1
|
Sagir B, Okutucu M, Arpa M, Findik H, Uzun F, Gokhan Aslan M, Şahin Ü, Kaim M. Evaluation of Choroidal Thickness and Retinal Vessel Density with Serum HIF-1α and TNF-α Level in Patients with OSAS. Curr Eye Res 2025; 50:66-73. [PMID: 39118389 DOI: 10.1080/02713683.2024.2386355] [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: 12/06/2023] [Revised: 06/07/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE To reveal changes in choroidal thickness, retinal vessel density, and serum HIF-1α and TNF-α levels in obstructive sleep apnea syndrome (OSAS) and their correlation. METHODS This prospective case-control study included 118 patients divided into mild-to-moderate OSAS (n = 40), severe OSAS (n = 39), and a control group (n = 39). Choroidal thickness was evaluated with OCT, vessel density with OCTA, AHI index with polysomnography, and serum HIF-1α and TNF-α levels were analyzed using the enzyme-linked immunosorbent assay. RESULTS The serum HIF-1α values of the participants in the mild-moderate OSAS and severe OSAS groups were [893.25(406.7-2068) and 1027(453-2527), respectively], and were both significantly higher than the control group [(521.5(231.6-2741))] (p < 0.001). Serum TNF-α levels did not differ significantly between the groups (p = 0.051).). Subfoveal choroidal thickness (SFCT) values of the severe OSAS groups were significantly lower than the control group (p < 0.05). The superficial and deep capillary plexus vascular density (SVD and DVD) values of the severe OSAS group were lower than the control group (p < 0.05). Serum HIF-1α and TNF-α levels of all participants were negatively correlated with both their SVD values (p < 0.05, r: -0.220 and p < 0.05, r: -0.252, respectively) and their DVD values (p < 0.001, r: -0.324 and p = 0.001, r: -0.299, respectively). CONCLUSIONS Increased serum levels of inflammatory mediators (HIF-1α ve TNF-α) in OSAS cause a decrease in SFCT, SVD, and DVD, which is an indication of systemic vascular damage. Further research on developing treatment strategies to modulate TNF-α ve HIF-1α may help recede vascular morbidity in OSAS patients.
Collapse
Affiliation(s)
- Busra Sagir
- Department of Ophtalmology, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Murat Okutucu
- Department of Ophthalmology, Recep Tayyip Erdoğan University Tip Fakultesi, Rize, Turkey
| | - Medeni Arpa
- Department of Biochemistry, Recep Tayyip Erdoğan University Tip Fakultesi, Rize, Turkey
| | - Hüseyin Findik
- Department of Ophthalmology, Recep Tayyip Erdoğan University Tip Fakultesi, Rize, Turkey
| | - Feyzahan Uzun
- Department of Ophthalmology, Recep Tayyip Erdoğan University School of Medicine, Rize, Turkey
| | | | - Ünal Şahin
- Department of Chest Diseases, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Muhammet Kaim
- Department of Ophthalmology, Recep Tayyip Erdoğan University School of Medicine, Rize, Turkey
| |
Collapse
|
2
|
Tehlivets O, Almer G, Brunner MS, Lechleitner M, Sommer G, Kolb D, Leitinger G, Diwoky C, Wolinski H, Habisch H, Opriessnig P, Bogoni F, Pernitsch D, Kavertseva M, Bourgeois B, Kukilo J, Tehlivets YG, Schwarz AN, Züllig T, Bubalo V, Schauer S, Groselj-Strele A, Hoefler G, Rechberger GN, Herrmann M, Eller K, Rosenkranz AR, Madl T, Frank S, Holzapfel GA, Kratky D, Mangge H, Hörl G. Homocysteine contributes to atherogenic transformation of the aorta in rabbits in the absence of hypercholesterolemia. Biomed Pharmacother 2024; 178:117244. [PMID: 39116783 DOI: 10.1016/j.biopha.2024.117244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Atherosclerosis, the leading cause of cardiovascular disease, cannot be sufficiently explained by established risk factors, including cholesterol. Elevated plasma homocysteine (Hcy) is an independent risk factor for atherosclerosis and is closely linked to cardiovascular mortality. However, its role in atherosclerosis has not been fully clarified yet. We have previously shown that rabbits fed a diet deficient in B vitamins and choline (VCDD), which are required for Hcy degradation, exhibit an accumulation of macrophages and lipids in the aorta, aortic stiffening and disorganization of aortic collagen in the absence of hypercholesterolemia, and an aggravation of atherosclerosis in its presence. In the current study, plasma Hcy levels were increased by intravenous injections of Hcy into balloon-injured rabbits fed VCDD (VCDD+Hcy) in the absence of hypercholesterolemia. While this treatment did not lead to thickening of aortic wall, intravenous injections of Hcy into rabbits fed VCDD led to massive accumulation of VLDL-triglycerides as well as significant impairment of vascular reactivity of the aorta compared to VCDD alone. In the aorta intravenous Hcy injections into VCDD-fed rabbits led to fragmentation of aortic elastin, accumulation of elastin-specific electron-dense inclusions, collagen disorganization, lipid degradation, and autophagolysosome formation. Furthermore, rabbits from the VCDD+Hcy group exhibited a massive decrease of total protein methylated arginine in blood cells and decreased creatine in blood cells, serum and liver compared to rabbits from the VCDD group. Altogether, we conclude that Hcy contributes to atherogenic transformation of the aorta not only in the presence but also in the absence of hypercholesterolemia.
Collapse
Affiliation(s)
- Oksana Tehlivets
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Division of General Radiology, Department of Radiology, Medical University of Graz, Graz, Austria.
| | - Gunter Almer
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Markus S Brunner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Margarete Lechleitner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gerhard Sommer
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria; Center for Medical Research, Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Clemens Diwoky
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Hansjörg Habisch
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Peter Opriessnig
- Division of General Neurology, Department of Neurology, Medical University of Graz, Graz, Austria; Division of Pediatric Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Francesca Bogoni
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Dominique Pernitsch
- Center for Medical Research, Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Maria Kavertseva
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Benjamin Bourgeois
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Jelena Kukilo
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Yuriy G Tehlivets
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Andreas N Schwarz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Thomas Züllig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Vladimir Bubalo
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | - Silvia Schauer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Andrea Groselj-Strele
- Center for Medical Research, Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Markus Herrmann
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Kathrin Eller
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | | | - Tobias Madl
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria; Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Gerd Hörl
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| |
Collapse
|
3
|
Song T, Cerruti M. Unraveling the role of carboxylate groups and elastin particle size in medial calcification. Int J Biol Macromol 2024; 274:133267. [PMID: 38906359 DOI: 10.1016/j.ijbiomac.2024.133267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
While it is known that calcium phosphate (CaP) minerals deposit in elastin-rich medial layers of arteries during medial calcification, their nucleation and growth sites are still debated. Neutral carbonyl groups and carboxylate groups are possible candidates. Also, while it is known that elastin degradation leads to calcification, it is unclear whether this is due to formation of new carboxylate groups or elastin fragmentation. In this work, we disentangle effects of carboxylate groups and particle size on elastin calcification; in doing so, we shed light on CaP mineralization sites on elastin. We find carboxylate groups accelerate calcification only in early stages; they mainly function as Ca2+ ion chelation sites but not calcification sites. Their presence promotes formation (likely on Ca2+ ions adsorbed on nearby carbonyl groups) of CaP minerals with high calcium-to-phosphate ratio as intermediate phases. Larger elastin particles calcify slower but reach similar amounts of CaP minerals in late stages; they promote direct formation of hydroxyapatite and CaP minerals with low calcium-to-phosphate ratio as intermediate phases. This work provides new perspectives on how carboxylate groups and elastin particle size influence calcification; these parameters can be tuned to study the mechanism of medial calcification and design drugs to inhibit the process.
Collapse
Affiliation(s)
- Tao Song
- Department of Mining and Materials Engineering, McGill University, Montreal, Quebec H3A 0C5, Canada.
| | - Marta Cerruti
- Department of Mining and Materials Engineering, McGill University, Montreal, Quebec H3A 0C5, Canada.
| |
Collapse
|
4
|
Fan Y, Moser J, van Meurs M, Kiers D, Sand JMB, Leeming DJ, Pickkers P, Burgess JK, Kox M, Pillay J. Neo-epitope detection identifies extracellular matrix turnover in systemic inflammation and sepsis: an exploratory study. Crit Care 2024; 28:120. [PMID: 38609959 PMCID: PMC11010428 DOI: 10.1186/s13054-024-04904-4] [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: 01/31/2024] [Accepted: 04/06/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Sepsis is associated with high morbidity and mortality, primarily due to systemic inflammation-induced tissue damage, resulting organ failure, and impaired recovery. Regulated extracellular matrix (ECM) turnover is crucial for maintaining tissue homeostasis in health and in response to disease-related changes in the tissue microenvironment. Conversely, uncontrolled turnover can contribute to tissue damage. Systemic Inflammation is implicated to play a role in the regulation of ECM turnover, but the relationship between the two is largely unclear. METHODS We performed an exploratory study in 10 healthy male volunteers who were intravenously challenged with 2 ng/kg lipopolysaccharide (LPS, derived from Escherichia coli) to induce systemic inflammation. Plasma samples were collected before (T0) and after (T 1 h, 3 h, 6 h and 24 h) the LPS challenge. Furthermore, plasma was collected from 43 patients with septic shock on day 1 of ICU admission. Circulating neo-epitopes of extracellular matrix turnover, including ECM degradation neo-epitopes of collagen type I (C1M), type III (C3M), type IV (C4Ma3), and type VI (C6M), elastin (ELP-3) and fibrin (X-FIB), as well as the ECM synthesis neo-epitopes of collagen type III (PRO-C3), collagen type IV (PRO-C4) and collagen type VI (PRO-C6) were measured by ELISA. Patient outcome data were obtained from electronic patient records. RESULTS Twenty-four hours after LPS administration, all measured ECM turnover neo-epitopes, except ELP-3, were increased compared to baseline levels. In septic shock patients, concentrations of all measured ECM neo-epitopes were higher compared to healthy controls. In addition, concentrations of C6M, ELP-3 and X-FIB were higher in patients with septic shock who ultimately did not survive (N = 7) compared to those who recovered (N = 36). CONCLUSION ECM turnover is induced in a model of systemic inflammation in healthy volunteers and was observed in patients with septic shock. Understanding interactions between systemic inflammation and ECM turnover may provide further insight into mechanisms underlying acute and persistent organ failure in sepsis.
Collapse
Affiliation(s)
- YiWen Fan
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- University Medical Center Groningen, Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Jill Moser
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Dorien Kiers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janette K Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janesh Pillay
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
- University Medical Center Groningen, Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
5
|
Tashiro R, Anzawa R, Inoue T, Mikagi A, Ozaki D, Tominaga K, Inoue T, Ishida T, Fujimura M, Usuki T, Endo H, Niizuma K, Tominaga T. The prognostic values of plasma desmosines, crosslinking molecules of elastic fibers, in the disease progression of Moyamoya disease. Bioorg Med Chem 2024; 100:117602. [PMID: 38324946 DOI: 10.1016/j.bmc.2024.117602] [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: 12/12/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/09/2024]
Abstract
Moyamoya disease (MMD) is a cerebrovascular disease which is characterized by the chronic progression of steno-occlusive changes at the terminal portion of internal carotid arteries and the development of "moyamoya vessels." Dysregulation of the extracellular matrix is regarded as a key pathophysiology underlying unique vascular remodeling. Here, we measured the concentration of elastin crosslinkers desmosine and isodesmosine in the plasma of MMD patients. We aimed to reveal its diagnostic values of desmosines in the progression of steno-occlusive lesions. The concentrations of plasma desmosines were determined by liquid chromatography-tandem mass spectrometry. The temporal profiles of steno-occlusive lesions on magnetic resonance angiography were retrospectively evaluated, and the correlation between the progression of steno-occlusive changes in intracranial arteries and plasma desmosines concentrations was further analyzed. Plasma desmosines were significantly higher in MMD patients with disease progression compared to MMD patients without disease progression. Also, the incidence of disease progression was higher in MMD patients with plasma desmosines levels over limit of quantitation (LOQ) than those with plasma desmosines levels below LOQ. In conclusion, plasma desmosines could be potential biomarkers to predict the progression of steno-occlusive changes in MMD patients.
