1
|
Wesley CD, Neutel CHG, De Meyer GRY, Martinet W, Guns PJ. Unravelling the impact of active and passive contributors to arterial stiffness in male mice and their role in vascular aging. Sci Rep 2024; 14:18337. [PMID: 39112507 PMCID: PMC11306354 DOI: 10.1038/s41598-024-68725-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Arterial stiffness, a key indicator of vascular health, encompassing active (vascular tone) and passive (extracellular matrix) components. This study aims to address how these different components affect arterial stiffness along the aorta and the influence of aging. Aortic segments of 12 week and 24 month old (both n = 6) male C57BL/6J mice were mounted in a Rodent Oscillatory Set-up to study Arterial Compliance, in order to measure arterial stiffness and vascular reactivity. Regional variations in arterial stiffness were evident, with abdominal infrarenal aorta (AIA) exhibiting highest stiffness and smallest diameters. AIA displayed both the highest amount of collagen and collagen:elastin ratio. Regional ex vivo vascular reactivity revealed heightened AIA contractions and lowered NO availability. Aging is a significant factor contributing towards vessel remodelling and arterial stiffness. Aging increased arterial stiffness, aortic diameters, collagen content, and reduced VSMC contraction. The results of this study could identify specific regions or mechanisms to target in the development of innovative therapeutic interventions aimed at enhancing overall vascular health.
Collapse
Affiliation(s)
- Callan D Wesley
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences and Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium.
| | - Cedric H G Neutel
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences and Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences and Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences and Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences and Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| |
Collapse
|
2
|
Bosman M, Krüger D, Van Assche C, Boen H, Neutel C, Favere K, Franssen C, Martinet W, Roth L, De Meyer GRY, Cillero-Pastor B, Delrue L, Heggermont W, Van Craenenbroeck EM, Guns PJ. Doxorubicin-induced cardiovascular toxicity: a longitudinal evaluation of functional and molecular markers. Cardiovasc Res 2023; 119:2579-2590. [PMID: 37625456 PMCID: PMC10676457 DOI: 10.1093/cvr/cvad136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 06/19/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
AIMS Apart from cardiotoxicity, the chemotherapeutic doxorubicin (DOX) induces vascular toxicity, represented by arterial stiffness and endothelial dysfunction. Both parameters are of interest for cardiovascular risk stratification as they are independent predictors of future cardiovascular events in the general population. However, the time course of DOX-induced cardiovascular toxicity remains unclear. Moreover, current biomarkers for cardiovascular toxicity prove insufficient. Here, we longitudinally evaluated functional and molecular markers of DOX-induced cardiovascular toxicity in a murine model. Molecular markers were further validated in patient plasma. METHODS AND RESULTS DOX (4 mg/kg) or saline (vehicle) was administered intra-peritoneally to young, male mice weekly for 6 weeks. In vivo cardiovascular function and ex vivo arterial stiffness and vascular reactivity were evaluated at baseline, during DOX therapy (Weeks 2 and 4) and after therapy cessation (Weeks 6, 9, and 15). Left ventricular ejection fraction (LVEF) declined from Week 4 in the DOX group. DOX increased arterial stiffness in vivo and ex vivo at Week 2, which reverted thereafter. Importantly, DOX-induced arterial stiffness preceded reduced LVEF. Further, DOX impaired endothelium-dependent vasodilation at Weeks 2 and 6, which recovered at Weeks 9 and 15. Conversely, contraction with phenylephrine was consistently higher in the DOX-treated group. Furthermore, proteomic analysis on aortic tissue identified increased thrombospondin-1 (THBS1) and alpha-1-antichymotrypsin (SERPINA3) at Weeks 2 and 6. Up-regulated THBS1 and SERPINA3 persisted during follow-up. Finally, THBS1 and SERPINA3 were quantified in plasma of patients. Cancer survivors with anthracycline-induced cardiotoxicity (AICT; LVEF < 50%) showed elevated THBS1 and SERPINA3 levels compared with age-matched control patients (LVEF ≥ 60%). CONCLUSIONS DOX increased arterial stiffness and impaired endothelial function, which both preceded reduced LVEF. Vascular dysfunction restored after DOX therapy cessation, whereas cardiac dysfunction persisted. Further, we identified SERPINA3 and THBS1 as promising biomarkers of DOX-induced cardiovascular toxicity, which were confirmed in AICT patients.
