in the progression of the disease and its inhibition has been proposed to be a promising pharmacological target to improve the prognosis of the disease condition. Asiatic acid and asiaticoside A are both pentacyclic triterpenoids isolated from Centella asiatica. Their use in treating various cardiovascular diseases has been reported. METHODS In this study through in silico and in vitro methods, the pharmacokinetic properties, residue interaction, and inhibitory activities of these compounds were checked against the CatS enzyme. The SwissADME online package and the ToxTree 3.01 version of the offline software were used to determine the physicochemical properties of the compounds. RESULT Asiatic acid reported no violation of the Lipinski rule while asiaticoside A violated the rule with regards to its molecular structure and size. The molecular docking was done using Molecular Operating Environment (MOE) and the S-score of - 7.25988, - 7.08466, and - 4.147913 Kcal/mol were recorded for LY300328, asiaticoside A, and asiatic acid respectively. Asiaticoside A has a docking score value (- 7.08466Kcal/mol) close to the co-crystallize compound. Apart from the close docking score, the amino acid residue glycine69 and asparagine163 both interact with the co-crystallized compound and asiaticoside A. The in vitro result clearly shows the inhibitory effect of asiaticoside and asiatic acid. Asiaticoside A has an inhibitory value of about 40% and asiatic acid has an inhibitory value of about 20%. CONCLUSION This clearly shows that asiaticoside will be a better drug candidate than asiatic acid in inhibiting the CatS enzyme for the purpose of improving the outcome of atherosclerosis. However, certain modifications need to be made to the structural make-up of asiaticoside A to improve its pharmacokinetics properties.
Collapse
Affiliation(s)
| | - Kenechukwu Obikeze
- University of the Western Cape, School of Pharmacy, Bellville, South Africa
| | - Zandisiwe E Magwebu
- South African Medical Research Council, Primate Unit and Delft Animal Centre (PUDAC), Cape town, South Africa
| | - Samuel Egieyeh
- University of the Western Cape, School of Pharmacy, Bellville, South Africa
| | - Chesa G Chauke
- South African Medical Research Council, Primate Unit and Delft Animal Centre (PUDAC), Cape town, South Africa
| |
Collapse
|
4
|
Zhou Z, Jiang WJ, Li L, Si JQ. Noise exposure increase apoptosis in the hippocampus of AD mice through the upregulation of CTSS. Biochem Biophys Res Commun 2023; 681:283-290. [PMID: 37801777 DOI: 10.1016/j.bbrc.2023.09.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023]
Abstract
In recent years, it has become an acknowledged fact that noise exposure can lead to cognitive impairments, and researchers have shown increasing interest in this area. However, the detrimental impact of noise exposure on Alzheimer's disease (AD) animal models might be considerably greater than on ordinary model mice, yet the mechanisms by which noise exposure affects the hippocampus in these models have been scarcely investigated. This study we used 4D Label-free proteomics to identify distinctive differentially expressed proteins in the hippocampus of AD model mice following noise exposure. Among these proteins, the presence of Cathepsin S(CTSS) cannot be disregarded. Utilizing experimental techniques such as Western blot, immunofluorescence, and rt-qPCR, we confirmed the expression of CTSS in the hippocampus of APP/PS1 mice after noise exposure. Additionally, we examined downstream molecules including P53,BCL-2, BAX, and CASPASE3 using KEGG pathway analysis. The results indicated an elevation in CTSS expression, a reduction in the anti-apoptotic gene BCL-2, and an increase in the expression of BAX and cleaved CASPASE3. Based on these findings, we hypothesize that noise exposure potentially heightens apoptosis within the hippocampus through upregulating CTSS expression, subsequently posing a threat to AD model animals.
Collapse
Affiliation(s)
- Zan Zhou
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China; Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Wen-Jun Jiang
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, China; Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310051, China
| | - Li Li
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, China.
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
5
|
Bastrup JA, Jepps TA. Proteomic mapping reveals dysregulated angiogenesis in the cerebral arteries of rats with early-onset hypertension. J Biol Chem 2023; 299:105221. [PMID: 37660920 PMCID: PMC10558802 DOI: 10.1016/j.jbc.2023.105221] [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: 03/14/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
Hypertension is associated with the presence of vascular abnormalities, including remodeling and rarefaction. These processes play an important role in cerebrovascular disease development; however, the mechanistic changes leading to these diseases are not well characterized. Using data-independent acquisition-based mass spectrometry analysis, here we determined the protein changes in cerebral arteries in pre- and early-onset hypertension from the spontaneously hypertensive rat (SHR), a model that resembles essential hypertension in humans. Our analysis identified 125 proteins with expression levels that were significantly upregulated or downregulated in 12-week-old spontaneously hypertensive rats compared to normotensive Wistar Kyoto rats. Using an angiogenesis enrichment analysis, we further identified a critical imbalance in angiogenic proteins that promoted an anti-angiogenic profile in cerebral arteries at early onset of hypertension. In a comparison to previously published data, we demonstrate that this angiogenic imbalance is not present in mesenteric and renal arteries from age-matched SHRs. Finally, we identified two proteins (Fbln5 and Cdh13), whose expression levels were critically altered in cerebral arteries compared to the other arterial beds. The observation of an angiogenic imbalance in cerebral arteries from the SHR reveals critical protein changes in the cerebrovasculature at the early onset of hypertension and provides novel insights into the early pathology of cerebrovascular disease.