Collapse
Affiliation(s)
- Ryosuke Tashiro
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Riki Anzawa
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Tomoo Inoue
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| | - Ayame Mikagi
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Dan Ozaki
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Keita Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Takashi Inoue
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Tomohisa Ishida
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo 060-0815, Japan
| | - Toyonobu Usuki
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Chiyoda-ku, Tokyo 102-8554, Japan.
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Biomedical Engineering, Sendai 980-8575, Japan; Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| |
Collapse
|
6
|
Halsey G, Sinha D, Dhital S, Wang X, Vyavahare N. Role of elastic fiber degradation in disease pathogenesis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166706. [PMID: 37001705 PMCID: PMC11659964 DOI: 10.1016/j.bbadis.2023.166706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
Elastin is a crucial extracellular matrix protein that provides structural integrity to tissues. Crosslinked elastin and associated microfibrils, named elastic fiber, contribute to biomechanics by providing the elasticity required for proper function. During aging and disease, elastic fiber can be progressively degraded and since there is little elastin synthesis in adults, degraded elastic fiber is not regenerated. There is substantial evidence linking loss or damage of elastic fibers to the clinical manifestation and pathogenesis of a variety of diseases. Disruption of elastic fiber networks by hereditary mutations, aging, or pathogenic stimuli results in systemic ailments associated with the production of elastin degradation products, inflammatory responses, and abnormal physiology. Due to its longevity, unique mechanical properties, and widespread distribution in the body, elastic fiber plays a central role in homeostasis of various physiological systems. While pathogenesis related to elastic fiber degradation has been more thoroughly studied in elastic fiber rich tissues such as the vasculature and the lungs, even tissues containing relatively small quantities of elastic fibers such as the eyes or joints may be severely impacted by elastin degradation. Elastic fiber degradation is a common observation in certain hereditary, age, and specific risk factor exposure induced diseases representing a converging point of pathological clinical phenotypes which may also help explain the appearance of co-morbidities. In this review, we will first cover the role of elastic fiber degradation in the manifestation of hereditary diseases then individually explore the structural role and degradation effects of elastic fibers in various tissues and organ systems. Overall, stabilizing elastic fiber structures and repairing lost elastin may be effective strategies to reverse the effects of these diseases.
Collapse
Affiliation(s)
- Gregory Halsey
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Dipasha Sinha
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Saphala Dhital
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Xiaoying Wang
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Naren Vyavahare
- Department of Bioengineering, Clemson University, SC 29634, United States of America.
| |
Collapse
|
7
|
Magnesium Improves Cardiac Function in Experimental Uremia by Altering Cardiac Elastin Protein Content. Nutrients 2023; 15:nu15061303. [PMID: 36986034 PMCID: PMC10056411 DOI: 10.3390/nu15061303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Cardiovascular complications are accompanied by life-threatening complications and represent the major cause of death in patients with chronic kidney disease (CKD). Magnesium is important for the physiology of cardiac function, and its deficiency is common in CKD. In the present study, we investigated the impact of oral magnesium carbonate supplementation on cardiac function in an experimental model of CKD induced in Wistar rats by an adenine diet. Echocardiographic analyses revealed restoration of impaired left ventricular cardiac function in animals with CKD. Cardiac histology and real-time PCR confirmed a high amount of elastin protein and increased collagen III expression in CKD rats supplemented with dietary magnesium as compared with CKD controls. Both structural proteins are crucial in maintaining cardiac health and physiology. Aortic calcium content increased in CKD as compared with tissue from control animals. Magnesium supplementation numerically lowered the increases in aortic calcium content as it remained statistically unchanged, compared with controls. In summary, the present study provides evidence for an improvement in cardiovascular function and aortic wall integrity in a rat model of CKD by magnesium, as evidenced by echocardiography and histology.
Collapse
|
8
|
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
|
9
|
Anthoulakis C, Mamopoulos A, Rousso D, Karagiannis A, Athanasiadis A, Grimbizis G, Athyros V. Arterial Stiffness as a Cardiovascular Risk Factor for the Development of Preeclampsia and Pharmacopreventive Options. Curr Vasc Pharmacol 2021; 20:52-61. [PMID: 34615450 DOI: 10.2174/1570161119666211006114258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
Arterial stiffness (AS) describes the rigidity of the arterial walls. Epidemiological studies have shown that increased AS is an independent predictive marker of cardiovascular (CV) morbidity and mortality in both pregnant and non-pregnant women. Preeclampsia (PE), a form of pregnancy-induced hypertension, affects approximately 5% of pregnancies worldwide. Preeclamptic women have a higher risk of CV disease (CVD), mainly because PE damages the heart's ability to relax between contractions. Different pharmacological approaches for the prevention of PE have been tested in clinical trials (e.g. aspirin, enoxaparin, metformin, pravastatin, and sildenafil citrate). In current clinical practice, only low-dose aspirin is used for PE pharmacoprevention. However, low-dose aspirin does not prevent term PE, which is the most common form of PE. Compromised vascular integrity precedes the onset of PE and therefore, AS assessment may constitute a promising predictive marker of PE. Several non-invasive techniques have been developed to assess AS. Compared with normotensive pregnancies, both carotid-femoral pulse wave velocity (cfPWV) and augmentation index (AIx) are increased in PE. In view of simplicity, reliability, and reproducibility, there is an interest in oscillometric AS measurements in pregnancies complicated by PE.
Collapse
Affiliation(s)
- Christos Anthoulakis
- First Department of Obstetrics & Gynecology, "Papageorgiou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Apostolos Mamopoulos
- Third Department of Obstetrics & Gynecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - David Rousso
- Third Department of Obstetrics & Gynecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Asterios Karagiannis
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Apostolos Athanasiadis
- Third Department of Obstetrics & Gynecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Grigoris Grimbizis
- First Department of Obstetrics & Gynecology, "Papageorgiou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Vasilios Athyros
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| |
Collapse
|
10
|
Adeva-Andany MM, Adeva-Contreras L, Fernández-Fernández C, González-Lucán M, Funcasta-Calderón R. Elastic tissue disruption is a major pathogenic factor to human vascular disease. Mol Biol Rep 2021; 48:4865-4878. [PMID: 34129188 DOI: 10.1007/s11033-021-06478-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/05/2021] [Indexed: 01/15/2023]
Abstract
Elastic fibers are essential components of the arterial extracellular matrix. They consist of the protein elastin and an array of microfibrils that support the protein and connect it to the surrounding matrix. The elastin gene encodes tropoelastin, a protein that requires extensive cross-linking to become elastin. Tropoelastin is expressed throughout human life, but its expression levels decrease with age, suggesting that the potential to synthesize elastin persists during lifetime although declines with aging. The initial abnormality documented in human atherosclerosis is fragmentation and loss of the elastic network in the medial layer of the arterial wall, suggesting an imbalance between elastic fiber injury and restoration. Damaged elastic structures are not adequately repaired by synthesis of new elastic elements. Progressive collagen accumulation follows medial elastic fiber disruption and fibrous plaques are formed, but advanced atherosclerosis lesions do not develop in the absence of prior elastic injury. Aging is associated with arterial extracellular matrix anomalies that evoke those present in early atherosclerosis. The reduction of elastic fibers with subsequent collagen accumulation leads to arterial stiffening and intima-media thickening, which are independent predictors of incident hypertension in prospective community-based studies. Arterial stiffening precedes the development of hypertension. The fundamental role of the vascular elastic network to arterial structure and function is emphasized by congenital disorders caused by mutations that disrupt normal elastic fiber production. Molecular changes in the genes coding tropoelastin, lysyl oxidase (tropoelastin cross-linking), and elastin-associated microfibrils, including fibrillin-1, fibulin-4, and fibulin-5 produce severe vascular injury due to absence of functional elastin.
Collapse
Affiliation(s)
- María M Adeva-Andany
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain.
| | | | - Carlos Fernández-Fernández
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Manuel González-Lucán
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Raquel Funcasta-Calderón
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| |
Collapse
|
11
|
Zhao S, Cao J, Li J, Yang X, Cao P, Lan J, Lu G. Association between serum elastin-derived peptides and abdominal aortic calcification in peritoneal dialysis patients: a cross-sectional study. Ren Fail 2021; 43:860-868. [PMID: 33993833 PMCID: PMC8143601 DOI: 10.1080/0886022x.2021.1918163] [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] [Indexed: 12/19/2022] Open
Abstract
Background Peritoneal dialysis (PD) patients experience accelerated arterial aging, which is characterized by elastin degradation. Elastin-derived peptides (EDPs) are direct products of elastin fragmentation. This study tried to explore the association between serum EDPs and abdominal aortic calcification (AAC) in PD patients. Methods Serum levels of EDPs were analyzed in 126 eligible PD patients and 30 controls. PD patients were grouped according to the annularity of AAC evaluated by an abdominal computed tomography (CT) scan. Serum EDPs were analyzed in relation to the presence of AAC or severe AAC in PD patients by logistic regression analysis. Results Serum EDPs in PD patients were significantly higher than age-matched controls. In 126 PD patients, higher EDPs was associated with greater risk of present AAC (OR = 1.056, 95%CI 1.010–1.103) and severe AAC (OR = 1.062, 95%CI 1.004–1.123). A combination of EDPs substantially improved the accuracy of diagnostic performance for AAC and severe AAC. Conclusions EDPs can predict the presence and extent of AAC in PD patients, indicating its possible role to recognize PD patients at risk for AAC and severe AAC.
Collapse
Affiliation(s)
- Shizhu Zhao
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingyuan Cao
- Department of Nephrology, Taizhou People's hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, China
| | - Jianzhong Li
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaochun Yang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Peiyang Cao
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingjing Lan
- Department of Nephology, Wuxi Traditional Chinese Medicine Hospital, Wuxi, China
| | - Guoyuan Lu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Lin SC, Wu TJ, Wu DA, Hsu BG. Hypoadiponectinemia is associated with aortic stiffness in nondialysis diabetic patients with stage 3-5 chronic kidney disease. Vascular 2021; 30:384-391. [PMID: 33866881 DOI: 10.1177/17085381211007602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Albuminuria and serum adiponectin levels are factors that have been associated with the development of cardiovascular disease in patients with diabetes mellitus. Here we investigated the relationship between serum adiponectin levels and aortic stiffness in nondialysis diabetic kidney disease patients with stage 3-5 chronic kidney disease. METHODS Fasting blood samples were obtained from 80 nondialysis diabetic kidney disease patients with stage 3-5 chronic kidney disease. Carotid-femoral pulse wave velocity (cfPWV) was measured using applanation tonometry; cfPWV values of >10 m/s were defined as aortic stiffness. Serum adiponectin levels were determined by enzyme immunoassay. RESULTS Forty-two patients (52.5%) with nondialysis diabetic kidney disease were diagnosed with aortic stiffness. The patients in this group were older (p = 0.011), had higher systolic blood pressure (p = 0.002) and urine albumin-to-creatinine ratios (p = 0.013), included fewer females (p = 0.024), and had lower serum adiponectin (p = 0.001) levels than those in the control group. Multivariable logistic regression analysis revealed that serum adiponectin was independently associated with aortic stiffness (odds ratio = 0.930, 95% confidence interval: 0.884-0.978, p = 0.005) and also positively correlated with cfPWV values by multivariable linear regression (β = -0.309, p = 0.002) in nondialysis diabetic kidney disease patients. CONCLUSIONS The results suggested that serum adiponectin levels could be used to predict aortic stiffness in nondialysis diabetic kidney disease patients with stage 3-5 chronic kidney disease.