Collapse
Affiliation(s)
- Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Dustin Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Charles Van Assche
- Research Group M4I—Imaging Mass Spectrometry (IMS); Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Hanne Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Cédric Neutel
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Kasper Favere
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Lynn Roth
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Berta Cillero-Pastor
- Research Group M4I—Imaging Mass Spectrometry (IMS); Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- Department of Cell Biology-Inspired Tissue Engineering, Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER Maastricht/Room C3.577, PO Box 616, Maastricht 6200 MD, The Netherlands
| | - Leen Delrue
- Department of Cardiology, Cardiovascular Center OLV Hospital Aalst, Moorselbaan 164, Aalst B-9300, Belgium
| | - Ward Heggermont
- Department of Cardiology, Cardiovascular Center OLV Hospital Aalst, Moorselbaan 164, Aalst B-9300, Belgium
- Department of Cardiology, Center for Molecular and Vascular Biology, KU Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| |
Collapse
|
3
|
De Moudt S, Hendrickx JO, De Meyer GRY, Martinet W, Fransen P. Disparate biomechanical properties of the aorta in non-aneurysmal and aneurysmal mice treated with angiotensin II. Physiol Rep 2022; 10:e15410. [PMID: 36117398 PMCID: PMC9483617 DOI: 10.14814/phy2.15410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023] Open
Abstract
In vivo angiotensin II (AngII)-treatment is a widely used experimental model to induce cardiovascular disease and results in a high likelihood of abdominal aorta aneurysm (AAA) formation. This involves progressive and irreversible focal dilation of the abdominal aorta and induces adverse aortic connective tissue remodeling contributing to aortic wall stiffening through inflammation, elastin degradation, and collagen restructuring. Hence, the present study aimed to investigate how AAA formation in AngII-treated mice affects aortic function and biomechanics. To this end, C57Bl/6J mice were treated with AngII (1000 ng/[kg.min]) or PBS infusion for 28 days. Peripheral blood pressure, echocardiography, and aortic pulse wave velocity were measured in vivo. Thoracic aorta rings were studied ex vivo in organ chambers, while aortic vascular smooth muscle cell (VSMC) phenotype was investigated histologically. We confirmed peripheral hypertension, cardiac hypertrophy, aortic stiffening, and increased VSMC proliferation and migration after AngII-treatment. Abdominal aorta aneurysm formation was observed in 8/13 AngII-treated mice. Ex vivo thoracic aortic rings of both aneurysmal and non-aneurysmal AngII-treated mice showed high isobaric aortic stiffness, endothelial dysfunction, heightened α1 -adrenergic contractility, and altered VSMC contractile calcium signaling. However, aortic biomechanics were differently affected, with heightened α1 -adrenoreceptor mediated aortic stiffening in non-aneurysmal mice, whereas contraction-dependent stiffening was impaired in aneurysmal mice. In conclusion, although aneurysmal and non-aneurysmal 4-week AngII-treated mice displayed similar changes in aortic physiology, aortic biomechanics were dissimilarly affected.
Collapse
Affiliation(s)
- Sofie De Moudt
- Laboratory of PhysiopharmacologyUniversity of AntwerpAntwerpBelgium
| | | | | | - Wim Martinet
- Laboratory of PhysiopharmacologyUniversity of AntwerpAntwerpBelgium
| | - Paul Fransen
- Laboratory of PhysiopharmacologyUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
4
|
Whitehead AK, Fried ND, Li Z, Neelamegam K, Pearson CS, LaPenna KB, Sharp TE, Lefer DJ, Lazartigues E, Gardner JD, Yue X. Alpha7 nicotinic acetylcholine receptor mediates chronic nicotine inhalation-induced cardiopulmonary dysfunction. Clin Sci (Lond) 2022; 136:973-987. [PMID: 35678315 PMCID: PMC10199464 DOI: 10.1042/cs20220083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/27/2022] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Cigarette smoking remains the leading modifiable risk factor for cardiopulmonary diseases; however, the effects of nicotine alone on cardiopulmonary function remain largely unknown. Previously, we have shown that chronic nicotine vapor inhalation in mice leads to the development of pulmonary hypertension (PH) with right ventricular (RV) remodeling. The present study aims to further examine the cardiopulmonary effects of nicotine and the role of the α7 nicotinic acetylcholine receptor (α7-nAChR), which is widely expressed in the cardiovascular system. Wild-type (WT) and α7-nAChR knockout (α7-nAChR-/-) mice were exposed to room air (control) or nicotine vapor daily for 12 weeks. Consistent with our previous study, echocardiography and RV catheterization reveal that male WT mice developed increased RV systolic pressure with RV hypertrophy and dilatation following 12-week nicotine vapor exposure; in contrast, these changes were not observed in male α7-nAChR-/- mice. In addition, chronic nicotine inhalation failed to induce PH and RV remodeling in female mice regardless of genotype. The effects of nicotine on the vasculature were further examined in male mice. Our results show that chronic nicotine inhalation led to impaired acetylcholine-mediated vasodilatory response in both thoracic aortas and pulmonary arteries, and these effects were accompanied by altered endothelial nitric oxide synthase phosphorylation (enhanced inhibitory phosphorylation at threonine 495) and reduced plasma nitrite levels in WT but not α7-nAChR-/- mice. Finally, RNA sequencing revealed up-regulation of multiple inflammatory pathways in thoracic aortas from WT but not α7-nAChR-/- mice. We conclude that the α7-nAChR mediates chronic nicotine inhalation-induced PH, RV remodeling and vascular dysfunction.