Collapse
Affiliation(s)
- Joakim A Bastrup
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Thomas A Jepps
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Abstract
Cardiovascular disease (CVD) is prevalent in patients with chronic kidney disease (CKD) and it is responsible for approximately half of all CKD-related deaths. CVDs are the primary cause of death in hemodialysis patients due to major adverse cardiovascular events. Therefore, better approaches for differentiating chronic hemodialysis patients at higher cardiovascular risk will help physicians improve clinical outcomes. Hence, there is an urgent need to discover feasible and reliable cardiac biomarkers to improve diagnostic accuracy, reflect myocardial injury, and identify high-risk patients. Numerous biomarkers that have significant prognostic value with respect to adverse CVD outcomes in the setting of mild to severe CKD have been identified. Therefore, a better understanding of the positive clinical impact of cardiac biomarkers on CVD patient outcomes is an important step toward prevention and improving treatment in the future. In this review, we address the relationship between cardiovascular biomarkers and CKD treatment strategies to elucidate the underlying importance of these biomarkers to patient outcomes.
Collapse
Affiliation(s)
- Ying-Ju Chen
- Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung, Taiwan
| | - Chih-Chieh Chen
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Tze-Kiong Er
- Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
7
|
Cathepsins in the extracellular space: Focusing on non-lysosomal proteolytic functions with clinical implications. Cell Signal 2023; 103:110531. [PMID: 36417977 DOI: 10.1016/j.cellsig.2022.110531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Cathepsins can be found in the extracellular space, cytoplasm, and nucleus. It was initially suspected that the primary physiological function of the cathepsins was to break down intracellular protein, and that they also had a role in pathological processes including inflammation and apoptosis. However, the many actions of cathepsins outside the cell and their complicated biological impacts have garnered much interest. Cathepsins play significant roles in a number of illnesses by regulating parenchymal cell proliferation, cell migration, viral invasion, inflammation, and immunological responses through extracellular matrix remodeling, signaling disruption, leukocyte recruitment, and cell adhesion. In this review, we outline the physiological roles of cathepsins in the extracellular space, the crucial pathological functions performed by cathepsins in illnesses, and the recent breakthroughs in the detection and therapy of specific inhibitors and fluorescent probes in associated dysfunction.
Collapse
|
8
|
Xie L, Zhang S, Huang L, Peng Z, Lu H, He Q, Chen R, Hu L, Wang B, Sun B, Yang Q, Xie Q. Single-cell RNA sequencing of peripheral blood reveals that monocytes with high cathepsin S expression aggravate cerebral ischemia-reperfusion injury. Brain Behav Immun 2023; 107:330-344. [PMID: 36371010 DOI: 10.1016/j.bbi.2022.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/19/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Stroke is a major cause of morbidity and mortality worldwide. After cerebral ischemia, peripheral immune cells infiltrate the brain and elicit an inflammatory response. However, it is not clear when and how these peripheral immune cells affect the central inflammatory response, and whether interventions that target these processes can alleviate ischemia-reperfusion (I/R) injury. METHODS Single-cell transcriptomic sequencing and bioinformatics analysis were performed on peripheral blood of mice at different times after I/R to analyze the key molecule of cell subsets. Then, the expression pattern of this molecule was determined through various biological experiments, including quantitative RT-PCR, western blot, ELISA, and in situ hybridization. Next, the function of this molecule was assessed using knockout mice and the corresponding inhibitor. RESULTS Single-cell transcriptomic sequencing revealed that peripheral monocyte subpopulations increased significantly after I/R. Cathepsin S (Ctss)was identified as a key molecule regulating monocyte activation by pseudotime trajectory analysis and gene function analysis. Next, Cathepsin S was confirmed to be expressed in monocytes with the highest expression level 3 days after I/R. Infarct size (p < 0.05), neurological function scores (p < 0.05), and apoptosis and vascular leakage rates were significantly reduced after Ctss knockout. In addition, CTSS destroyed the blood-brain barrier (BBB) by binding to junctional adhesion molecule (JAM) family proteins to cause their degradation. CONCLUSIONS Cathepsin S inhibition attenuated cerebral I/R injury; therefore, cathepsin S can be used as a novel target for drug intervention after stroke.