Collapse
Affiliation(s)
- Ssu-Chin Lin
- Division of Nephrology, Department of Medicine, Hualien Armed Forces General Hospital, Hualien.,Department of Nursing, Hualien Armed Forces General Hospital, Hualien
| | - Tsung-Jui Wu
- Division of Nephrology, Department of Medicine, Hualien Armed Forces General Hospital, Hualien
| | - Du-An Wu
- Division of Metabolism and Endocrinology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,School of Medicine, Tzu Chi University, Hualien
| | - Bang-Gee Hsu
- School of Medicine, Tzu Chi University, Hualien.,Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
| |
Collapse
|
13
|
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
|
14
|
Balestrini CS, Al-Khazraji BK, Suskin N, Shoemaker JK. Does vascular stiffness predict white matter hyperintensity burden in ischemic heart disease with preserved ejection fraction? Am J Physiol Heart Circ Physiol 2020; 318:H1401-H1409. [PMID: 32357114 DOI: 10.1152/ajpheart.00057.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The survival rate of patients with ischemic heart disease (IHD) is increasing. However, survivors experience increased risk for neurological complications. The mechanisms for this increased risk are unknown. We tested the hypothesis that patients with IHD have greater carotid and cerebrovascular stiffness, and these indexes predict white matter small vessel disease. Fifty participants (age, 40-78 yr), 30 with IHD with preserved ejection fraction and 20 healthy age-matched controls, were studied using ultrasound imaging of the common carotid artery (CCA) and middle cerebral artery (MCA), as well as magnetic resonance imaging (T1, T2-FLAIR), to measure white matter lesion volume (WMLv). Carotid β-stiffness provided the primary measure of peripheral vascular stiffness. Carotid-cerebral pulse wave transit time (ccPWTT) provided a marker of cerebrovascular stiffness. Pulsatility index (PI) and resistive index (RI) of the MCA were calculated as measures of downstream cerebrovascular resistance. When compared with controls, patients with IHD exhibited greater β-stiffness [8.5 ± 3.3 vs. 6.8 ± 2.2 arbitrary units (AU); P = 0.04], MCA PI (1.1 ± 0.20 vs. 0.98 ± 0.18 AU; P = 0.02), and MCA RI (0.66 ± 0.06 vs. 0.62 ± 0.07 AU; P = 0.04). There was no difference in WMLv between IHD and control groups (0.95 ± 1.2 vs. 0.86 ± 1.4 mL; P = 0.81). In pooled patient data, WMLv correlated with both β-stiffness (R = 0.34, P = 0.02) and cerebrovascular ccPWTT (R = -0.43, P = 0.02); however, β-stiffness and ccPWTT were not associated (P = 0.13). In multivariate analysis, WMLv remained independently associated with ccPWTT (P = 0.02) and carotid β-stiffness (P = 0.04). Patients with IHD expressed greater β-stiffness and cerebral microvascular resistance. However, IHD did not increase risk of WMLv or cerebrovascular stiffness. Nonetheless, pooled data indicate that both carotid and cerebrovascular stiffness are independently associated with WMLv.NEW & NOTEWORTHY This study found that patients with ischemic heart disease (IHD) with preserved ejection fraction and normal blood pressures exhibit greater carotid β-stiffness, as well as middle cerebral artery pulsatility and resistive indexes, than controls. White matter lesion volume (WMLv) was not different between vascular pathology groups. Cerebrovascular pulse wave transit time (ccPWTT) and carotid β-stiffness independently associate with WMLv in pooled participant data, suggesting that regardless of heart disease history, ccPWTT and β-stiffness are associated with structural white matter damage.
Collapse
Affiliation(s)
| | | | - Neville Suskin
- Cardiac Rehabilitation and Secondary Prevention Program of Saint Joseph's Health Care London, London, Ontario, Canada.,Division of Cardiology, Department of Medicine, and Program of Experimental Medicine, Western University, London, Ontario, Canada
| | - J Kevin Shoemaker
- School of Kinesiology, Western University, London, Ontario, Canada.,Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| |
Collapse
|
15
|
Khaltourina D, Matveyev Y, Alekseev A, Cortese F, Ioviţă A. Aging Fits the Disease Criteria of the International Classification of Diseases. Mech Ageing Dev 2020; 189:111230. [PMID: 32251691 DOI: 10.1016/j.mad.2020.111230] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 12/24/2022]
Abstract
The disease criteria used by the World Health Organization (WHO) were applied to human biological aging in order to assess whether aging can be classified as a disease. These criteria were developed for the 11th revision of the International Classification of Diseases (ICD) and included disease diagnostics, mechanisms, course and outcomes, known interventions, and linkage to genetic and environmental factors. RESULTS: Biological aging can be diagnosed with frailty indices, functional, blood-based biomarkers. A number of major causal mechanisms of human aging involved in various organs have been described, such as inflammation, replicative cellular senescence, immune senescence, proteostasis failures, mitochondrial dysfunctions, fibrotic propensity, hormonal aging, body composition changes, etc. We identified a number of clinically proven interventions, as well as genetic and environmental factors of aging. Therefore, aging fits the ICD-11 criteria and can be considered a disease. Our proposal was submitted to the ICD-11 Joint Task force, and this led to the inclusion of the extension code for "Ageing-related" (XT9T) into the "Causality" section of the ICD-11. This might lead to greater focus on biological aging in global health policy and might provide for more opportunities for the new therapy developers.
Collapse
Affiliation(s)
- Daria Khaltourina
- Department of Risk Factor Prevention, Federal Research Institute for Health Organization and Informatics of Ministry of Health of the Russian Federation, Dobrolyubova St. 11, Moscow, 127254, Russia; International Longevity Alliance, 19 avenue Jean Jaurès, Sceaux, 92330, France.
| | - Yuri Matveyev
- Research Lab, Moscow Regional Research and Clinical Institute, Schepkina St. 61/2 k.1, Moscow, 129110, Russia
| | - Aleksey Alekseev
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory, GSP-1, Moscow, 119991, Russia
| | - Franco Cortese
- Biogerontology Research Foundation, Apt 2354 Chynoweth House, Trevissome Park, Truro, London, TR4 8UN, UK
| | - Anca Ioviţă
- International Longevity Alliance, 19 avenue Jean Jaurès, Sceaux, 92330, France
| |
Collapse
|
16
|
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
|
17
|
Smith ER, Hewitson TD, Holt SG. Diagnostic Tests for Vascular Calcification. Adv Chronic Kidney Dis 2019; 26:445-463. [PMID: 31831123 DOI: 10.1053/j.ackd.2019.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/22/2019] [Accepted: 07/28/2019] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is the heterogeneous endpoint of multiple vascular insults, which varies by arterial bed, the layer of the arterial wall affected, and is propagated by diverse cellular and biochemical mechanisms. A variety of in vivo and ex vivo techniques have been applied to the analysis of VC in preclinical studies, but clinical examination has principally relied on a number of noninvasive and invasive imaging modalities for detection and quantitation. Most imaging methods suffer from suboptimal spatial resolution, leading to the inability to distinguish medial from intimal VC and insufficient sensitivity to detect microcalcifications that are indicative of active mineral deposition and of vulnerable plaques which may be prone to rupture. Serum biomarkers lack specificity for VC and cannot discriminate pathology. Overall, uncertainties surrounding the sensitivity and specificity of different VC testing modalities, the absence of a clear cause-effect relationship, and lack of any evidence-based diagnostic or therapeutic protocols in relation to VC testing in chronic kidney disease has yielded weak or ungraded recommendations for their use in clinical practice. While VC is recognized as a key manifestation of chronic kidney disease-mineral and bone disorder and those with an increasing burden of VC are considered to be at higher cardiovascular risk, routine screening is not currently recommended.
Collapse
|
18
|
Lee SJ, Avolio A, Seo DC, Kim BS, Kang JH, Lee MY, Sung KC. Relationship Between Brachial-Ankle Pulse Wave Velocity and Incident Hypertension According to 2017 ACC/AHA High Blood Pressure Guidelines. J Am Heart Assoc 2019; 8:e013019. [PMID: 31412746 PMCID: PMC6759909 DOI: 10.1161/jaha.119.013019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background Arterial stiffness predicts both cardiovascular events and incident hypertension. However, whether brachial‐ankle pulse wave velocity (baPWV) is predictive of incident hypertension based on the 2017 American College of Cardiology/American Heart Association (ACC/AHA) High Blood Pressure Guidelines has not been established. We performed a large cohort study to investigate whether incident hypertension could be predicted from baPWV measurements as a measure of arterial stiffness, even when applying updated hypertension criteria. Methods and Results A total of 10 360 Korean adults who underwent baPWV examination during a health‐screening program between 2010 and 2016 were enrolled. Hypertension was defined according to the 2017 ACC/AHA Guidelines as 130/80 mm Hg. Cox proportional hazard analysis was used to assess the risk of incident hypertension according to baPWV quartiles. The mean age of the study subjects was 40.2 years and 75.6% were men. During the follow‐up period (median 2.17 years), 2000 subjects (19.3%) developed hypertension. The subjects in the highest baPWV quartile group showed an increased risk of hypertension compared with the lowest baPWV quartile group as confirmed by multivariate adjusted hazard ratios of 1.64 (95% CI 1.41–1.89; P<0.001) in men and 12.36 (95% CI 4.41–34.62; P=0.005) in women. The increased risk of developing hypertension was consistent after adjusting for several confounding factors. Conclusions Arterial stiffness measured by baPWV is associated with incident hypertension according to the updated 2017 ACC/AHA Guidelines and is a useful independent predictor of incident hypertension among relatively healthy people.