Collapse
Affiliation(s)
- Anna K. Whitehead
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Nicholas D. Fried
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Zhen Li
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Kandasamy Neelamegam
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Charlotte S. Pearson
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Kyle B. LaPenna
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Thomas E. Sharp
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Department of Medicine Section of Cardiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - David J. Lefer
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Southeast Louisiana Veterans Health Care Systems, New Orleans, LA 70119, U.S.A
| | - Jason D. Gardner
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| |
Collapse
|
5
|
Van den Bergh G, Van den Branden A, Opdebeeck B, Fransen P, Neven E, De Meyer G, D’Haese PC, Verhulst A. Endothelial dysfunction aggravates arterial media calcification in warfarin administered rats. FASEB J 2022; 36:e22315. [DOI: 10.1096/fj.202101919r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Geoffrey Van den Bergh
- Laboratory of Pathophysiology Department of Biomedical Sciences University of Antwerp Wilrijk Belgium
| | - Astrid Van den Branden
- Laboratory of Pathophysiology Department of Biomedical Sciences University of Antwerp Wilrijk Belgium
| | - Britt Opdebeeck
- Laboratory of Pathophysiology Department of Biomedical Sciences University of Antwerp Wilrijk Belgium
| | - Paul Fransen
- Laboratory of Physiopharmacology Department of Pharmaceutical Sciences University of Antwerp Wilrijk Belgium
| | - Ellen Neven
- Laboratory of Pathophysiology Department of Biomedical Sciences University of Antwerp Wilrijk Belgium
| | - Guido De Meyer
- Laboratory of Physiopharmacology Department of Pharmaceutical Sciences University of Antwerp Wilrijk Belgium
| | - Patrick C. D’Haese
- Laboratory of Pathophysiology Department of Biomedical Sciences University of Antwerp Wilrijk Belgium
| | - Anja Verhulst
- Laboratory of Pathophysiology Department of Biomedical Sciences University of Antwerp Wilrijk Belgium
| |
Collapse
|
6
|
Doxorubicin Impairs Smooth Muscle Cell Contraction: Novel Insights in Vascular Toxicity. Int J Mol Sci 2021; 22:ijms222312812. [PMID: 34884612 PMCID: PMC8657832 DOI: 10.3390/ijms222312812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 01/12/2023] Open
Abstract
Clinical and animal studies have demonstrated that chemotherapeutic doxorubicin (DOX) increases arterial stiffness, a predictor of cardiovascular risk. Despite consensus about DOX-impaired endothelium-dependent vasodilation as a contributing mechanism, some studies have reported conflicting results on vascular smooth muscle cell (VSMC) function after DOX treatment. The present study aimed to investigate the effects of DOX on VSMC function. To this end, mice received a single injection of 4 mg DOX/kg, or mouse aortic segments were treated ex vivo with 1 μM DOX, followed by vascular reactivity evaluation 16 h later. Phenylephrine (PE)-induced VSMC contraction was decreased after DOX treatment. DOX did not affect the transient PE contraction dependent on Ca2+ release from the sarcoplasmic reticulum (0 mM Ca2+), but it reduced the subsequent tonic phase characterised by Ca2+ influx. These findings were supported by similar angiotensin II and attenuated endothelin-1 contractions. The involvement of voltage-gated Ca2+ channels in DOX-decreased contraction was excluded by using levcromakalim and diltiazem in PE-induced contraction and corroborated by similar K+ and serotonin contractions. Despite the evaluation of multiple blockers of transient receptor potential channels, the exact mechanism for DOX-decreased VSMC contraction remains elusive. Surprisingly, DOX reduced ex vivo but not in vivo arterial stiffness, highlighting the importance of appropriate timing for evaluating arterial stiffness in DOX-treated patients.
Collapse
|
7
|
Tehrani AY, Ciufolini MA, Bernatchez P. Nitric oxide in the Marfan vasculature: Friend or foe? Nitric Oxide 2021; 116:27-34. [PMID: 34478846 DOI: 10.1016/j.niox.2021.08.006] [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: 07/04/2021] [Revised: 08/13/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the FBN1 gene, which encodes fibrillin-1, a protein essential for the formation and stabilization of elastic fibers as well as signaling homeostasis. Progressive aortic root widening is the most serious manifestation of MFS as it can lead to aortic dissection, aneurysm formation and rupture. However, despite their ability to decrease the hemodynamic stress the aorta is subjected to, anti-hypertensive medications often lead to underwhelming reductions in the rate of aortic root dilation, which illustrates how fragmental our understanding of MFS-associated aortic remodeling is. This manuscript summarizes recent evidence that document nitric oxide (NO) synthase (NOS)-related changes to the vasculature during the pathogenesis of MFS and how they result in a unique state of vascular dysfunction that likely plays a causal role in the aortic root widening process. We also review how clinic-approved and experimental therapies as well lifestyle approaches may promote aortic root stability by correcting NO homeostasis, which if properly optimized may improve outcomes in this population afflicted by a notoriously refractory type of aortopathy.
Collapse
Affiliation(s)
- Arash Y Tehrani
- Centre for Heart + Lung Innovation, St. Paul's Hospital, Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, Canada
| | | | - Pascal Bernatchez
- Centre for Heart + Lung Innovation, St. Paul's Hospital, Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, Canada.
| |
Collapse
|
8
|
Endothelial Contribution to Warfarin-Induced Arterial Media Calcification in Mice. Int J Mol Sci 2021; 22:ijms222111615. [PMID: 34769044 PMCID: PMC8583869 DOI: 10.3390/ijms222111615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Arterial media calcification (AMC) is predominantly regulated by vascular smooth muscle cells (VSMCs), which transdifferentiate into pro-calcifying cells. In contrast, there is little evidence for endothelial cells playing a role in the disease. The current study investigates cellular functioning and molecular pathways underlying AMC, respectively by, an ex vivo isometric organ bath set-up to explore the interaction between VSMCs and ECs and quantitative proteomics followed by functional pathway interpretation. AMC development, which was induced in mice by dietary warfarin administration, was proved by positive Von Kossa staining and a significantly increased calcium content in the aorta compared to that of control mice. The ex vivo organ bath set-up showed calcified aortic segments to be significantly more sensitive to phenylephrine induced contraction, compared to control segments. This, together with the fact that calcified segments as compared to control segments, showed a significantly smaller contraction in the absence of extracellular calcium, argues for a reduced basal NO production in the calcified segments. Moreover, proteomic data revealed a reduced eNOS activation to be part of the vascular calcification process. In summary, this study identifies a poor endothelial function, next to classic pro-calcifying stimuli, as a possible initiator of arterial calcification.