Collapse
Affiliation(s)
- Lexing Xie
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Shuang Zhang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Li Huang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Zhouzhou Peng
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Hui Lu
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Qian He
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Ru Chen
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Linlin Hu
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Bingqiao Wang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China; Chongqing Institute for Brain and Intelligence, CIBI, China
| | - Baoliang Sun
- Department of Neurology, The Second Affiliated Hospital, Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Qingwu Yang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China.
| | - Qi Xie
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China.
| |
Collapse
|
9
|
Biasizzo M, Javoršek U, Vidak E, Zarić M, Turk B. Cysteine cathepsins: A long and winding road towards clinics. Mol Aspects Med 2022; 88:101150. [PMID: 36283280 DOI: 10.1016/j.mam.2022.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022]
Abstract
Biomedical research often focuses on properties that differentiate between diseased and healthy tissue; one of the current focuses is elevated expression and altered localisation of proteases. Among these proteases, dysregulation of cysteine cathepsins can frequently be observed in inflammation-associated diseases, which tips the functional balance from normal physiological to pathological manifestations. Their overexpression and secretion regularly exhibit a strong correlation with the development and progression of such diseases, making them attractive pharmacological targets. But beyond their mostly detrimental role in inflammation-associated diseases, cysteine cathepsins are physiologically highly important enzymes involved in various biological processes crucial for maintaining homeostasis and responding to different stimuli. Consequently, several challenges have emerged during the efforts made to translate basic research data into clinical applications. In this review, we present both physiological and pathological roles of cysteine cathepsins and discuss the clinical potential of cysteine cathepsin-targeting strategies for disease management and diagnosis.
Collapse
Affiliation(s)
- Monika Biasizzo
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Miki Zarić
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
10
|
Sinha S, Haque M. Obesity, Diabetes Mellitus, and Vascular Impediment as Consequences of Excess Processed Food Consumption. Cureus 2022; 14:e28762. [PMID: 36105908 PMCID: PMC9441778 DOI: 10.7759/cureus.28762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2022] [Indexed: 12/15/2022] Open
Abstract
Regular intake of ready-to-eat meals is related to obesity and several noninfectious illnesses, such as cardiovascular diseases, hypertension, diabetes mellitus (DM), and tumors. Processed foods contain high calories and are often enhanced with excess refined sugar, saturated and trans fat, Na+ andphosphate-containing taste enhancers, and preservatives. Studies showed that monosodium glutamate (MSG) induces raised echelons of oxidative stress, and excessive hepatic lipogenesis is concomitant to obesity and type 2 diabetes mellitus (T2DM). Likewise, more than standard salt intake adversely affects the cardiovascular system, renal system, and central nervous system (CNS), especially the brain. Globally, excessive utilization of phosphate-containing preservatives and additives contributes unswervingly to excessive phosphate intake through food. In addition, communities and even health experts, including medical doctors, are not well-informed about the adverse effects of phosphate preservatives on human health. Dietary phosphate excess often leads to phosphate toxicity, ultimately potentiating kidney disease development. The mechanisms involved in phosphate-related adverse effects are not explainable. Study reports suggested that high blood level of phosphate causes vascular ossification through the deposition of Ca2+ and substantially alters fibroblast growth factor-23 (FGF23) and calcitriol.
Collapse
|
11
|
Oostveen RF, Kaiser Y, Stroes ES, Verberne HJ. Molecular Imaging of Aortic Valve Stenosis with Positron Emission Tomography. Pharmaceuticals (Basel) 2022; 15:ph15070812. [PMID: 35890111 PMCID: PMC9319069 DOI: 10.3390/ph15070812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Aortic valve stenosis (AVS) is an increasingly prevalent disease in our aging population. Although multiple risk factors for AVS have been elucidated, medical therapies capable of slowing down disease progression remain unavailable. Molecular imaging technologies are opening up avenues for the non-invasive assessment of disease progression, allowing the assessment of (early) medical interventions. This review will focus on the role of positron emission tomography of the aortic valve with 18F-fluorodeoxyglucose and 18F-sodium fluoride but will also shed light on novel tracers which have potential in AVS, ranging from the healthy aortic valve to end-stage valvular disease.
Collapse
Affiliation(s)
- Reindert F. Oostveen
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (R.F.O.); (Y.K.); (E.S.G.S.)
| | - Yannick Kaiser
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (R.F.O.); (Y.K.); (E.S.G.S.)