Collapse
Affiliation(s)
- Seung Jae Lee
- Division of Cardiology Department of Internal Medicine Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Alberto Avolio
- Department of Biomedical Sciences Faculty of Medicine and Health Sciences Macquarie University Sydney New South Wales Australia
| | - Dae Chul Seo
- Division of Cardiology Department of Internal Medicine Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Bum Soo Kim
- Division of Cardiology Department of Internal Medicine Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Jin Ho Kang
- Division of Cardiology Department of Internal Medicine Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Mi Yeon Lee
- Division of Biostatistics Department of R&D Management Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Ki-Chul Sung
- Division of Cardiology Department of Internal Medicine Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine Seoul Republic of Korea
| |
Collapse
|
19
|
Perrault R, Omelchenko A, Taylor CG, Zahradka P. Establishing the interchangeability of arterial stiffness but not endothelial function parameters in healthy individuals. BMC Cardiovasc Disord 2019; 19:190. [PMID: 31387535 PMCID: PMC6685177 DOI: 10.1186/s12872-019-1167-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 07/23/2019] [Indexed: 12/15/2022] Open
Abstract
Background Development of instruments capable of detecting early stage vascular disease has increased interest in employing arterial stiffness (e.g. pulse wave velocity (PWV), augmentation index (AIx)) and endothelial dysfunction (e.g. reactive hyperemia index (RHI)) to diagnose atherosclerotic disease before occurrence of a cardiovascular event. However, amongst the equipment designed for this purpose, there is insufficient information regarding each of these parameters to establish appropriate cutoffs to distinguish between healthy and unhealthy blood vessels. To address these limitations, the study was designed to establish the upper arterial stiffness and endothelial function thresholds in a healthy population, by comparing the outputs from different instruments capable of measuring PWV, AIx and RHI. Methods A systematic comparison of PWV, AIx and RHI was conducted to determine the inter-relationships between these parameters of vascular functionality. Outputs were obtained non-invasively using three instruments, the VP-1000 (VP), SphygmoCor (SC), and EndoPAT (EP), in 40 apparently healthy males and females. Results Correlations were found between the brachial-ankle PWV and radial-ankle PWV (by VP and SC), and PWV (VP) with AIx (SC). The interchangeability of these outputs was demonstrated by the Bland Altman test, making it feasible to extrapolate cut-offs for radial-ankle PWV and AIx equivalent to brachial-ankle PWV that signify healthy vessels. In contrast, RHI showed no association with AIx, suggesting these endothelial and arterial parameters are functionally distinct. Conclusions It was concluded that it is possible to compare the vascular function outputs of different instruments and identify healthy from unhealthy vessels, even though the approaches for quantifying the underlying physiological processes may differ. In this way, non-invasive determination of arterial function could be a new paradigm for detecting existing early stage asymptomatic atherosclerotic disease in individuals using techniques that are amenable to the clinical setting. Electronic supplementary material The online version of this article (10.1186/s12872-019-1167-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raissa Perrault
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada.,Department of Physiology and Pathophysiology, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Alexander Omelchenko
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Carla G Taylor
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada.,Department of Physiology and Pathophysiology, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Department of Food and Human Nutritional Sciences, Faculty of Agriculture and Food Science, University of Manitoba, Winnipeg, Canada
| | - Peter Zahradka
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada. .,Department of Physiology and Pathophysiology, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada. .,Department of Food and Human Nutritional Sciences, Faculty of Agriculture and Food Science, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
20
|
Reesink KD, Spronck B. Constitutive interpretation of arterial stiffness in clinical studies: a methodological review. Am J Physiol Heart Circ Physiol 2019; 316:H693-H709. [DOI: 10.1152/ajpheart.00388.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Clinical assessment of arterial stiffness relies on noninvasive measurements of regional pulse wave velocity or local distensibility. However, arterial stiffness measures do not discriminate underlying changes in arterial wall constituent properties (e.g., in collagen, elastin, or smooth muscle), which is highly relevant for development and monitoring of treatment. In arterial stiffness in recent clinical-epidemiological studies, we systematically review clinical-epidemiological studies (2012–) that interpreted arterial stiffness changes in terms of changes in arterial wall constituent properties (63 studies included of 514 studies found). Most studies that did so were association studies (52 of 63 studies) providing limited causal evidence. Intervention studies (11 of 63 studies) addressed changes in arterial stiffness through the modulation of extracellular matrix integrity (5 of 11 studies) or smooth muscle tone (6 of 11 studies). A handful of studies (3 of 63 studies) used mathematical modeling to discriminate between extracellular matrix components. Overall, there exists a notable gap in the mechanistic interpretation of stiffness findings. In constitutive model-based interpretation, we first introduce constitutive-based modeling and use it to illustrate the relationship between constituent properties and stiffness measurements (“forward” approach). We then review all literature on modeling approaches for the constitutive interpretation of clinical arterial stiffness data (“inverse” approach), which are aimed at estimation of constitutive properties from arterial stiffness measurements to benefit treatment development and monitoring. Importantly, any modeling approach requires a tradeoff between model complexity and measurable data. Therefore, the feasibility of changing in vivo the biaxial mechanics and/or vascular smooth muscle tone should be explored. The effectiveness of modeling approaches should be confirmed using uncertainty quantification and sensitivity analysis. Taken together, constitutive modeling can significantly improve clinical interpretation of arterial stiffness findings.
Collapse
Affiliation(s)
- Koen D. Reesink
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Bart Spronck
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, Connecticut
| |
Collapse
|
21
|
Vizovišek M, Fonović M, Turk B. Cysteine cathepsins in extracellular matrix remodeling: Extracellular matrix degradation and beyond. Matrix Biol 2019; 75-76:141-159. [DOI: 10.1016/j.matbio.2018.01.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/14/2018] [Accepted: 01/29/2018] [Indexed: 12/21/2022]
|
22
|
Ngai D, Lino M, Bendeck MP. Cell-Matrix Interactions and Matricrine Signaling in the Pathogenesis of Vascular Calcification. Front Cardiovasc Med 2018; 5:174. [PMID: 30581820 PMCID: PMC6292870 DOI: 10.3389/fcvm.2018.00174] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification is a complex pathological process occurring in patients with atherosclerosis, type 2 diabetes, and chronic kidney disease. The extracellular matrix, via matricrine-receptor signaling plays important roles in the pathogenesis of calcification. Calcification is mediated by osteochondrocytic-like cells that arise from transdifferentiating vascular smooth muscle cells. Recent advances in our understanding of the plasticity of vascular smooth muscle cell and other cells of mesenchymal origin have furthered our understanding of how these cells transdifferentiate into osteochondrocytic-like cells in response to environmental cues. In the present review, we examine the role of the extracellular matrix in the regulation of cell behavior and differentiation in the context of vascular calcification. In pathological calcification, the extracellular matrix not only provides a scaffold for mineral deposition, but also acts as an active signaling entity. In recent years, extracellular matrix components have been shown to influence cellular signaling through matrix receptors such as the discoidin domain receptor family, integrins, and elastin receptors, all of which can modulate osteochondrocytic differentiation and calcification. Changes in extracellular matrix stiffness and composition are detected by these receptors which in turn modulate downstream signaling pathways and cytoskeletal dynamics, which are critical to osteogenic differentiation. This review will focus on recent literature that highlights the role of cell-matrix interactions and how they influence cellular behavior, and osteochondrocytic transdifferentiation in the pathogenesis of cardiovascular calcification.
Collapse
Affiliation(s)
- David Ngai
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Marsel Lino
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Copper as the most likely pathogenic divergence factor between lung fibrosis and emphysema. Med Hypotheses 2018; 120:49-54. [DOI: 10.1016/j.mehy.2018.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/04/2018] [Indexed: 01/17/2023]
|
24
|
Coenen AMJ, Bernaerts KV, Harings JAW, Jockenhoevel S, Ghazanfari S. Elastic materials for tissue engineering applications: Natural, synthetic, and hybrid polymers. Acta Biomater 2018; 79:60-82. [PMID: 30165203 DOI: 10.1016/j.actbio.2018.08.027] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/03/2018] [Accepted: 08/21/2018] [Indexed: 02/08/2023]
Abstract
Elastin and collagen are the two main components of elastic tissues and provide the tissue with elasticity and mechanical strength, respectively. Whereas collagen is adequately produced in vitro, production of elastin in tissue-engineered constructs is often inadequate when engineering elastic tissues. Therefore, elasticity has to be artificially introduced into tissue-engineered scaffolds. The elasticity of scaffold materials can be attributed to either natural sources, when native elastin or recombinant techniques are used to provide natural polymers, or synthetic sources, when polymers are synthesized. While synthetic elastomers often lack the biocompatibility needed for tissue engineering applications, the production of natural materials in adequate amounts or with proper mechanical strength remains a challenge. However, combining natural and synthetic materials to create hybrid components could overcome these issues. This review explains the synthesis, mechanical properties, and structure of native elastin as well as the theories on how this extracellular matrix component provides elasticity in vivo. Furthermore, current methods, ranging from proteins and synthetic polymers to hybrid structures that are being investigated for providing elasticity to tissue engineering constructs, are comprehensively discussed. STATEMENT OF SIGNIFICANCE Tissue engineered scaffolds are being developed as treatment options for malfunctioning tissues throughout the body. It is essential that the scaffold is a close mimic of the native tissue with regards to both mechanical and biological functionalities. Therefore, the production of elastic scaffolds is of key importance to fabricate tissue engineered scaffolds of the elastic tissues such as heart valves and blood vessels. Combining naturally derived and synthetic materials to reach this goal proves to be an interesting area where a highly tunable material that unites mechanical and biological functionalities can be obtained.
Collapse
Affiliation(s)
- Anna M J Coenen
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Katrien V Bernaerts
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Jules A W Harings
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Stefan Jockenhoevel
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands; Department of Biohybrid & Medical Textiles (BioTex), AME-Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Forckenbeckstraβe 55, 52072 Aachen, Germany
| | - Samaneh Ghazanfari
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands.
| |
Collapse
|
25
|
Salesse S, Odoul L, Chazée L, Garbar C, Duca L, Martiny L, Mahmoudi R, Debelle L. Elastin molecular aging promotes MDA-MB-231 breast cancer cell invasiveness. FEBS Open Bio 2018; 8:1395-1404. [PMID: 30186741 PMCID: PMC6120250 DOI: 10.1002/2211-5463.12455] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/30/2018] [Accepted: 05/15/2018] [Indexed: 01/22/2023] Open
Abstract
Elastin is a long-lived extracellular matrix protein responsible for the structural integrity and function of tissues. Breast cancer elastosis is a complex phenomenon resulting in both the deposition of elastotic masses and the local production of elastin fragments. In invasive human breast cancers, an increase in elastosis is correlated with severity of the disease and age of the patient. Elastin-derived peptides (EDPs) are a hallmark of aging and are matrikines - matrix fragments having the ability to regulate cell physiology. They are known to promote processes linked to tumor progression, but their effects on breast cancer cells remain unexplored. Our data show that EDPs enhance the invasiveness of MDA-MB-231 breast cancer cells through the engagement of matrix metalloproteases 14 and 2. We therefore suggest that elastosis and/or an aged stroma could promote breast cancer cell invasiveness.
Collapse
Affiliation(s)
- Stéphanie Salesse
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Ludivine Odoul
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Lise Chazée
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Christian Garbar
- Biopathology Department Institut Jean Godinot-Unicancer Reims France.,DERM-I-C EA7319 Université de Reims Champagne Ardenne France
| | - Laurent Duca
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Laurent Martiny
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Rachid Mahmoudi
- Faculty of Medicine, EA3797 University of Reims Champagne-Ardenne France.,Department of Geriatrics and Internal Medicine Maison Blanche Hospital Reims University Hospitals France
| | - Laurent Debelle
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| |
Collapse
|
26
|
Soler A, Hunter I, Joseph G, Hutcheson R, Hutcheson B, Yang J, Zhang FF, Joshi SR, Bradford C, Gotlinger KH, Maniyar R, Falck JR, Proctor S, Schwartzman ML, Gupte SA, Rocic P. Elevated 20-HETE in metabolic syndrome regulates arterial stiffness and systolic hypertension via MMP12 activation. J Mol Cell Cardiol 2018; 117:88-99. [PMID: 29428638 PMCID: PMC5877315 DOI: 10.1016/j.yjmcc.2018.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/08/2018] [Accepted: 02/07/2018] [Indexed: 11/24/2022]
Abstract
Arterial stiffness plays a causal role in development of systolic hypertension. 20-hydroxyeicosatetraeonic acid (20-HETE), a cytochrome P450 (CYP450)-derived arachidonic acid metabolite, is known to be elevated in resistance arteries in hypertensive animal models and loosely associated with obesity in humans. However, the role of 20-HETE in the regulation of large artery remodeling in metabolic syndrome has not been investigated. We hypothesized that elevated 20-HETE in metabolic syndrome increases matrix metalloproteinase 12 (MMP12) activation leading to increased degradation of elastin, increased large artery stiffness and increased systolic blood pressure. 20-HETE production was increased ~7 fold in large, conduit arteries of metabolic syndrome (JCR:LA-cp, JCR) vs. normal Sprague-Dawley (SD) rats. This correlated with increased elastin degradation (~7 fold) and decreased arterial compliance (~75% JCR vs. SD). 20-HETE antagonists blocked elastin degradation in JCR rats concomitant with blocking MMP12 activation. 20-HETE antagonists normalized, and MMP12 inhibition (pharmacological and MMP12-shRNA-Lnv) significantly improved (~50% vs. untreated JCR) large artery compliance in JCR rats. 20-HETE antagonists also decreased systolic (182 ± 3 mmHg JCR, 145 ± 3 mmHg JCR + 20-HETE antagonists) but not diastolic blood pressure in JCR rats. Whereas diastolic pressure was fully angiotensin II (Ang II)-dependent, systolic pressure was only partially Ang II-dependent, and large artery stiffness was Ang II-independent. Thus, 20-HETE-dependent regulation of systolic blood pressure may be a unique feature of metabolic syndrome related to high 20-HETE production in large, conduit arteries, which results in increased large artery stiffness and systolic blood pressure. These findings may have implications for management of systolic hypertension in patients with metabolic syndrome.
