1
|
Johnson RT, Solanki R, Wostear F, Ahmed S, Taylor JCK, Rees J, Abel G, McColl J, Jørgensen HF, Morris CJ, Bidula S, Warren DT. Piezo1-mediated regulation of smooth muscle cell volume in response to enhanced extracellular matrix rigidity. Br J Pharmacol 2024; 181:1576-1595. [PMID: 38044463 DOI: 10.1111/bph.16294] [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: 04/12/2023] [Revised: 11/06/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Decreased aortic compliance is a precursor to numerous cardiovascular diseases. Compliance is regulated by the rigidity of the aortic wall and the vascular smooth muscle cells (VSMCs). Extracellular matrix stiffening, observed during ageing, reduces compliance. In response to increased rigidity, VSMCs generate enhanced contractile forces that result in VSMC stiffening and a further reduction in compliance. Mechanisms driving VSMC response to matrix rigidity remain poorly defined. EXPERIMENTAL APPROACH Human aortic-VSMCs were seeded onto polyacrylamide hydrogels whose rigidity mimicked either healthy (12 kPa) or aged/diseased (72 kPa) aortae. VSMCs were treated with pharmacological agents prior to agonist stimulation to identify regulators of VSMC volume regulation. KEY RESULTS On pliable matrices, VSMCs contracted and decreased in cell area. Meanwhile, on rigid matrices VSMCs displayed a hypertrophic-like response, increasing in area and volume. Piezo1 activation stimulated increased VSMC volume by promoting calcium ion influx and subsequent activation of PKC and aquaporin-1. Pharmacological blockade of this pathway prevented the enhanced VSMC volume response on rigid matrices whilst maintaining contractility on pliable matrices. Importantly, both piezo1 and aquaporin-1 gene expression were up-regulated during VSMC phenotypic modulation in atherosclerosis and after carotid ligation. CONCLUSIONS AND IMPLICATIONS In response to extracellular matrix rigidity, VSMC volume is increased by a piezo1/PKC/aquaporin-1 mediated pathway. Pharmacological targeting of this pathway specifically blocks the matrix rigidity enhanced VSMC volume response, leaving VSMC contractility on healthy mimicking matrices intact. Importantly, upregulation of both piezo1 and aquaporin-1 gene expression is observed in disease relevant VSMC phenotypes.
Collapse
Affiliation(s)
| | - Reesha Solanki
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - Finn Wostear
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - Sultan Ahmed
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - James C K Taylor
- Section of Cardiorespiratory Medicine, University of Cambridge, VPD Heart and Lung Research Institute, Cambridge, UK
| | - Jasmine Rees
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - Geraad Abel
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - James McColl
- Henry Wellcome Laboratory for Cell Imaging, University of East Anglia, Norfolk, UK
| | - Helle F Jørgensen
- Section of Cardiorespiratory Medicine, University of Cambridge, VPD Heart and Lung Research Institute, Cambridge, UK
| | - Chris J Morris
- School of Pharmacy, University College London, London, UK
| | - Stefan Bidula
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - Derek T Warren
- School of Pharmacy, University of East Anglia, Norwich, UK
| |
Collapse
|
2
|
Kilanowski-Doroh IM, McNally AB, Wong T, Visniauskas B, Blessinger SA, Sugi AI, Richard C, Diaz Z, Horton A, Natale CA, Ogola BO, Lindsey SH. Ovariectomy-Induced Arterial Stiffening Differs From Vascular Aging and Is Reversed by GPER Activation. Hypertension 2024; 81:e51-e62. [PMID: 38445498 PMCID: PMC11023783 DOI: 10.1161/hypertensionaha.123.22024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/16/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Arterial stiffness is a cardiovascular risk factor and dramatically increases as women transition through menopause. The current study assessed whether a mouse model of menopause increases arterial stiffness in a similar manner to aging and whether activation of the G-protein-coupled estrogen receptor could reverse stiffness. METHODS Female C57Bl/6J mice were ovariectomized at 10 weeks of age or aged to 52 weeks, and some mice were treated with G-protein-coupled estrogen receptor agonists. RESULTS Ovariectomy and aging increased pulse wave velocity to a similar extent independent of changes in blood pressure. Aging increased carotid wall thickness, while ovariectomy increased material stiffness without altering vascular geometry. RNA-sequencing analysis revealed that ovariectomy downregulated smooth muscle contractile genes. The enantiomerically pure G-protein-coupled estrogen receptor agonist, LNS8801, reversed stiffness in ovariectomy mice to a greater degree than the racemic agonist G-1. In summary, ovariectomy and aging induced arterial stiffening via potentially different mechanisms. Aging was associated with inward remodeling, while ovariectomy-induced material stiffness independent of geometry and a loss of the contractile phenotype. CONCLUSIONS This study enhances our understanding of the impact of estrogen loss on vascular health in a murine model and warrants further studies to examine the ability of LNS8801 to improve vascular health in menopausal women.