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (R.F.O.); (Y.K.); (E.S.G.S.)
| | - Hein J. Verberne
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-562-8436
| |
Collapse
|
12
|
Nagy EE, Puskás A, Kelemen P, Makó K, Brassai Z, Hársfalvi J, Frigy A. Elevated Serum Cystatin C and Decreased Cathepsin S/Cystatin C Ratio Are Associated with Severe Peripheral Arterial Disease and Polyvascular Involvement. Diagnostics (Basel) 2022; 12:diagnostics12040833. [PMID: 35453881 PMCID: PMC9029365 DOI: 10.3390/diagnostics12040833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 11/16/2022] Open
Abstract
Peripheral arterial disease (PAD) is frequently associated with atherosclerotic manifestations of the carotids and coronaries. Polyvascular involvement and low ankle−brachial index predict major cardiovascular events and high mortality. Cathepsin S (Cat S) promotes the inflammatory pathways of the arterial wall, while Cystatin C (Cys C) functions as its inhibitor; therefore, Cys C was proposed to be a biomarker of progression in PAD. In a single-center observational study, we investigated the correlations of serum Cys C and Cat S/Cys C ratio in a group of 90 PAD patients, predominantly with polyvascular involvement. Cys C and Cat S/Cys C were associated with ankle−brachial index (ABI) scores <0.4 in univariate and multiple regression models. Furthermore, both markers correlated positively with the plasma Von Willebrand Factor Antigen (VWF: Ag) and Von Willebrand Factor collagen-binding activity (VWF: CB). In addition, Cat S/Cys C was significantly decreased, whereas Cys C increased in subjects with three-bed atherosclerotic involvement. According to our results, high serum Cys C and low Cat S/Cys C ratios may indicate severe peripheral arterial disease and polyvascular atherosclerotic involvement.
Collapse
Affiliation(s)
- Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
- Laboratory of Medical Analysis, Clinical County Hospital Mures, 540394 Targu Mures, Romania
- Correspondence: ; Tel.: +40-733-956-395
| | - Attila Puskás
- Angio-Center Vascular Medicine, 540074 Targu Mures, Romania;
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
| | - Piroska Kelemen
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
| | - Katalin Makó
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
- Hestia General Practioner Ltd., H-1188 Budapest, Hungary
| | - Zoltán Brassai
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
| | - Jolán Hársfalvi
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, H-1444 Budapest, Hungary;
| | - Attila Frigy
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Department of Cardiology, Clinical County Hospital Mures, 540072 Targu Mures, Romania
| |
Collapse
|
13
|
Sonkar SK, Singh PK, Chandra S, Sonkar GK, Bhosale V, Sharma S. Cathepsin S as an early biomarker for cardiovascular disease in chronic kidney disease patients. J Bras Nefrol 2022; 44:329-335. [PMID: 35023538 PMCID: PMC9518627 DOI: 10.1590/2175-8239-jbn-2021-0135] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION A high incidence of cardiovascular disease (CVD) events and premature mortality is observed in patients with chronic kidney disease (CKD). Thus, new biomarkers that may help predict the development of CVD in early stages of CKD are being investigated along with other traditional risk factors. OBJECTIVE To investigate cathepsin S as an early biomarker for CVD in patients with CKD. METHODS A total of 64 patients with CKD were included and classified into 2 groups: CKD patients with established CVD and CKD patients with non-established CVD. All patients were submitted to routine investigations including complete blood count, random blood sugar, glycated hemoglobin (HbA1c), serum electrolytes, urea, creatinine, total protein, total albumin, calcium total, phosphorous, uric acid, vitamin D, parathormone, lipid profile, liver function test, measurement of serum cathepsin S (Cat S), and 2D Echo of the heart. RESULTS The level of serum Cat S was increased in CKD patients with CVD (p <0.05) as well as in later stages of CKD (p <0.05). CVD was also more common in patients in early stage CKD. In early stages CKD, Cat S and CVD were positively correlated. CONCLUSION These findings suggest that serum Cat S might be useful as an early biomarker for CVD in CKD patients.