Collapse
Affiliation(s)
- Amanda Soler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Ian Hunter
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Gregory Joseph
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Rebecca Hutcheson
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Brenda Hutcheson
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Jenny Yang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Frank Fan Zhang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Sachindra Raj Joshi
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Chastity Bradford
- Department of Biology, Tuskegee University, Tuskegee, AL 36088, United States
| | - Katherine H Gotlinger
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Rachana Maniyar
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - John R Falck
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Spencer Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Alberta Institute for Human Nutrition, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | | | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Petra Rocic
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States.
| |
Collapse
|
27
|
Sena BF, Figueiredo JL, Aikawa E. Cathepsin S As an Inhibitor of Cardiovascular Inflammation and Calcification in Chronic Kidney Disease. Front Cardiovasc Med 2018; 4:88. [PMID: 29379789 PMCID: PMC5770806 DOI: 10.3389/fcvm.2017.00088] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/14/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease (CVD) is responsible for the majority of deaths in the developed world. Particularly, in patients with chronic kidney disease (CKD), the imbalance of calcium and phosphate may lead to the acceleration of both vascular and valve inflammation and calcification. One in two patients with CKD are reported as dying from cardiovascular causes due to the resulting acceleration in the development of atherosclerosis plaques. In addition, CKD patients on hemodialysis are prone to aortic valve calcification and often need valve replacement before kidney transplantation. The lysosomal proteases, cathepsins, are composed of 11 cysteine members (cathepsin B, C, F, H, K, L, O, S, V, W, and Z), as well as serine proteases cathepsin A and G, which cleave peptide bonds with serine as the amino acid, and aspartyl proteases D and E, which use an activated water molecule bound to aspartate to break peptide substrate. Cysteine proteases, also known as thiol proteases, degrade protein via the deprotonation of a thiol and have been found to play a significant role in autoimmune disease, atherosclerosis, aortic valve calcification, cardiac repair, and cardiomyopathy, operating within extracellular spaces. This review sought to evaluate recent findings in this field, highlighting how among cathepsins, the inhibition of cathepsin S in particular, could play a significant role in diminishing the effects of CVD, especially for patients with CKD.
Collapse
Affiliation(s)
- Brena F Sena
- Boston University School of Public Health, Boston, MA, United States
| | - Jose Luiz Figueiredo
- Department of Surgery, Introduction to Clinical and Surgical Techniques Division, Laboratory of Experimental Surgery, Federal University of Pernambuco, Recife, Brazil
| | - Elena Aikawa
- The Center of Excellence in Vascular Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Does elastin deficiency cause chronic kidney disease? Kidney Int 2017; 92:1036-1038. [DOI: 10.1016/j.kint.2017.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 11/18/2022]
|
29
|
Peeters SA, Engelen L, Buijs J, Chaturvedi N, Fuller JH, Jorsal A, Parving HH, Tarnow L, Theilade S, Rossing P, Schalkwijk CG, Stehouwer CDA. Circulating matrix metalloproteinases are associated with arterial stiffness in patients with type 1 diabetes: pooled analysis of three cohort studies. Cardiovasc Diabetol 2017; 16:139. [PMID: 29070037 PMCID: PMC5657128 DOI: 10.1186/s12933-017-0620-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/15/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Altered regulation of extracellular matrix (ECM) composition by matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinase (TIMPs) may contribute to arterial stiffening. We investigated associations between circulating MMP-1, -2, -3, -9, -10 and TIMP-1, and carotid-femoral pulse wave velocity (cfPWV) and pulse pressure (PP), as markers of arterial stiffness in type 1 diabetic patients. METHODS Individuals with type 1 diabetes from three different cohorts were included in this study: EURODIAB Prospective Complications study (n = 509), LEACE (n = 370) and PROFIL (n = 638). Linear regression analyses were used to investigate cross-sectional associations between circulating levels of MMP-1, -2, -3, -9, -10, and TIMP-1 and cfPWV (n = 614) as well as office PP (n = 1517). Data on 24-h brachial and 24-h central PP were available in 638 individuals from PROFIL. Analyses were adjusted for age, sex, duration of diabetes, HbA1c, mean arterial pressure (MAP), and eGFR, and additionally for other cardiovascular risk factors and presence of vascular complications. RESULTS After adjustment for potential confounders and presence of vascular complications, circulating MMP-3 was associated with cfPWV [β per 1 SD higher lnMMP3 0.29 m/s (0.02; 0.55)]. In addition, brachial and central 24-h PP measurements in PROFIL were significantly associated with MMP-2 [(1.40 (0.47:2.33) and 1.43 (0.63:2.23)]. Pooled data analysis showed significant associations of circulating levels of MMP-1 and MMP-2 with office PP [β per 1 SD higher lnMMP-1 and lnMMP-2 = - 0.83 mmHg (95% CI - 1.50; - 0.16) and = 1.33 mmHg (0.55; 2.10), respectively]. CONCLUSIONS MMPs-1, -2, and -3 are independently associated with markers of arterial stiffening in patients with type 1 diabetes and may become therapeutic targets.
Collapse
Affiliation(s)
- Stijn A. Peeters
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- Department of Internal Medicine, Zuyderland hospital, Heerlen, The Netherlands
| | - Lian Engelen
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- Centraal Bureau voor de Statistiek, Heerlen, The Netherlands
| | - Jacqueline Buijs
- Department of Internal Medicine, Zuyderland hospital, Heerlen, The Netherlands
| | - Nish Chaturvedi
- Institute of Cardiovascular Sciences, University College London, London, UK
| | - John H. Fuller
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Anders Jorsal
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Hans-Henrik Parving
- Department of Medical Endocrinology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lise Tarnow
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Faculty of Health, Aarhus University, Aarhus, Denmark
| | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - Casper G. Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Coen D. A. Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
30
|
Janssen R. Magnesium to counteract elastin degradation and vascular calcification in chronic obstructive pulmonary disease. Med Hypotheses 2017; 107:74-77. [PMID: 28915968 DOI: 10.1016/j.mehy.2017.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/08/2017] [Accepted: 08/11/2017] [Indexed: 01/26/2023]
Abstract
Accelerated elastin degradation is an important pathogenic mechanism in chronic obstructive pulmonary disease (COPD) leading to irreversible lung function loss and cardiovascular comorbidities. The rate of elastin breakdown is a predictor of mortality in patients with COPD. Decelerating elastinolysis might be an attractive therapeutic target in this debilitating condition. Vascular calcification starts in the elastin network of the arterial wall and is enhanced in patients with COPD. Elastin calcification is accompanied by an upregulation of matrix metalloproteinase gene expression and consequently a shift in the elastase/anti-elastase balance towards degradation. Magnesium can be regarded as a natural calcium antagonist and has the proven ability to ameliorate vascular calcification. Furthermore, an animal study has suggested that magnesium deficiency promotes elastin degradation. I hypothesize that inhibiting elastin calcification by means of magnesium supplementation might counteract both vascular calcification and elastin degradation in COPD. This could potentially have a favorable impact on cardiovascular and respiratory related morbidity/mortality in patients with COPD.
Collapse
Affiliation(s)
- Rob Janssen
- Department of Pulmonary Medicine, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands.
| |
Collapse
|
31
|
Hunter I, Soler A, Joseph G, Hutcheson B, Bradford C, Zhang FF, Potter B, Proctor S, Rocic P. Cardiovascular function in male and female JCR:LA-cp rats: effect of high-fat/high-sucrose diet. Am J Physiol Heart Circ Physiol 2017; 312:H742-H751. [PMID: 28087518 DOI: 10.1152/ajpheart.00535.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 12/31/2022]
Abstract
Thirty percent of the world population is diagnosed with metabolic syndrome. High-fat/high-sucrose (HF/HS) diet (Western diet) correlates with metabolic syndrome prevalence. We characterized effects of the HF/HS diet on vascular (arterial stiffness, vasoreactivity, and coronary collateral development) and cardiac (echocardiography) function, oxidative stress, and inflammation in a rat model of metabolic syndrome (JCR rats). Furthermore, we determined whether male versus female animals were affected differentially by the Western diet. Cardiovascular function in JCR male rats was impaired versus normal Sprague-Dawley (SD) rats. HF/HS diet compromised cardiovascular (dys)function in JCR but not SD male rats. In contrast, cardiovascular function was minimally impaired in JCR female rats on normal chow. However, cardiovascular function in JCR female rats on the HF/HS diet deteriorated to levels comparable to JCR male rats on the HF/HS diet. Similarly, oxidative stress was markedly increased in male but not female JCR rats on normal chow but was equally exacerbated by the HF/HS diet in male and female JCR rats. These results indicate that the Western diet enhances oxidative stress and cardiovascular dysfunction in metabolic syndrome and eliminates the protective effect of female sex on cardiovascular function, implying that both males and females with metabolic syndrome are at equal risk for cardiovascular disease.NEW & NOTEWORTHY Western diet abolished protective effect of sex against cardiovascular disease (CVD) development in premenopausal animals with metabolic syndrome. Western diet accelerates progression of CVD in male and female animals with preexisting metabolic syndrome but not normal animals. Exacerbation of baseline oxidative stress correlates with accelerated progression of CVD in metabolic syndrome animals on Western diet.
Collapse
Affiliation(s)
- Ian Hunter
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Amanda Soler
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Gregory Joseph
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Brenda Hutcheson
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | | | - Frank Fan Zhang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Barry Potter
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| | - Spencer Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Alberta Institute for Human Nutrition, University of Alberta, Edmonton, Alberta, Canada
| | - Petra Rocic
- Department of Pharmacology, New York Medical College, Valhalla, New York;
| |
Collapse
|
32
|
Lau WL, Huisa BN, Fisher M. The Cerebrovascular-Chronic Kidney Disease Connection: Perspectives and Mechanisms. Transl Stroke Res 2016; 8:67-76. [PMID: 27628245 PMCID: PMC5241336 DOI: 10.1007/s12975-016-0499-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/21/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is an independent risk factor for the development of cerebrovascular disease, particularly small vessel disease which can manifest in a variety of phenotypes ranging from lacunes to microbleeds. Small vessel disease likely contributes to cognitive dysfunction in the CKD population. Non-traditional risk factors for vascular injury in uremia include loss of calcification inhibitors, hyperphosphatemia, increased blood pressure variability, elastinolysis, platelet dysfunction, and chronic inflammation. In this review, we discuss the putative pathways by which these mechanisms may promote cerebrovascular disease and thus increase risk of future stroke in CKD patients.