Collapse
|
9
|
GSK-7975A, an inhibitor of Ca 2+ release-activated calcium channels, depresses isometric contraction of mouse aorta. Eur J Pharmacol 2021; 906:174197. [PMID: 34052216 DOI: 10.1016/j.ejphar.2021.174197] [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: 02/02/2021] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 11/21/2022]
Abstract
GSK-7975A is described to inhibit stromal interaction molecule 1(STIM1)-mediated Ca2+ release-activated Ca2+ channels ORAI 1, ORAI 2 and ORAI 3 in different cell types. The present study investigated whether isometric contractions of mouse aortic segments were affected by this selective store-operated calcium channel inhibitor. Depending on the way by which Ca2+ influx pathways were activated during contraction, GSK-7975A inhibited contractility of mouse aortic segments with different affinity. When contractile effects were induced by depolarization as with elevated extracellular K+ and opening of voltage-gated calcium channels, the affinity was approximately 10 times lower than when contraction was elicited with Ca2+ influx via non-selective cation channels. GSK-7975A may repolarize the aortic smooth muscle cells by inhibiting non-selective cation channels, has no effect on IP3-mediated phenylephrine-induced phasic contractions or on refilling of the contractile sarcoplasmic reticulum Ca2+ store, but has significant effects on non-contractile store-operated Ca2+ influx.
Collapse
|
10
|
Bosman M, Favere K, Neutel CHG, Jacobs G, De Meyer GRY, Martinet W, Van Craenenbroeck EM, Guns PJDF. Doxorubicin induces arterial stiffness: A comprehensive in vivo and ex vivo evaluation of vascular toxicity in mice. Toxicol Lett 2021; 346:23-33. [PMID: 33895255 DOI: 10.1016/j.toxlet.2021.04.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/29/2021] [Accepted: 04/20/2021] [Indexed: 01/09/2023]
Abstract
Arterial stiffness is an important predictor of cardiovascular risk. Clinical studies have demonstrated that arterial stiffness increases in cancer patients treated with the chemotherapeutic doxorubicin (DOX). However, the mechanisms of DOX-induced arterial stiffness remain largely unknown. This study aimed to evaluate artery stiffening in DOX-treated mice using in vivo and ex vivo techniques. Male C57BL/6J mice were treated for 2 weeks with 2 mg/kg (low dose) or 4 mg/kg (high dose) of DOX weekly. Arterial stiffness was assessed in vivo with ultrasound imaging (abdominal aorta pulse wave velocity (aaPWV)) and applanation tonometry (carotid-femoral PWV) combined with ex vivo vascular stiffness and reactivity evaluation. The high dose increased aaPWV, while cfPWV did not reach statistical significance. Phenylephrine (PE)-contracted aortic segments showed a higher Peterson's modulus (Ep) in the high dose group, while Ep did not differ when vascular smooth muscle cells (VSMCs) were relaxed by a NO donor (DEANO). In addition, aortic rings of DOX-treated mice showed increased PE contraction, decreased basal nitric oxide (NO) index and impaired acetylcholine-induced endothelium-dependent relaxation. DOX treatment contributed to endothelial cell loss and reduced endothelial nitric oxide synthase (eNOS) expression in the aorta. In conclusion, we have replicated DOX-induced arterial stiffness in a murine model and this aortic stiffness is driven by impaired endothelial function, contributing to increased vascular tone.
Collapse
Affiliation(s)
- Matthias Bosman
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium.
| | - Kasper Favere
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium; University of Antwerp, Research Group Cardiovascular Diseases, GENCOR, Antwerp, Belgium; Ghent University, Faculty of Medicine and Health Sciences, Department of Internal Medicine, C. Heymanslaan 10, B-9000, Ghent, Belgium; Antwerp University Hospital (UZA), Department of Cardiology, Drie Eikenstraat 655, B-2650, Edegem, Belgium
| | - Cédric H G Neutel
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Griet Jacobs
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Guido R Y De Meyer
- University of Antwerp, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Wim Martinet
- University of Antwerp, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- University of Antwerp, Research Group Cardiovascular Diseases, GENCOR, Antwerp, Belgium; Antwerp University Hospital (UZA), Department of Cardiology, Drie Eikenstraat 655, B-2650, Edegem, Belgium
| | - Pieter-Jan D F Guns
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| |
Collapse
|
11
|
Leloup AJA, Van Hove CE, De Moudt S, De Keulenaer GW, Fransen P. Ex vivo aortic stiffness in mice with different eNOS activity. Am J Physiol Heart Circ Physiol 2020; 318:H1233-H1244. [DOI: 10.1152/ajpheart.00737.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endothelial function and NO bioavailability are important determinants of aortic biomechanics and function. With a new technique we investigated the ex vivo aortic segment biomechanics of different mouse models with altered NO signaling. Our experiments clearly show that chronic distortion of NO signaling triggered several compensatory mechanisms that reflect the organism’s attempt to maintain optimal central hemodynamics.