Collapse
Affiliation(s)
| | | | - Sharad Chandra
- King George's Medical University, Cardiology Department, Lucknow, India
| | | | - Vivek Bhosale
- Central Drug Research Institute, Clinical & Experimental Medicine, Lucknow, India
| | - Sharad Sharma
- Central Drug Research Institute, Clinical & Experimental Medicine, Lucknow, India
| |
Collapse
|
14
|
Urine peptidome analysis in cardiorenal syndrome reflects molecular processes. Sci Rep 2021; 11:16219. [PMID: 34376786 PMCID: PMC8355128 DOI: 10.1038/s41598-021-95695-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
The cardiorenal syndrome (CRS) is defined as the confluence of heart-kidney dysfunction. This study investigates the molecular differences at the level of the urinary peptidome between CRS patients and controls and their association to disease pathophysiology. The urinary peptidome of CRS patients (n = 353) was matched for age and sex with controls (n = 356) at a 1:1 ratio. Changes in the CRS peptidome versus controls were identified after applying the Mann-Whitney test, followed by correction for multiple testing. Proteasix tool was applied to investigate predicted proteases involved in CRS-associated peptide generation. Overall, 559 differentially excreted urinary peptides were associated with CRS patients. Of these, 193 peptides were specifically found in CRS when comparing with heart failure and chronic kidney disease urinary peptide profiles. Proteasix predicted 18 proteases involved in > 1% of proteolytic cleavage events including multiple forms of MMPs, proprotein convertases, cathepsins and kallikrein 4. Forty-four percent of the cleavage events were produced by 3 proteases including MMP13, MMP9 and MMP2. Pathway enrichment analysis supported that ECM-related pathways, fibrosis and inflammation were represented. Collectively, our study describes the changes in urinary peptides of CRS patients and potential proteases involved in their generation, laying the basis for further validation.
Collapse
|
15
|
Borhanjoo P, Singh N, Nath S, Chowdhury MS, Swanson C, Kaiser R, Geraghty P, Foronjy RF, Chow L. Systemic inflammation and protease profile of Afro-Caribbean patients with sepsis. SAGE Open Med 2021; 9:20503121211012521. [PMID: 33996084 PMCID: PMC8107660 DOI: 10.1177/20503121211012521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/05/2021] [Indexed: 11/25/2022] Open
Abstract
Objectives: Sepsis is one of the leading causes of morbidity and mortality within the healthcare system and remains a diagnostic and therapeutic challenge. A major issue in the diagnosis of sepsis is understanding the pathophysiologic mechanism, which revolves around host immune system activation and dysregulated responses. African Americans are more likely to experience severe sepsis with higher mortality rates compared to the general population. This pilot study characterized multiple inflammatory markers and proteases in plasma of primarily African American and Afro-Caribbean patients with mild sepsis. Methods: Plasma was collected from 16 healthy controls and 15 subjects presenting with sepsis, on admission, and again upon resolution of the signs of sepsis, defined as a resolution of sepsis criteria. Plasma samples were analyzed for cytokines, chemokines, and proteases using multiplex bead assays. Results: Elevated levels of granulocyte colony-stimulating factor, interleukin-10, interleukin-15, interleukin-1 receptor antagonist, interleukin-8, interferon gamma-induced protein 10, monocyte chemoattractant protein-1, matrix metallopeptidase 12, and cathepsin S were identified in plasma from sepsis patients on admission compared to control subjects. Interleukin-6, interleukin-8, granulocyte colony-stimulating factor, and cathepsin S were reduced in sepsis patients upon clinical resolution of sepsis. Conclusion: These findings profile the circulating inflammatory cytokines, chemokines, and proteases in African Americans and Afro-Caribbean patients during sepsis. The role of these targets in sepsis needs addressing in this patient population.
Collapse
Affiliation(s)
- Panid Borhanjoo
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Navneet Singh
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Sridesh Nath
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | | | - Carl Swanson
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Ryan Kaiser
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Patrick Geraghty
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Robert F Foronjy
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Lillian Chow
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
16
|
Zhao J, Yang Y, Wu Y. The Clinical Significance and Potential Role of Cathepsin S in IgA Nephropathy. Front Pediatr 2021; 9:631473. [PMID: 33912521 PMCID: PMC8071879 DOI: 10.3389/fped.2021.631473] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/17/2021] [Indexed: 12/26/2022] Open
Abstract
Objective: Cathepsin S (CTSS) is an important lysosomal cysteine protease. This study aimed at investigating the clinical significance of CTSS and underlying mechanism in immunoglobulin A nephropathy (IgAN). Methods: This study recruited 25 children with IgAN and age-matched controls and their serum CTSS levels were measured by enzyme-linked immunosorbent assay (ELISA). Following induction of IgAN in rats, their kidney CTSS expression, IgA accumulation and serum CTSS were characterized by immunohistochemistry, immunofluorescence, and ELISA. The impact of IgA1 aggregates on the proliferation of human mesangial cells (HMCs) was determined by Cell Counting Kit-8 and Western blot analysis of Ki67. Results: Compared to the non-IgAN controls, significantly up-regulated CTSS expression was detected in the renal tissues, particularly in the glomerular mesangium and tubular epithelial cells of IgAN patients, accompanied by higher levels of serum CTSS (P < 0.05), which were correlated with the levels of 24-h-urine proteins and microalbumin and urine erythrocytes and grades of IgAN Lee's classification in children with IgAN (P < 0.01 for all). Following induction of IgAN, we detected inducible IgA accumulation and increased levels of CTSS expression in the glomerular mesangium and glomerular damages in rats, which were mitigated by LY3000328, a CTSS-specific inhibitor. Treatment with LY3000328 significantly mitigated the Ki67 expression in the kidney of IgAN rats (P < 0.01) and significantly minimized the IgA1 aggregate-stimulated proliferation of HMCs and their Ki67 expression in vitro (P < 0.01). Conclusions: CTSS promoted the proliferation of glomerular mesangial cells, contributing to the pathogenesis of IgAN and may be a new therapeutic target for intervention of aberrant mesangial cell proliferation during the process of IgAN.