Collapse
Affiliation(s)
- Wei Ling Lau
- Department of Medicine, Division of Nephrology, University of California, Irvine, CA, USA
| | - Branko N Huisa
- Department of Neurology, University of California, San Diego, CA, USA
| | - Mark Fisher
- Departments of Neurology, Anatomy & Neurobiology, and Pathology & Laboratory Medicine, University of California, Irvine, CA, USA. .,Department of Neurology, UC Irvine Medical Center, 101 The City Drive South, Shanbrom Hall, Room 121, Orange, CA, 92868, USA.
| |
Collapse
|
33
|
Rabinovich RA, Miller BE, Wrobel K, Ranjit K, Williams MC, Drost E, Edwards LD, Lomas DA, Rennard SI, Agustí A, Tal-Singer R, Vestbo J, Wouters EFM, John M, van Beek EJR, Murchison JT, Bolton CE, MacNee W, Huang JTJ. Circulating desmosine levels do not predict emphysema progression but are associated with cardiovascular risk and mortality in COPD. Eur Respir J 2016; 47:1365-73. [PMID: 27009168 DOI: 10.1183/13993003.01824-2015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/16/2016] [Indexed: 11/05/2022]
Abstract
Elastin degradation is a key feature of emphysema and may have a role in the pathogenesis of atherosclerosis associated with chronic obstructive pulmonary disease (COPD). Circulating desmosine is a specific biomarker of elastin degradation. We investigated the association between plasma desmosine (pDES) and emphysema severity/progression, coronary artery calcium score (CACS) and mortality.pDES was measured in 1177 COPD patients and 110 healthy control subjects from two independent cohorts. Emphysema was assessed on chest computed tomography scans. Aortic arterial stiffness was measured as the aortic-femoral pulse wave velocity.pDES was elevated in patients with cardiovascular disease (p<0.005) and correlated with age (rho=0.39, p<0.0005), CACS (rho=0.19, p<0.0005) modified Medical Research Council dyspnoea score (rho=0.15, p<0.0005), 6-min walking distance (rho=-0.17, p<0.0005) and body mass index, airflow obstruction, dyspnoea, exercise capacity index (rho=0.10, p<0.01), but not with emphysema, emphysema progression or forced expiratory volume in 1 s decline. pDES predicted all-cause mortality independently of several confounding factors (p<0.005). In an independent cohort of 186 patients with COPD and 110 control subjects, pDES levels were higher in COPD patients with cardiovascular disease and correlated with arterial stiffness (p<0.05).In COPD, excess elastin degradation relates to cardiovascular comorbidities, atherosclerosis, arterial stiffness, systemic inflammation and mortality, but not to emphysema or emphysema progression. pDES is a good biomarker of cardiovascular risk and mortality in COPD.
Collapse
Affiliation(s)
- Roberto A Rabinovich
- Edinburgh Lung and the Environment Group Initiative (ELEGI), Centre for Inflammation and Research, Queens' Medical Research Institute, Edinburgh, UK
| | - Bruce E Miller
- Respiratory Therapy Area Unit, GSK, King of Prussia, PA, USA
| | - Karolina Wrobel
- Medical Research Institute, School of Medicine, University of Dundee, Dundee, UK
| | - Kareshma Ranjit
- Edinburgh Lung and the Environment Group Initiative (ELEGI), Centre for Inflammation and Research, Queens' Medical Research Institute, Edinburgh, UK
| | | | - Ellen Drost
- Edinburgh Lung and the Environment Group Initiative (ELEGI), Centre for Inflammation and Research, Queens' Medical Research Institute, Edinburgh, UK
| | | | - David A Lomas
- Faculty of Medical Sciences, University College London, London, UK
| | - Stephen I Rennard
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska, Omaha, NE, USA Clinical Discovery Unit, AstraZeneca, Cambridge, UK
| | - Alvar Agustí
- Servei de Pneumologia, Thorax Institute, Hospital Clinic, IDIBAPS, Universitat de Barcelona and CIBER Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Ruth Tal-Singer
- Respiratory Therapy Area Unit, GSK, King of Prussia, PA, USA
| | - Jørgen Vestbo
- Centre for Respiratory Medicine and Allergy, Manchester Academic Health Science Centre, University Hospital South Manchester NHS Foundation Trust, Manchester, UK
| | - Emiel F M Wouters
- Dept of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Michelle John
- Nottingham Respiratory Research Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Edwin J R van Beek
- Clinical Research Imaging Centre, Queens Medical Research Institute, Edinburgh, UK
| | | | - Charlotte E Bolton
- Nottingham Respiratory Research Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - William MacNee
- Edinburgh Lung and the Environment Group Initiative (ELEGI), Centre for Inflammation and Research, Queens' Medical Research Institute, Edinburgh, UK
| | - Jeffrey T J Huang
- Medical Research Institute, School of Medicine, University of Dundee, Dundee, UK
| | | |
Collapse
|
34
|
Duca L, Blaise S, Romier B, Laffargue M, Gayral S, El Btaouri H, Kawecki C, Guillot A, Martiny L, Debelle L, Maurice P. Matrix ageing and vascular impacts: focus on elastin fragmentation. Cardiovasc Res 2016; 110:298-308. [DOI: 10.1093/cvr/cvw061] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/07/2016] [Indexed: 12/17/2022] Open
|
35
|
Hsiao KC, Tsai JP, Yang SF, Lee WC, Huang JY, Chang SC, Hso CS, Chang HR. MMP-2 serum concentrations predict mortality in hemodialysis patients: a 5-year cohort study. Clin Chim Acta 2015; 452:161-6. [PMID: 26612771 DOI: 10.1016/j.cca.2015.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND We evaluated the ability of matrix metalloproteinase (MMP)-2, MMP-9, myeloperoxidase, osteopontin and stromal cell-derived factor 1 to predict mortality in hemodialysis (HD) patients. METHODS One hundred forty HD patients were enrolled and followed from December 2007 until December 2012. At the end of this 5-year period, data were compared between the patients who were alive and those who had died. RESULTS The patients who alive were younger (56 vs. 63y), with lower frequency of diabetes mellitus (34.34% vs. 58.53%), higher concentrations of albumin (4.13 vs. 3.91mg/dl) and lower concentrations of MMP-2 (430.76 vs. 521.59ng/ml). Multivariate analysis showed that age (HR=1.03, p=0.02), diabetes mellitus (HR=2.395, p=0.012), albumin (HR=0.475, p=0.047) and MMP-2 (HR=1.003, p=0.005) were independent factors predicting mortality in HD patients. Receiver operating characteristic curve analysis showed that albumin (AUC=0.628, p=0.027) and MMP-2 (AUC=0.643, p=0.004) had a similar ability (p=0.76) to predict survival of HD patients. CONCLUSIONS Compared with albumin, serum MMP-2 is a non-inferior prognostic marker for predicting the survival of HD patients.
Collapse
Affiliation(s)
- Kuang-Chih Hsiao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Nephrology, Department of Medicine, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Jen-Pi Tsai
- Department of Nephrology, Buddhist Dalin Tzu Chi General Hospital, Chiayi, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Chin Lee
- Division of Nephrology, Department of Medicine, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Jong-Yu Huang
- Division of Nephrology, Department of Medicine, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Shun-Chi Chang
- Division of Nephrology, Department of Medicine, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Chun-Shuo Hso
- Division of Nephrology, Department of Medicine, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Horng-Rong Chang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Nephrology, Department of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| |
Collapse
|
36
|
Kumar Vr S, Darisipudi MN, Steiger S, Devarapu SK, Tato M, Kukarni OP, Mulay SR, Thomasova D, Popper B, Demleitner J, Zuchtriegel G, Reichel C, Cohen CD, Lindenmeyer MT, Liapis H, Moll S, Reid E, Stitt AW, Schott B, Gruner S, Haap W, Ebeling M, Hartmann G, Anders HJ. Cathepsin S Cleavage of Protease-Activated Receptor-2 on Endothelial Cells Promotes Microvascular Diabetes Complications. J Am Soc Nephrol 2015; 27:1635-49. [PMID: 26567242 DOI: 10.1681/asn.2015020208] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 08/25/2015] [Indexed: 01/13/2023] Open
Abstract
Endothelial dysfunction is a central pathomechanism in diabetes-associated complications. We hypothesized a pathogenic role in this dysfunction of cathepsin S (Cat-S), a cysteine protease that degrades elastic fibers and activates the protease-activated receptor-2 (PAR2) on endothelial cells. We found that injection of mice with recombinant Cat-S induced albuminuria and glomerular endothelial cell injury in a PAR2-dependent manner. In vivo microscopy confirmed a role for intrinsic Cat-S/PAR2 in ischemia-induced microvascular permeability. In vitro transcriptome analysis and experiments using siRNA or specific Cat-S and PAR2 antagonists revealed that Cat-S specifically impaired the integrity and barrier function of glomerular endothelial cells selectively through PAR2. In human and mouse type 2 diabetic nephropathy, only CD68(+) intrarenal monocytes expressed Cat-S mRNA, whereas Cat-S protein was present along endothelial cells and inside proximal tubular epithelial cells also. In contrast, the cysteine protease inhibitor cystatin C was expressed only in tubules. Delayed treatment of type 2 diabetic db/db mice with Cat-S or PAR2 inhibitors attenuated albuminuria and glomerulosclerosis (indicators of diabetic nephropathy) and attenuated albumin leakage into the retina and other structural markers of diabetic retinopathy. These data identify Cat-S as a monocyte/macrophage-derived circulating PAR2 agonist and mediator of endothelial dysfunction-related microvascular diabetes complications. Thus, Cat-S or PAR2 inhibition might be a novel strategy to prevent microvascular disease in diabetes and other diseases.
Collapse
Affiliation(s)
- Santhosh Kumar Vr
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Murthy N Darisipudi
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Stefanie Steiger
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Satish Kumar Devarapu
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Maia Tato
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Onkar P Kukarni
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Shrikant R Mulay
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Dana Thomasova
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Bastian Popper
- Department of Anatomy and Cell Biology, Ludwig-Maximilians Universität, Munich, Germany
| | | | - Gabriele Zuchtriegel
- Walter Brendel Centre of Experimental Medicine, and Department of Otorhinolaryngology, Head and Neck Surgery, University of Munich, Munich, Germany
| | - Christoph Reichel
- Walter Brendel Centre of Experimental Medicine, and Department of Otorhinolaryngology, Head and Neck Surgery, University of Munich, Munich, Germany
| | - Clemens D Cohen
- Division of Nephrology, Krankenhaus Harlaching, Munich, Germany; Division of Nephrology and Institute of Physiology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | - Helen Liapis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Solange Moll
- Institute of Clinical Pathology, University Hospital Geneva, Geneva, Switzerland
| | - Emma Reid
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Ireland; and
| | - Alan W Stitt
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Ireland; and
| | - Brigitte Schott
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Sabine Gruner
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Wolfgang Haap
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Martin Ebeling
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Guido Hartmann
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Hans-Joachim Anders
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany;
| |
Collapse
|
37
|
Angiotensin-II induced hypertension and renovascular remodelling in tissue inhibitor of metalloproteinase 2 knockout mice. J Hypertens 2015; 31:2270-81; discussion 2281. [PMID: 24077247 DOI: 10.1097/hjh.0b013e3283649b33] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sustained hypertension induces renovascular remodelling by altering extracellular matrix (ECM) components. Matrix metalloproteinases (MMPs) are Zn-dependent enzymes that regulate ECM turnover in concert with their inhibitors, tissue inhibitors of metalloproteinases (TIMPs). Increased MMP-2 and MMP-9 have been implicated in hypertensive complications; however, the contribution of individual MMPs/TIMPs in renal remodelling has not been fully elucidated. The purpose of this study was to determine the effect of TIMP2 deficiency and thus MMP-2 on angiotensin-II (Ang-II) induced renal remodelling. METHOD C57BL/6J (wild-type) and TIMP2 knockout mice were infused with Ang-II at 250 ng/kg per min for 4 weeks. Blood pressure was measured weekly and end-point laser Doppler flowmetry was done to assess cortical blood flow. Immunohistochemical staining was performed for collagen and elastin analyses. The activity of MMP-9 and MMP-2 was determined by Gelatin zymography. RESULTS Ang-II induced similar elevation in mean blood pressure in TIMP2 and wild-type mice. In TIMP2 mice, Ang-II treatment was associated with a greater reduction in renal cortical blood flow and barium angiography demonstrated decreased vascular density compared with Ang-II treated wild-type mice. Peri-glomerular and vascular collagen deposition was increased and elastin content was decreased causing increased wall-to-lumen ratio in TIMP2 mice compared with wild-type mice receiving Ang-II. Ang-II increased the expression and activity of MMP-9 predominantly in TIMP2 mice than in wild-type mice. CONCLUSION These results suggest that TIMP2 deficiency exacerbates renovascular remodelling in agonist-induced hypertension by a mechanism that may, in part, be attributed to increased activity of MMP-9.