Collapse
Affiliation(s)
- Arthur J. A. Leloup
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Cor E. Van Hove
- Faculty of Medicine and Health Sciences, Laboratory of Pharmacology, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Gilles W. De Keulenaer
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Middelheim Hospital, Antwerp, Belgium
| | - Paul Fransen
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
12
|
Fransen P, Chen J, Vangheluwe P, Guns PJ. Contractile Behavior of Mouse Aorta Depends on SERCA2 Isoform Distribution: Effects of Replacing SERCA2a by SERCA2b. Front Physiol 2020; 11:282. [PMID: 32296344 PMCID: PMC7136392 DOI: 10.3389/fphys.2020.00282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/12/2020] [Indexed: 01/16/2023] Open
Abstract
The Sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) actively pumps Ca2+ into the sarco/endoplasmic reticulum, thereby regulating intracellular Ca2+ concentrations and associated physiological processes. Different SERCA isoforms have been described (SERCA1, 2, and 3) with SERCA2 playing a pivotal role in Ca2+ homeostasis in cardiovascular tissues. In the heart, SERCA2a is the dominant isoform and has been proposed as therapeutic target in patients with heart failure. In the vasculature, both SERCA2a and SERCA2b are expressed with SERCA2b being the predominant isoform. The physiological role of SERCA2a in the vasculature, however, remains incompletely understood. In the present study, we used gene-modified mice in which the alternative splicing of the SERCA2-encoding gene (Atp2a2), underlying the expression of SERCA2a, is prevented and SERCA2a is replaced by SERCA2b. The resulting SERCA2b/b mice provide a unique opportunity to investigate the specific contribution of SERCA2a versus SERCA2b to vascular physiology. Aortic segments of SERCA2b/b (SERCA2a-deficient) and SERCA2a/b (control) mice were mounted in organ baths to evaluate vascular reactivity. SERCA2b/b aortic rings displayed higher contractions induced by phenylephrine (1 μM). Surprisingly, the initial inositol-3-phosphate mediated phasic contraction showed a faster decay of force in SERCA2b/b mice, while the subsequent tonic contraction was larger in SERCA2b/b segments. Moreover, in the presence of the calcium channel blocker diltiazem (35 μM) SERCA2b/b aortic rings showed higher contractions compared to SERCA2a/b, suggesting that SERCA2a (deficiency) modulates the activity of non-selective cation channels. Additionally, in endothelial cell (EC)-denuded aortic segments, the SERCA-inhibitor cyclopiazonic acid (CPA) caused markedly larger contractions in SERCA2b/b mice, while the increases of cytosolic Ca2+ were similar in both strains. Hence, aortas of SERCA2b/b mice appear to have a stronger coupling of intracellular Ca2+ to contraction, which may be in agreement with the reported difference in intracellular localization of SERCA2a versus SERCA2b. Finally, EC-mediated relaxation by acetylcholine and ATP was assessed. Concentration-response-curves for ATP showed a higher sensitivity of aortic segments of SERCA2b/b mice, while no difference in potency between strains were observed for acetylcholine. In summary, despite the relative low expression of SERCA2a in the murine aorta, our results point toward a distinct role in vascular physiology.
Collapse
Affiliation(s)
- Paul Fransen
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical Sciences and Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Jialin Chen
- Laboratory of Cellular Transport Systems, KU Leuven, Leuven, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, KU Leuven, Leuven, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical Sciences and Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
13
|
Leloup AJA, Van Hove CE, De Moudt S, De Meyer GRY, De Keulenaer GW, Fransen P. Vascular smooth muscle cell contraction and relaxation in the isolated aorta: a critical regulator of large artery compliance. Physiol Rep 2019; 7:e13934. [PMID: 30810292 PMCID: PMC6391714 DOI: 10.14814/phy2.13934] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, isometric contraction studies of isolated thoracic aorta segments have significantly contributed to our overall understanding of the active, contractile properties of aortic vascular smooth muscle cells (VSMCs) and their cross-talk with endothelial cells. However, the physiological role of VSMC contraction or relaxation in the healthy aorta and its contribution to the pulse-smoothening capacity of the aorta is currently unclear. Therefore, we investigated the acute effects of VSMC contraction and relaxation on the isobaric biomechanical properties of healthy mouse aorta. An in-house developed set-up was used to measure isobaric stiffness parameters of periodically stretched (10 Hz) aortic segments at an extended pressure range, while pharmacologically modulating VSMC tone and endothelial cell function. We found that the effects of α1-adrenergic stimulation with phenylephrine on the pressure-stiffness relationship varied in sensitivity, magnitude and direction, with the basal, unstimulated NO production by the endothelium playing a pivotal role. We also investigated how arterial disease affected this system by using the angiotensin-II-treated mouse. Our results show that isobaric stiffness was increased and that the aortic segments demonstrated a reduced capacity for modulating the pressure-stiffness relationship. This suggests that not only increased isobaric stiffness at normal pressure, but also a reduced capacity of the VSMCs to limit the pressure-associated increase in aortic stiffness, may contribute to the pathogenesis of this mouse model. Overall, this study provides more insight in how aortic VSMC tone affects the pressure-dependency of aortic biomechanics at different physiological and pathological conditions.
Collapse
Affiliation(s)
- Arthur J. A. Leloup
- Laboratory of PhysiopharmacologyDepartment of Pharmaceutical SciencesUniversity of AntwerpAntwerpBelgium
| | - Cor E. Van Hove
- Laboratory of PharmacologyFaculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Sofie De Moudt
- Laboratory of PhysiopharmacologyDepartment of Pharmaceutical SciencesUniversity of AntwerpAntwerpBelgium
| | - Guido R. Y. De Meyer
- Laboratory of PhysiopharmacologyDepartment of Pharmaceutical SciencesUniversity of AntwerpAntwerpBelgium
| | - Gilles W. De Keulenaer
- Laboratory of PhysiopharmacologyDepartment of Pharmaceutical SciencesUniversity of AntwerpAntwerpBelgium
| | - Paul Fransen
- Laboratory of PhysiopharmacologyDepartment of Pharmaceutical SciencesUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
14
|
Leloup AJA, De Moudt S, Van Hove CE, Dugaucquier L, Vermeulen Z, Segers VFM, De Keulenaer GW, Fransen P. Short-Term Angiotensin II Treatment Affects Large Artery Biomechanics and Function in the Absence of Small Artery Alterations in Mice. Front Physiol 2018; 9:582. [PMID: 29867592 PMCID: PMC5964213 DOI: 10.3389/fphys.2018.00582] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/01/2018] [Indexed: 02/01/2023] Open
Abstract
Induction of hypertension by angiotensin II (AngII) is a widely used experimental stimulus to study vascular aging in mice. It is associated with large artery stiffness, a hallmark of arterial aging and a root cause of increased cardiovascular risk. We reported earlier that long term (4 week) AngII treatment in mice altered the active, contractile properties of the arteries in a vascular bed-specific manner and that, in healthy mice aorta, active contractile properties of the aortic wall determine isobaric aortic stiffness. Given the huge physiological relevance of large artery stiffening, we aimed to characterize the early (1 week) changes in the active properties of the aorta of AngII-treated mice. We were not able to detect a significant effect of AngII treatment on anesthetized blood pressure or abdominal aorta pulse wave velocity. Ex vivo biomechanical and functional studies of the aorta revealed increased arterial stiffness and altered vascular smooth muscle cell (VSMC) and endothelial cell reactivity. Interestingly, the AngII-associated changes in the aorta could be largely attributed to alterations in basal VSMC tone and basal nitric oxide efficacy, indicating that, besides structural remodeling of the arterial wall, dysfunctional active components of the aorta play a crucial role in the pathophysiological mechanisms by which AngII treatment induces arterial stiffness.