Collapse
Affiliation(s)
- Jingying Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongchang Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yubin Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
Peng K, Liu H, Yan B, Meng XW, Song SY, Ji FH, Xia Z. Inhibition of cathepsin S attenuates myocardial ischemia/reperfusion injury by suppressing inflammation and apoptosis. J Cell Physiol 2020; 236:1309-1320. [PMID: 32657442 DOI: 10.1002/jcp.29938] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/16/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury leads to high mortality and morbidity due to the incomplete understanding of the underlying mechanism and the consequent lack of effective therapy. The present study revealed and validated key candidate genes in relation to inflammation and apoptosis pathways underlying myocardial I/R injury. Cathepsin S was identified as the top hub protein based on the protein-protein interaction analysis, and, thus, its role during myocardial I/R injury was further investigated. Myocardial I/R in mice resulted in significantly increased levels of myocardial injury biomarkers (cardiac troponin I, lactic dehydrogenase, and creatinine kinase-MB) and inflammatory cytokines (interleukin-1β [IL-1β], IL-6, and tumor necrosis factor-α), elevated apoptosis rate, and upregulated protein expression of cleaved caspase-8, cleaved caspase-3, and cleaved poly ADP-ribose polymerase. These abovementioned changes were blocked by two different selective cathepsin S inhibitors, LY3000328 or MIV-247. Moreover, Kaplan-Meier survival plot showed that cathepsin S inhibition improved 21-day survival rate following myocardial I/R injury. This study demonstrated that the inhibition of cathepsin S alleviated myocardial I/R-induced injury by suppressing inflammation and apoptosis, which may be used in clinical applications of cardioprotection.
Collapse
Affiliation(s)
- Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, California
| | - Bin Yan
- Department of Intervention and Cell Therapy, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Computer Science, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shao-Yong Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengyuan Xia
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, California.,Department of Anesthesiology, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
18
|
De Pasquale V, Moles A, Pavone LM. Cathepsins in the Pathophysiology of Mucopolysaccharidoses: New Perspectives for Therapy. Cells 2020; 9:cells9040979. [PMID: 32326609 PMCID: PMC7227001 DOI: 10.3390/cells9040979] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Cathepsins (CTSs) are ubiquitously expressed proteases normally found in the endolysosomal compartment where they mediate protein degradation and turnover. However, CTSs are also found in the cytoplasm, nucleus, and extracellular matrix where they actively participate in cell signaling, protein processing, and trafficking through the plasma and nuclear membranes and between intracellular organelles. Dysregulation in CTS expression and/or activity disrupts cellular homeostasis, thus contributing to many human diseases, including inflammatory and cardiovascular diseases, neurodegenerative disorders, diabetes, obesity, cancer, kidney dysfunction, and others. This review aimed to highlight the involvement of CTSs in inherited lysosomal storage disorders, with a primary focus to the emerging evidence on the role of CTSs in the pathophysiology of Mucopolysaccharidoses (MPSs). These latter diseases are characterized by severe neurological, skeletal and cardiovascular phenotypes, and no effective cure exists to date. The advance in the knowledge of the molecular mechanisms underlying the activity of CTSs in MPSs may open a new challenge for the development of novel therapeutic approaches for the cure of such intractable diseases.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Anna Moles
- Institute of Biomedical Research of Barcelona, Spanish Research Council, 08036 Barcelona, Spain;
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
- Correspondence: ; Tel.: +39-081-7463043
| |
Collapse
|
19
|
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
|
20
|
Zheng KH, Tzolos E, Dweck MR. Pathophysiology of Aortic Stenosis and Future Perspectives for Medical Therapy. Cardiol Clin 2020; 38:1-12. [DOI: 10.1016/j.ccl.2019.09.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
21
|
Characterization of cathepsin S exosites that govern its elastolytic activity. Biochem J 2020; 477:227-242. [DOI: 10.1042/bcj20190847] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 11/17/2022]
Abstract
We have previously determined that the elastolytic activities of cathepsins (Cat) K and V require two exosites sharing the same structural localization on both enzymes. The structural features involved in the elastolytic activity of CatS have not yet been identified. We first mutated the analogous CatK and V putative exosites of CatS into the elastolytically inactive CatL counterparts. The modification of the exosite 1 did not affect the elastase activity of CatS whilst mutation of the Y118 of exosite 2 decreased the cleavage of elastin by ∼70% without affecting the degradation of other macromolecular substrates (gelatin, thyroglobulin). T06, an ectosteric inhibitor that disrupt the elastolytic activity of CatK, blocked ∼80% of the elastolytic activity of CatS without blocking the cleavage of gelatin and thyroglobulin. Docking studies showed that T06 preferentially interacts with a binding site located on the Right domain of the enzyme, outside of the active site. The structural examination of this binding site showed that the loop spanning the L174N175G176K177 residues of CatS is considerably different from that of CatL. Mutation of this loop into the CatL-like equivalent decreased elastin degradation by ∼70% and adding the Y118 mutation brought down the loss of elastolysis to ∼80%. In addition, the Y118 mutation selectively reduced the cleavage of the basement membrane component laminin by ∼50%. In summary, our data show that the degradation of elastin by CatS requires two exosites where one of them is distinct from those of CatK and V whilst the cleavage of laminin requires only one exosite.