Collapse
|
38
|
Camici GG, Savarese G, Akhmedov A, Lüscher TF. Molecular mechanism of endothelial and vascular aging: implications for cardiovascular disease. Eur Heart J 2015; 36:3392-403. [PMID: 26543043 DOI: 10.1093/eurheartj/ehv587] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023] Open
Abstract
Western societies are aging due to an increasing life span, decreased birth rates, and improving social and health conditions. On the other hand, the prevalence of cardiovascular (CV) and cerebrovascular (CBV) diseases rises with age. Thus, in view of the ongoing aging pandemic, it is appropriate to better understand the molecular pathways of aging as well as age-associated CV and CBV diseases. Oxidative stress contributes to aging of organs and the whole body by an accumulation of reactive oxygen species promoting oxidative damage. Indeed, increased oxidative stress produced in the mitochondria and cytosol of heart and brain is a common denominator to almost all CV and CBV diseases. The mitochondrial adaptor protein p66(Shc) and the family of deacetylase enzymes, the sirtuins, regulate the aging process, determine lifespan of many species and are involved in CV diseases. GDF11, a member of TGFβ superfamily with homology to myostatin also retards the aging process via yet unknown mechanisms. Recent evidence points towards a promising role of this novel 'rejuvenation' factor in reducing age-related heart disease. Finally, telomere length is also involved in aging and the development of age-related CV dysfunction. This review focuses on the latest scientific advances in understanding age-related changes of the CV and CBV system, as well as delineating potential novel therapeutic targets derived from aging research for CV and CBV diseases.
Collapse
Affiliation(s)
- Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Gianluigi Savarese
- Cardiology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland University Heart Center, Cardiology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Lee SH, Shin K, Park S, Kang SM, Choi D, Lee SH, Lee SH. Circulating Anti-Elastin Antibody Levels and Arterial Disease Characteristics: Associations with Arterial Stiffness and Atherosclerosis. Yonsei Med J 2015; 56:1545-51. [PMID: 26446635 PMCID: PMC4630041 DOI: 10.3349/ymj.2015.56.6.1545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 12/26/2014] [Accepted: 12/27/2014] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Elastin is a major arterial structural protein, and elastin-derived peptides are related to arterial change. We previously reported on a novel assay developed using aortic elastin peptides; however, its clinical implications remain unclear. In this study, we assessed whether anti-elastin antibody titers reflect the risk of coronary artery disease (CAD) or its characteristics. MATERIALS AND METHODS We included 174 CAD patients and 171 age- and sex-matched controls. Anti-elastin antibody titers were quantified by enzyme-linked immunosorbent assay. Parameters of arterial stiffness, including the augmentation index (AI) and heart-to-femoral pulse wave velocity (hfPWV), were measured non-invasively. The clinical and angiographic characteristics of CAD patients were also evaluated. Associations between anti-elastin levels and vascular characteristics were examined by linear regression analysis. RESULTS The median blood level of anti-elastin was significantly lower in the CAD group than in the controls [197 arbitrary unit (a.u.) vs. 63 a.u., p<0.001]. Levels of anti-elastin were significantly lower in men and in subjects with hypertension, diabetes mellitus, hyperlipidemia, or high hfPWV. Nevertheless, anti-elastin levels were not dependent on atherothrombotic events or the angiographic severity of CAD. In a multivariate analysis, male sex (β=-0.38, p<0.001), diabetes mellitus (β=-0.62, p<0.001), hyperlipidemia (β=-0.29, p<0.001), and AI (β=-0.006, p=0.02) were ultimately identified as determinants of anti-elastin levels. CONCLUSION Lower levels of anti-elastin are related to CAD. The association between antibody titers and CAD is linked to arterial stiffness rather than the advancement of atherosclerosis.
Collapse
Affiliation(s)
- Seung-Hyun Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kihyuk Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea; Department of Dermatology, School of Medicine, Pusan National University, Busan, Korea
| | - Sungha Park
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University Health System, Seoul, Korea
| | - Seok-Min Kang
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University Health System, Seoul, Korea
| | - Donghoon Choi
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University Health System, Seoul, Korea
| | - Seung-Hyo Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University Health System, Seoul, Korea.
| |
Collapse
|
40
|
Payne CD, Deeg MA, Chan M, Tan LH, LaBell ES, Shen T, DeBrota DJ. Pharmacokinetics and pharmacodynamics of the cathepsin S inhibitor, LY3000328, in healthy subjects. Br J Clin Pharmacol 2015; 78:1334-42. [PMID: 25039273 DOI: 10.1111/bcp.12470] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 07/11/2014] [Indexed: 11/27/2022] Open
Abstract
AIM The aim of this study was to assess the safety and tolerability, pharmacokinetics and pharmacodynamics of LY3000328 when administered as single escalating doses to healthy volunteers. METHODS This was a phase 1, placebo-controlled, dose escalation study with LY3000328 in 21 healthy male volunteers. Subjects were administered escalating LY3000328 doses up to 300 mg with food in this single dose study. Blood samples were collected at set times post-dose for the assessment of LY3000328 pharmacokinetics and the measurement of cathepsin S (CatS) activity, CatS mass and calculated CatS specific activity. RESULTS All doses of LY3000328 were well tolerated, with linear pharmacokinetics up to the 300 mg dose. The pharmacodynamic activity of LY3000328 was measured ex vivo showing a biphasic response to LY3000328, where CatS activity declines, then returns to baseline, and then increases to a level above baseline. CatS mass was also assessed post-dose which increased in a dose-dependent manner, and continued to increase after LY3000328 had been cleared from the body. CatS specific activity was additionally calculated to normalize CatS activity for changes in CatS mass. This demonstrated the increase in CatS activity was attributable to the increase in CatS mass detected in plasma. CONCLUSION A specific inhibitor of CatS which is cleared quickly from plasma may produce a transient decrease in plasma CatS activity which is followed by a more prolonged increase in plasma CatS mass which may have implications for the future clinical development of inhibitors of CatS.
Collapse
Affiliation(s)
- Christopher D Payne
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Affiliation(s)
- Zhongjie Sun
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City.
| |
Collapse
|
42
|
Kawecki C, Hézard N, Bocquet O, Poitevin G, Rabenoelina F, Kauskot A, Duca L, Blaise S, Romier B, Martiny L, Nguyen P, Debelle L, Maurice P. Elastin-derived peptides are new regulators of thrombosis. Arterioscler Thromb Vasc Biol 2014; 34:2570-8. [PMID: 25341794 DOI: 10.1161/atvbaha.114.304432] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Elastin is the major structural extracellular matrix component of the arterial wall that provides the elastic recoil properties and resilience essential for proper vascular function. Elastin-derived peptides (EDP) originating from elastin fragmentation during vascular remodeling have been shown to play an important role in cell physiology and development of cardiovascular diseases. However, their involvement in thrombosis has been unexplored to date. In this study, we investigated the effects of EDP on (1) platelet aggregation and related signaling and (2) thrombus formation. We also characterized the mechanism by which EDP regulate thrombosis. APPROACH AND RESULTS We show that EDP, derived from organo-alkaline hydrolysate of bovine insoluble elastin (kappa-elastin), decrease human platelet aggregation in whole blood induced by weak and strong agonists, such as ADP, epinephrine, arachidonic acid, collagen, TRAP, and U46619. In a mouse whole blood perfusion assay over a collagen matrix, kappa-elastin and VGVAPG, the canonical peptide recognizing the elastin receptor complex, significantly decrease thrombus formation under arterial shear conditions. We confirmed these results in vivo by demonstrating that both kappa-elastin and VGVAPG significantly prolonged the time for complete arteriole occlusion in a mouse model of thrombosis and increased tail bleeding times. Finally, we demonstrate that the regulatory role of EDP on thrombosis relies on platelets that express a functional elastin receptor complex and on the ability of EDP to disrupt plasma von Willebrand factor interaction with collagen. CONCLUSIONS These results highlight the complex nature of the mechanisms governing thrombus formation and reveal an unsuspected regulatory role for circulating EDP in thrombosis.
Collapse
Affiliation(s)
- Charlotte Kawecki
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Nathalie Hézard
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Olivier Bocquet
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Gaël Poitevin
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Fanja Rabenoelina
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Alexandre Kauskot
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Laurent Duca
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Sébastien Blaise
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Béatrice Romier
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Laurent Martiny
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Philippe Nguyen
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Laurent Debelle
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.)
| | - Pascal Maurice
- From the URCA, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), laboratoire SiRMa, UFR Sciences Exactes et Naturelles, Reims, France (C.K., O.B., F.R., L. Ducca, S.B., B.R., L.M., L. Debelle, P.M.); EA3801, Hémostase et remodelage vasculaire post-ischémique (HERVI), UFR de Médecine, Reims, France (N.H., G.P., P.N.); CHU Reims, Hôpital Robert Debré, Laboratoire d'Hématologie, Reims, France (N.H., P.N.); INSERM UMRS 1140, Université Paris Descartes, Sorbonne Paris Cité, France (A.K.); and INSERM U770, Le Kremlin Bicêtre, Université Paris-Sud, Le Kremlin Bicêtre, France (A.K.).
| |
Collapse
|
43
|
Ameer OZ, Salman IM, Avolio AP, Phillips JK, Butlin M. Opposing changes in thoracic and abdominal aortic biomechanical properties in rodent models of vascular calcification and hypertension. Am J Physiol Heart Circ Physiol 2014; 307:H143-51. [DOI: 10.1152/ajpheart.00139.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated the effects of hypertension on regional aortic biomechanical and structural properties in three rat models of vascular calcification: the hypertensive Lewis polycystic kidney (LPK; n = 13) model of chronic kidney disease, spontaneously hypertensive rats (SHRs; n = 12), and calcification in normotensive Lewis rats induced by vitamin D3 and nicotine (VDN; n = 8). Lewis and Wistar-Kyoto rats were controls. Thoracic and abdominal aortic stiffness parameters were assessed by tensile testing. In models where aortic stiffness differences compared with controls existed in both thoracic and abdominal segments, an additional cohort was quantified by histology for thoracic and abdominal aortic elastin, collagen, and calcification. LPK and VDN animals had higher thoracic breaking strain than control animals ( P < 0.01 and P < 0.05, respectively) and lower energy absorption within the tensile curve of the abdominal aorta ( P < 0.05). SHRs had a lower abdominal breaking stress than Wistar-Kyoto rats. LPK and VDN rats had more elastic lamellae fractures than control rats ( P < 0.001), which were associated with calcium deposition (thoracic R = 0.37, P = 0.048; abdominal: R = 0.40, P = 0.046). LPK rats had higher nuclear density than control rats ( P < 0.01), which was also evident in the thoracic but not abdominal aorta of VDN rats ( P < 0.01). In LPK and VDN rats, but not in control rats, media thickness and cross-sectional area were at least 1.5-fold greater in thoracic than abdominal regions. The calcification models chronic kidney disease and induced calcification in normotension caused differences in regional aortic stiffness not seen in a genetic form of hypertension. Detrimental abdominal aortic remodeling but lower stiffness in the thoracic aorta with disease indicates possible compensatory mechanisms in the proximal aorta.