Collapse
Affiliation(s)
- Arthur J A Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Cor E Van Hove
- Laboratory of Pharmacology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Lindsey Dugaucquier
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Zarha Vermeulen
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Antwerp University Hospital, Edegem, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Middelheim Hospital, Antwerp, Belgium
| | - Paul Fransen
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
15
|
Leloup A, De Moudt S, Van Hove C, Fransen P. Cyclic Stretch Alters Vascular Reactivity of Mouse Aortic Segments. Front Physiol 2017; 8:858. [PMID: 29163203 PMCID: PMC5674939 DOI: 10.3389/fphys.2017.00858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/13/2017] [Indexed: 11/13/2022] Open
Abstract
Large, elastic arteries buffer the pressure wave originating in the left ventricle and are constantly exposed to higher amplitudes of cyclic stretch (10%) than muscular arteries (2%). As a crucial factor for endothelial and smooth muscle cell function, cyclic stretch has, however, never been studied in ex vivo aortic segments of mice. To investigate the effects of cyclic stretch on vaso-reactivity of mouse aortic segments, we used the Rodent Oscillatory Tension Set-up to study Arterial Compliance (ROTSAC). The aortic segments were clamped at frequencies of 6–600 bpm between two variable preloads, thereby mimicking dilation as upon left ventricular systole and recoiling as during diastole. The preloads corresponding to different transmural pressures were chosen to correspond to a low, normal or high amplitude of cyclic stretch. At different time intervals, cyclic stretch was interrupted, the segments were afterloaded and isometric contractions by α1-adrenergic stimulation with 2 μM phenylephrine in the absence and presence of 300 μM L-NAME (eNOS inhibitor) and/or 35 μM diltiazem (blocker of voltage-gated Ca2+ channels) were measured. As compared with static or cyclic stretch at low amplitude (<10 mN) or low frequency (0.1 Hz), cyclic stretch at physiological amplitude (>10 mN) and frequency (1–10 Hz) caused better ex vivo conservation of basal NO release with time after mounting. The relaxation of PE-precontracted segments by addition of ACh to stimulate NO release was unaffected by cyclic stretch. In the absence of basal NO release (hence, presence of L-NAME), physiological in comparison with aberrant cyclic stretch decreased the baseline tension, attenuated the phasic contraction by phenylephrine in the absence of extracellular Ca2+ and shifted the smaller tonic contraction more from a voltage-gated Ca2+ channel-mediated to a non-selective cation channel-mediated. Data highlight the need of sufficient mechanical activation of endothelial and vascular smooth muscle cells to maintain basal NO release and low intracellular Ca2+ in the smooth muscle cells in large arteries. Both phenomena may play a vital role in maintaining the high compliance of large arteries.
Collapse
Affiliation(s)
- Arthur Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Cor Van Hove
- Laboratory of Pharmacology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Paul Fransen
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
16
|
Gevaert AB, Shakeri H, Leloup AJ, Van Hove CE, De Meyer GRY, Vrints CJ, Lemmens K, Van Craenenbroeck EM. Endothelial Senescence Contributes to Heart Failure With Preserved Ejection Fraction in an Aging Mouse Model. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003806. [PMID: 28611124 DOI: 10.1161/circheartfailure.116.003806] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/01/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Because of global aging, the prevalence of heart failure with preserved ejection fraction (HFpEF) continues to rise. Although HFpEF pathophysiology remains incompletely understood, endothelial inflammation is stated to play a central role. Cellular senescence is a process of cellular growth arrest linked with aging and inflammation. We used mice with accelerated aging to investigate the role of cellular senescence in HFpEF development. METHODS AND RESULTS Senescence-accelerated mice (SAM, n=18) and control mice with normal senescence (n=15) were fed normal chow or a high-fat, high-salt diet (WD). Vascular and cardiac function was assessed at 8, 16, and 24 weeks of age. At 24 weeks, both SAM on WD (SAM-WD) and SAM on regular diet displayed endothelial dysfunction, as evidenced by impaired acetylcholine-induced relaxation of aortic segments and reduced basal nitric oxide. At week 24, SAM-WD had developed HFpEF, characterized by diastolic dysfunction, left ventricular hypertrophy, left atrial dilatation, and interstitial fibrosis. Also, exercise capacity was reduced and lung weight increased. Cardiovascular inflammation and senescence were assessed by immunohistochemical and immunofluorescence staining of hearts and aortas. SAM-WD showed increased endothelial inflammation (intercellular adhesion molecule 1 expression) and increased endothelial senescence (acetyl-p53/CD31 costaining). The latter correlated with diastolic function and intercellular adhesion molecule 1 expression. CONCLUSIONS SAM develop endothelial dysfunction. Adding a high-salt, high-fat diet accelerates endothelial senescence and instigates endothelial inflammation. This coincides with hemodynamic and structural changes typical of HFpEF. Targeting endothelial senescence could be a new therapeutic avenue in HFpEF.