Collapse
|
22
|
Abstract
Lysosomes are membrane-bound organelles with roles in processes involved in degrading and recycling cellular waste, cellular signalling and energy metabolism. Defects in genes encoding lysosomal proteins cause lysosomal storage disorders, in which enzyme replacement therapy has proved successful. Growing evidence also implicates roles for lysosomal dysfunction in more common diseases including inflammatory and autoimmune disorders, neurodegenerative diseases, cancer and metabolic disorders. With a focus on lysosomal dysfunction in autoimmune disorders and neurodegenerative diseases - including lupus, rheumatoid arthritis, multiple sclerosis, Alzheimer disease and Parkinson disease - this Review critically analyses progress and opportunities for therapeutically targeting lysosomal proteins and processes, particularly with small molecules and peptide drugs.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Fengjuan Wang
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France.
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France.
- University of Strasbourg Institute for Advanced Study, Strasbourg, France.
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, Strasbourg, France.
| |
Collapse
|
23
|
Voelkl J, Lang F, Eckardt KU, Amann K, Kuro-O M, Pasch A, Pieske B, Alesutan I. Signaling pathways involved in vascular smooth muscle cell calcification during hyperphosphatemia. Cell Mol Life Sci 2019; 76:2077-2091. [PMID: 30887097 PMCID: PMC6502780 DOI: 10.1007/s00018-019-03054-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023]
Abstract
Medial vascular calcification has emerged as a putative key factor contributing to the excessive cardiovascular mortality of patients with chronic kidney disease (CKD). Hyperphosphatemia is considered a decisive determinant of vascular calcification in CKD. A critical role in initiation and progression of vascular calcification during elevated phosphate conditions is attributed to vascular smooth muscle cells (VSMCs), which are able to change their phenotype into osteo-/chondroblasts-like cells. These transdifferentiated VSMCs actively promote calcification in the medial layer of the arteries by producing a local pro-calcifying environment as well as nidus sites for precipitation of calcium and phosphate and growth of calcium phosphate crystals. Elevated extracellular phosphate induces osteo-/chondrogenic transdifferentiation of VSMCs through complex intracellular signaling pathways, which are still incompletely understood. The present review addresses critical intracellular pathways controlling osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification during hyperphosphatemia. Elucidating these pathways holds a significant promise to open novel therapeutic opportunities counteracting the progression of vascular calcification in CKD.
Collapse
MESH Headings
- Animals
- Calcium Phosphates/chemistry
- Calcium Phosphates/metabolism
- Cell Transdifferentiation
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Gene Expression Regulation
- Humans
- Hyperphosphatemia/complications
- Hyperphosphatemia/genetics
- Hyperphosphatemia/metabolism
- Hyperphosphatemia/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Osteoblasts/metabolism
- Osteoblasts/pathology
- RANK Ligand/genetics
- RANK Ligand/metabolism
- Receptor Activator of Nuclear Factor-kappa B/genetics
- Receptor Activator of Nuclear Factor-kappa B/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- Vascular Calcification/complications
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria.
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany.