Collapse
Affiliation(s)
- Omar Z. Ameer
- The Australian School of Advanced Medicine, Macquarie University, Sydney, New South Wales, Australia
| | - Ibrahim M. Salman
- The Australian School of Advanced Medicine, Macquarie University, Sydney, New South Wales, Australia
| | - Alberto P. Avolio
- The Australian School of Advanced Medicine, Macquarie University, Sydney, New South Wales, Australia
| | - Jacqueline K. Phillips
- The Australian School of Advanced Medicine, Macquarie University, Sydney, New South Wales, Australia
| | - Mark Butlin
- The Australian School of Advanced Medicine, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
44
|
Li X, Li Y, Jin J, Jin D, Cui L, Li X, Rei Y, Jiang H, Zhao G, Yang G, Zhu E, Nan Y, Cheng X. Increased serum cathepsin K in patients with coronary artery disease. Yonsei Med J 2014; 55:912-9. [PMID: 24954318 PMCID: PMC4075394 DOI: 10.3349/ymj.2014.55.4.912] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Cathepsin K is a potent collagenase implicated in human and animal atherosclerosis-based vascular remodeling. This study examined the hypothesis that serum CatK is associated with the prevalence of coronary artery disease (CAD). MATERIALS AND METHODS Between January 2011 and December 2012, 256 consecutive subjects were enrolled from among patients who underwent coronary angiography and percutaneous coronary intervention treatment. A total of 129 age-matched subjects served as controls. RESULTS The subjects' serum cathepsin K and high sensitive C-reactive protein (hs-CRP) and high-density lipoprotein cholesterol were measured. The patients with CAD had significantly higher serum cathepsin K levels compared to the controls (130.8±25.5 ng/mL vs. 86.9±25.5 ng/mL, p<0.001), and the patients with acute coronary syndrome had significantly higher serum cathepsin K levels compared to those with stable angina pectoris (137.1±26.9 ng/mL vs. 102.6±12.9 ng/mL, p<0.001). A linear regression analysis showed that overall, the cathepsin K levels were inversely correlated with the high-density lipoprotein levels (r=-0.29, p<0.01) and positively with hs-CRP levels (r=0.32, p<0.01). Multiple logistic regression analyses shows that cathepsin K levels were independent predictors of CAD (odds ratio, 1.76; 95% confidence interval, 1.12 to 1.56; p<0.01). CONCLUSION These data indicated that elevated levels of cathepsin K are closely associated with the presence of CAD and that circulating cathepsin K serves a useful biomarker for CAD.
Collapse
Affiliation(s)
- Xiang Li
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Yuzi Li
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Jiyong Jin
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Dehao Jin
- Intervention Laboratory, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Lan Cui
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China.
| | - Xiangshan Li
- Central Laboratory, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Yanna Rei
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China. ; Department of Anesthesiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Haiying Jiang
- Department of Physiology and Pathophysiology, Yanbian University Medical College, Yanji, Jilin P.R., China
| | - Guangxian Zhao
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Guang Yang
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Enbo Zhu
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Yongshan Nan
- Department of Anesthesiology, Yanbian University Hospital, Yanji, Jilin P.R., China
| | - Xianwu Cheng
- Department of Cardiology, Yanbian University Hospital, Yanji, Jilin P.R., China. ; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan. ; Department of Internal Medicine, Kyung Hee University Hospital, Seoul, Korea
| |
Collapse
|
45
|
Filip S, Pontillo C, Peter Schanstra J, Vlahou A, Mischak H, Klein J. Urinary proteomics and molecular determinants of chronic kidney disease: possible link to proteases. Expert Rev Proteomics 2014; 11:535-48. [DOI: 10.1586/14789450.2014.926224] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
46
|
Kidher E, Harling L, Nihoyannopoulos P, Shenker N, Ashrafian H, Francis DP, Mayet J, Athanasiou T. High aortic pulse wave velocity is associated with poor quality of life in surgical aortic valve stenosis patients. Interact Cardiovasc Thorac Surg 2014; 19:189-97. [DOI: 10.1093/icvts/ivu156] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
47
|
|
48
|
Huyard F, Yzydorczyk C, Castro MM, Cloutier A, Bertagnolli M, Sartelet H, Germain N, Comte B, Schulz R, DeBlois D, Nuyt AM. Remodeling of aorta extracellular matrix as a result of transient high oxygen exposure in newborn rats: implication for arterial rigidity and hypertension risk. PLoS One 2014; 9:e92287. [PMID: 24743169 PMCID: PMC3990546 DOI: 10.1371/journal.pone.0092287] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 02/20/2014] [Indexed: 12/22/2022] Open
Abstract
Neonatal high-oxygen exposure leads to elevated blood pressure, microvascular rarefaction, vascular dysfunction and arterial (aorta) rigidity in adult rats. Whether structural changes are present in the matrix of aorta wall is unknown. Considering that elastin synthesis peaks in late fetal life in humans, and early postnatal life in rodents, we postulated that transient neonatal high-oxygen exposure can trigger premature vascular remodelling. Sprague Dawley rat pups were exposed from days 3 to 10 after birth to 80% oxygen (vs. room air control) and were studied at 4 weeks. Blood pressure and vasomotor response of the aorta to angiotensin II and to the acetylcholine analogue carbachol were not different between groups. Vascular superoxide anion production was similar between groups. There was no difference between groups in aortic cross sectional area, smooth muscle cell number or media/lumen ratio. In oxygen-exposed rats, aorta elastin/collagen content ratio was significantly decreased, the expression of elastinolytic cathepsin S was increased whereas collagenolytic cathepsin K was decreased. By immunofluorescence we observed an increase in MMP-2 and TIMP-1 staining in aortas of oxygen-exposed rats whereas TIMP-2 staining was reduced, indicating a shift in the balance towards degradation of the extra-cellular matrix and increased deposition of collagen. There was no significant difference in MMP-2 activity between groups as determined by gelatin zymography. Overall, these findings indicate that transient neonatal high oxygen exposure leads to vascular wall alterations (decreased elastin/collagen ratio and a shift in the balance towards increased deposition of collagen) which are associated with increased rigidity. Importantly, these changes are present prior to the elevation of blood pressure and vascular dysfunction in this model, and may therefore be contributory.
Collapse
Affiliation(s)
- Fanny Huyard
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Catherine Yzydorczyk
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Michele M. Castro
- Departments of Pediatrics & Pharmacology, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Anik Cloutier
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Mariane Bertagnolli
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Hervé Sartelet
- Sainte-Justine University Hospital Research Center, Department of Pathology, Université de Montréal, Montreal, Québec, Canada
| | - Nathalie Germain
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Blandine Comte
- Unit of Human Nutrition UMR 1019, INRA, Research Centre of Clermont-Ferrand/Theix, Saint-Genès-Champanelle, France
| | - Richard Schulz
- Departments of Pediatrics & Pharmacology, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Denis DeBlois
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Anne Monique Nuyt
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
- * E-mail:
| |
Collapse
|
49
|
Smith ER, Ford ML, Tomlinson LA, Bodenham E, McMahon LP, Farese S, Rajkumar C, Holt SG, Pasch A. Serum calcification propensity predicts all-cause mortality in predialysis CKD. J Am Soc Nephrol 2013; 25:339-48. [PMID: 24179171 DOI: 10.1681/asn.2013060635] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Medial arterial calcification is accelerated in patients with CKD and strongly associated with increased arterial rigidity and cardiovascular mortality. Recently, a novel in vitro blood test that provides an overall measure of calcification propensity by monitoring the maturation time (T50) of calciprotein particles in serum was described. We used this test to measure serum T50 in a prospective cohort of 184 patients with stages 3 and 4 CKD, with a median of 5.3 years of follow-up. At baseline, the major determinants of serum calcification propensity included higher serum phosphate, ionized calcium, increased bone osteoclastic activity, and lower free fetuin-A, plasma pyrophosphate, and albumin concentrations, which accounted for 49% of the variation in this parameter. Increased serum calcification propensity at baseline independently associated with aortic pulse wave velocity in the complete cohort and progressive aortic stiffening over 30 months in a subgroup of 93 patients. After adjustment for demographic, renal, cardiovascular, and biochemical covariates, including serum phosphate, risk of death among patients in the lowest T50 tertile was more than two times the risk among patients in the highest T50 tertile (adjusted hazard ratio, 2.2; 95% confidence interval, 1.1 to 5.4; P=0.04). This effect was lost, however, after additional adjustment for aortic stiffness, suggesting a shared causal pathway. Longitudinally, serum calcification propensity measurements remained temporally stable (intraclass correlation=0.81). These results suggest that serum T50 may be helpful as a biomarker in designing methods to improve defenses against vascular calcification.
Collapse
Affiliation(s)
- Edward R Smith
- Department of Renal Medicine, Eastern Health Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lau WL, Ix JH. Clinical detection, risk factors, and cardiovascular consequences of medial arterial calcification: a pattern of vascular injury associated with aberrant mineral metabolism. Semin Nephrol 2013; 33:93-105. [PMID: 23465497 DOI: 10.1016/j.semnephrol.2012.12.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Patients with end-stage renal disease are characterized by extensive vascular calcification and high cardiovascular disease (CVD) risk. Calcification in end-stage renal disease patients represents at least two distinct pathologic processes. Calcification within the tunica intima frequently is associated with lipid-laden, flow-limiting atherosclerotic plaques. These appear as spotty areas of calcification interspersed with noncalcified arterial segments on plain radiography and generally are found near arterial branch points in medium-sized conduit arteries. In contrast, medial arterial calcification (MAC) involves deeper layers of the arterial wall; tends to affect the artery diffusely, appearing as a linear contiguous tram-track pattern of calcification on plain radiography; and often involves smaller muscular arteries such as the radial artery, intermammary arteries, and arteries in the ankle and foot. Both are related to CVD events, but potentially through different mechanisms. Atherosclerotic calcification may be marking the total burden of atherosclerosis, whereas MAC may lead to arterial stiffness and left ventricular hypertrophy. Existing data suggest that altered mineral metabolism may promote MAC, whereas heightened inflammation and oxidative stress contribute to atherosclerosis. Dysregulation of normal anticalcification factors and elastin degradation are common to both processes. Risk of vascular calcification also may be increased by the use of certain medications in the setting of chronic kidney disease. This review compares and contrasts known risk factors for MAC and atherosclerosis, describes existing and emerging technologies to distinguish between them, and reviews the existing literature linking each with CVD events in dialysis patients and in other settings.
Collapse
Affiliation(s)
- Wei Ling Lau
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
| | | |
Collapse
|