Collapse
Affiliation(s)
- Andreas B Gevaert
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.).
| | - Hadis Shakeri
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Arthur J Leloup
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Cor E Van Hove
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Guido R Y De Meyer
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Christiaan J Vrints
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Katrien Lemmens
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Emeline M Van Craenenbroeck
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| |
Collapse
|
17
|
De Moudt S, Leloup A, Van Hove C, De Meyer G, Fransen P. Isometric Stretch Alters Vascular Reactivity of Mouse Aortic Segments. Front Physiol 2017; 8:157. [PMID: 28360864 PMCID: PMC5352655 DOI: 10.3389/fphys.2017.00157] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/28/2017] [Indexed: 02/02/2023] Open
Abstract
Most vaso-reactive studies in mouse aortic segments are performed in isometric conditions and at an optimal preload, which is the preload corresponding to a maximal contraction by non-receptor or receptor-mediated stimulation. In general, this optimal preload ranges from about 1.2 to 8.0 mN/mm, which according to Laplace's law roughly correlates with transmural pressures of 10-65 mmHg. For physiologic transmural pressures around 100 mmHg, preloads of 15.0 mN/mm should be implemented. The present study aimed to compare vascular reactivity of 2 mm mouse (C57Bl6) aortic segments preloaded at optimal (8.0 mN/mm) vs. (patho) physiological (10.0-32.5 mN/mm) preload. Voltage-dependent contractions of aortic segments, induced by increasing extracellular K+, and contractions by α1-adrenergic stimulation with phenylephrine (PE) were studied at these preloads in the absence and presence of L-NAME to inhibit basal release of NO from endothelial cells (EC). In the absence of basal NO release and with higher than optimal preload, contractions evoked by depolarization or PE were attenuated, whereas in the presence of basal release of NO PE-, but not depolarization-induced contractions were preload-independent. Phasic contractions by PE, as measured in the absence of external Ca2+, were decreased at higher than optimal preload suggestive for a lower contractile SR Ca2+ content at physiological preload. Further, in the presence of external Ca2+, contractions by Ca2+ influx via voltage-dependent Ca2+ channels were preload-independent, whereas non-selective cation channel-mediated contractions were increased. The latter contractions were very sensitive to the basal release of NO, which itself seemed to be preload-independent. Relaxation by endogenous NO (acetylcholine) of aortic segments pre-contracted with PE was preload-independent, whereas relaxation by exogenous NO (diethylamine NONOate) displayed higher sensitivity at high preload. Results indicated that stretching aortic segments to higher than optimal preload depolarizes the SMC and causes Ca2+ unloading of the contractile SR, making them extremely sensitive to small changes in the basal release of NO from EC as can occur in hypertension or arterial stiffening.
Collapse
Affiliation(s)
- Sofie De Moudt
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Arthur Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Cor Van Hove
- Laboratory of Pharmacology, Faculty of Medicine and Health Sciences, University of Antwerp Antwerp, Belgium
| | - Guido De Meyer
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Paul Fransen
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| |
Collapse
|
18
|
Leloup AJA, Van Hove CE, Heykers A, Schrijvers DM, De Meyer GRY, Fransen P. Elastic and Muscular Arteries Differ in Structure, Basal NO Production and Voltage-Gated Ca(2+)-Channels. Front Physiol 2015; 6:375. [PMID: 26696904 PMCID: PMC4678217 DOI: 10.3389/fphys.2015.00375] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022] Open
Abstract
In the last decades, the search for mechanisms underlying progressive arterial stiffening and for interventions to avoid or reverse this process has gained much attention. In general, arterial stiffening displays regional variation and is, for example, during aging more prominent in elastic than in muscular arteries. We hypothesize that besides passive also active regulators of arterial compliance [i.e., endothelial and vascular smooth muscle cell (VSMC) function] differ between these arteries. Hence, it is conceivable that these vessel types will display different time frames of stiffening. To investigate this hypothesis segments of muscular arteries such as femoral and mesenteric arteries and elastic arteries such as the aorta and carotid artery were isolated from female C57Bl6 mice (5–6 months of age, n = 8). Both microscopy and passive stretching of the segments in a myograph confirmed that passive mechanical properties (elastin, collagen) of elastic and muscular arteries were significantly different. Endothelial function, more specifically basal nitric oxide (NO) efficacy, and VSMC function, more specifically L-type voltage-gated Ca2+ channel (VGCC)-mediated contractions, were determined by α1-adrenoceptor stimulation with phenylephrine (PE) and by gradual depolarization with elevated extracellular K+ in the absence and presence of eNOS inhibition with L-NAME. PE-mediated isometric contractions significantly increased after inhibition of NO release with L-NAME in elastic, but not in muscular vessel segments. This high basal eNOS activity in elastic vessels was also responsible for shifts of K+ concentration-contraction curves to higher external K+. VGCC-mediated contractions were similarly affected by depolarization with elevated K+ in muscular artery segments or in elastic artery segments in the absence of basal NO. However, K+-induced contractions were inhibited by the VGCC blocker diltiazem with significantly higher sensitivity in the muscular arteries, suggestive of different populations of VGCC isoforms in both vessel types. The results from the present study demonstrate that, besides passive arterial wall components, also active functional components contribute to the heterogeneity of arterial compliance along the vascular tree. This crucially facilitates the search for (patho) physiological mechanisms and potential therapeutic targets to treat or reverse large artery stiffening as occurring in aging-induced arterial stiffening.