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany.
| | - Florian Lang
- Department of Physiology I, Eberhard-Karls University, Wilhelmstr. 56, 72076, Tübingen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany
| | - Kerstin Amann
- Department of Nephropathology, Universität Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Andreas Pasch
- Calciscon AG, Aarbergstrasse 5, 2560, Nidau-Biel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178, Berlin, Germany
- Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178, Berlin, Germany
| |
Collapse
|
24
|
Vidak E, Javoršek U, Vizovišek M, Turk B. Cysteine Cathepsins and their Extracellular Roles: Shaping the Microenvironment. Cells 2019; 8:cells8030264. [PMID: 30897858 PMCID: PMC6468544 DOI: 10.3390/cells8030264] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 12/17/2022] Open
Abstract
For a long time, cysteine cathepsins were considered primarily as proteases crucial for nonspecific bulk proteolysis in the endolysosomal system. However, this view has dramatically changed, and cathepsins are now considered key players in many important physiological processes, including in diseases like cancer, rheumatoid arthritis, and various inflammatory diseases. Cathepsins are emerging as important players in the extracellular space, and the paradigm is shifting from the degrading enzymes to the enzymes that can also specifically modify extracellular proteins. In pathological conditions, the activity of cathepsins is often dysregulated, resulting in their overexpression and secretion into the extracellular space. This is typically observed in cancer and inflammation, and cathepsins are therefore considered valuable diagnostic and therapeutic targets. In particular, the investigation of limited proteolysis by cathepsins in the extracellular space is opening numerous possibilities for future break-through discoveries. In this review, we highlight the most important findings that establish cysteine cathepsins as important players in the extracellular space and discuss their roles that reach beyond processing and degradation of extracellular matrix (ECM) components. In addition, we discuss the recent developments in cathepsin research and the new possibilities that are opening in translational medicine.
Collapse
Affiliation(s)
- Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Matej Vizovišek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich Otto-Stern-Weg 3, 8093 Zürich, Switzerland.
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
25
|
Koukos PI, Xue LC, Bonvin AMJJ. Protein-ligand pose and affinity prediction: Lessons from D3R Grand Challenge 3. J Comput Aided Mol Des 2019; 33:83-91. [PMID: 30128928 PMCID: PMC6373529 DOI: 10.1007/s10822-018-0148-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/09/2018] [Indexed: 12/30/2022]
Abstract
We report the performance of HADDOCK in the 2018 iteration of the Grand Challenge organised by the D3R consortium. Building on the findings of our participation in last year's challenge, we significantly improved our pose prediction protocol which resulted in a mean RMSD for the top scoring pose of 3.04 and 2.67 Å for the cross-docking and self-docking experiments respectively, which corresponds to an overall success rate of 63% and 71% when considering the top1 and top5 models respectively. This performance ranks HADDOCK as the 6th and 3rd best performing group (excluding multiple submissions from a same group) out of a total of 44 and 47 submissions respectively. Our ligand-based binding affinity predictor is the 3rd best predictor overall, behind only the two leading structure-based implementations, and the best ligand-based one with a Kendall's Tau correlation of 0.36 for the Cathepsin challenge. It also performed well in the classification part of the Kinase challenges, with Matthews Correlation Coefficients of 0.49 (ranked 1st), 0.39 (ranked 4th) and 0.21 (ranked 4th) for the JAK2, vEGFR2 and p38a targets respectively. Through our participation in last year's competition we came to the conclusion that template selection is of critical importance for the successful outcome of the docking. This year we have made improvements in two additional areas of importance: ligand conformer selection and initial positioning, which have been key to our excellent pose prediction performance this year.
Collapse
Affiliation(s)
- Panagiotis I Koukos
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Li C Xue
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Alexandre M J J Bonvin
- Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
26
|
Vascular Calcification in Chronic Kidney Disease: The Role of Inflammation. Int J Nephrol 2018; 2018:4310379. [PMID: 30186632 PMCID: PMC6109995 DOI: 10.1155/2018/4310379] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular complications are extremely frequent in patients with chronic kidney disease (CKD) and death from cardiac causes is the most common cause of death in this particular population. Cardiovascular disease is approximately 3 times more frequent in patients with CKD than in other known cardiovascular risk groups and cardiovascular mortality is approximately 10-fold more frequent in patients on dialysis compared to the age- and sex-matched segments of the nonrenal population. Among other structural and functional factors advanced calcification of atherosclerotic plaques as well as of the arterial and venous media has been described as potentially relevant for this high cardiovascular morbidity and mortality. One potential explanation for this exceedingly high vascular calcification in animal models as well as in patients with CKD increased systemic and most importantly local (micro)inflammation that has been shown to favor the development of calcifying particles by multiple ways. Of note, local vascular upregulation of proinflammatory and proosteogenic molecules is already present at early stages of CKD and may thus be operative for vascular calcification. In addition, increased expression of costimulatory molecules and mast cells has also been documented in patients with CKD pointing to a more inflammatory and potentially less stable phenotype of coronary atherosclerotic plaques in CKD.
Collapse
|