Collapse
Affiliation(s)
- Arthur J A Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Cor E Van Hove
- Laboratory of Pharmacology, Faculty of Medicine and Health Sciences, University of Antwerp Antwerp, Belgium
| | - Annick Heykers
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Dorien M Schrijvers
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Paul Fransen
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| |
Collapse
|
19
|
Fransen P, Van Hove CE, Leloup AJA, Schrijvers DM, De Meyer GRY, De Keulenaer GW. Effect of angiotensin II-induced arterial hypertension on the voltage-dependent contractions of mouse arteries. Pflugers Arch 2015; 468:257-67. [PMID: 26432297 DOI: 10.1007/s00424-015-1737-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 09/22/2015] [Indexed: 11/28/2022]
Abstract
Arterial hypertension (AHT) affects the voltage dependency of L-type Ca(2+) channels in cardiomyocytes. We analyzed the effect of angiotensin II (AngII)-induced AHT on L-type Ca(2+) channel-mediated isometric contractions in conduit arteries. AHT was induced in C57Bl6 mice with AngII-filled osmotic mini-pumps (4 weeks). Normotensive mice treated with saline-filled osmotic mini-pumps were used for comparison. Voltage-dependent contractions mediated by L-type Ca(2+) channels were studied in vaso-reactive studies in vitro in isolated aortic and femoral arteries by using extracellular K(+) concentration-response (KDR) experiments. In aortic segments, AngII-induced AHT significantly sensitized isometric contractions induced by elevated extracellular K(+) and depolarization. This sensitization was partly prevented by normalizing blood pressure with hydralazine, suggesting that it was caused by AHT rather than by direct AngII effects on aortic smooth muscle cells. The EC50 for extracellular K(+) obtained in vitro correlated significantly with the rise in arterial blood pressure induced by AngII in vivo. The AHT-induced sensitization persisted when aortic segments were exposed to levcromakalim or to inhibitors of basal nitric oxide release. Consistent with these observations, AngII-treatment also sensitized the vaso-relaxing effects of the L-type Ca(2+) channel blocker diltiazem during K(+)-induced contractions. Unlike aorta, AngII-treatment desensitized the isometric contractions to depolarization in femoral arteries pointing to vascular bed specific responses of arteries to hypertension. AHT affects the voltage-dependent L-type Ca(2+) channel-mediated contraction of conduit arteries. This effect may contribute to the decreased vascular compliance in AHT and explain the efficacy of Ca(2+) channel blockers to reduce vascular stiffness and central blood pressure in AHT.
Collapse
Affiliation(s)
- Paul Fransen
- Department of Pharmaceutical Sciences, Physiopharmacology, Campus Drie Eiken, University of Antwerp, T2, Universiteitsplein 1, 2610, Antwerp, Belgium.
| | - Cor E Van Hove
- Faculty of Medicine & Health Sciences, Pharmacology, Campus Drie Eiken, University of Antwerp, T2, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Arthur J A Leloup
- Department of Pharmaceutical Sciences, Physiopharmacology, Campus Drie Eiken, University of Antwerp, T2, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Dorien M Schrijvers
- Department of Pharmaceutical Sciences, Physiopharmacology, Campus Drie Eiken, University of Antwerp, T2, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Guido R Y De Meyer
- Department of Pharmaceutical Sciences, Physiopharmacology, Campus Drie Eiken, University of Antwerp, T2, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Department of Pharmaceutical Sciences, Physiopharmacology, Campus Drie Eiken, University of Antwerp, T2, Universiteitsplein 1, 2610, Antwerp, Belgium
| |
Collapse
|
20
|
Contribution of α-Adrenoceptor Stimulation by Phenylephrine to Basal Nitric Oxide Production in the Isolated Mouse Aorta. J Cardiovasc Pharmacol 2013; 61:318-23. [DOI: 10.1097/fjc.0b013e318281fa2d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Abstract
In the hippocampus, as in many other CNS areas, nitric oxide (NO) participates in synaptic plasticity, manifested as changes in pre- and/or postsynaptic function. While it is known that these changes are brought about by cGMP following activation of guanylyl cyclase-coupled NO receptors attempts to locate cGMP by immunocytochemistry in hippocampal slices in response to NO have failed to detect the cGMP elevation where expected, i.e. in the pyramidal neurones. Instead, astrocytes, unidentified varicose fibres and GABA-ergic nerve terminals are reported to be the prominent NO targets, raising the possibility that NO acts indirectly via other cells. We have re-investigated the distribution of cGMP generated in response to endogenous and exogenous NO in hippocampal slices using immunohistochemistry and new conditions designed to optimise cGMP accumulation and, hence, its detectability. The conditions included use of tissue from the developing rat hippocampus, a potent inhibitor of phosphodiesterase-2, and an allosteric enhancer of the NO-receptive guanylyl cyclase. Under these conditions, cGMP was formed in response to endogenous NO and was found in a population of pyramidal cell somata in area CA3 and subiculum as well as in structures described previously. The additional presence of exogenous NO resulted in hippocampal cGMP reaching the highest level recorded for brain tissue (1700 pmol/mg protein) and in cGMP immunolabelling throughout the pyramidal cell layer. Populations of axons and interneurones were also stained. According with these results, immunohistochemistry for the common NO receptor β1-subunit indicated widespread expression. A similar staining pattern for the α1-subunit with an antibody used previously in the hippocampus and elsewhere, however, proved to be artefactual. The results indicate that the targets of NO in the hippocampus are more varied and extensive than previous evidence had suggested and, in particular, that the pyramidal neurones participating in NO-dependent synaptic plasticity are direct NO targets.
Collapse
|