Collapse
Affiliation(s)
| | | | - Tristen Wong
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Bruna Visniauskas
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | | | - Chase Richard
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| | - Zaidmara Diaz
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Alec Horton
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | - Benard O. Ogola
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| |
Collapse
|
3
|
Mozzicato AM, Bastrup JA, Sanchez-Alonso JL, van der Horst J, Gorelik J, Hägglund P, Jepps TA. Mesenteric artery smooth muscle cells from hypertensive rats have increased microtubule acetylation. Biochem J 2024; 481:387-403. [PMID: 38373073 DOI: 10.1042/bcj20230420] [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: 10/03/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 02/21/2024]
Abstract
The dynamic nature of the microtubule network is dependent in part by post-translational modifications (PTMs) - particularly through acetylation, which stabilizes the microtubule network. Whether PTMs of the microtubule network in vascular smooth muscle cells (VSMCs) contribute to the pathophysiology of hypertension is unknown. The aim of this study was to determine the acetylated state of the microtubule network in the mesenteric arteries of spontaneously hypertensive rats (SHR). Experiments were performed on male normotensive rats and SHR mesenteric arteries. Western blotting and mass spectrometry determined changes in tubulin acetylation. Wire myography was used to investigate the effect of tubacin on isoprenaline-mediated vasorelaxations. Isolated cells from normotensive rats were used for scanning ion conductance microscopy (SICM). Mass spectrometry and Western blotting showed that tubulin acetylation is increased in the mesenteric arteries of the SHR compared with normotensive rats. Tubacin enhanced the β-adrenoceptor-mediated vasodilatation by isoprenaline when the endothelium was intact, but attenuated relaxations when the endothelium was denuded or nitric oxide production was inhibited. By pre-treating vessels with colchicine to disrupt the microtubule network, we were able to confirm that the effects of tubacin were microtubule-dependent. Using SICM, we examined the cell surface Young's modulus of VSMCs, but found no difference in control, tubacin-treated, or taxol-treated cells. Acetylation of tubulin at Lys40 is elevated in mesenteric arteries from the SHR. Furthermore, this study shows that tubacin has an endothelial-dependent bimodal effect on isoprenaline-mediated vasorelaxation.
Collapse
Affiliation(s)
- Anthony M Mozzicato
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joakim A Bastrup
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jose L Sanchez-Alonso
- Myocardial Function, National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, London, U.K
| | - Jennifer van der Horst
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Julia Gorelik
- Myocardial Function, National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, London, U.K
| | - Per Hägglund
- Protein Oxidation Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Tsilingiris D, Schimpfle L, Κender Z, Sulaj A, von Rauchhaupt E, Herzig S, Szendroedi J, Kopf S. Utility of bioelectrical phase angle for cardiovascular risk assessment among individuals with and without diabetes mellitus. Diab Vasc Dis Res 2024; 21:14791641231223701. [PMID: 38305220 PMCID: PMC10838040 DOI: 10.1177/14791641231223701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
PURPOSE Low values of bioimpedance-derived phase angle (PA) have been associated with various adverse outcomes. We investigated the association of PA with cardiovascular markers in individuals with and without diabetes mellitus (DM). METHODS PA was measured in 452 adults (without DM n = 153, T1DM n = 67, T2DM n = 232). Carotid intima-media thickness (IMT), renal resistive index (RRI), ankle-brachial index (ABI) and carotid-femoral Pulse Wave Velocity (cfPWV) were estimated. Furthermore, the levels of high-sensitive Troponin-T [hsTnT], N-terminal brain natriuretic peptide [NT-pro-BNP]) were measured. RESULTS PA values were lower in DM independently of age, gender, and BMI (estimated marginal means 6.21, 5.83, 5.95 for controls, T1DM, T2DM p < .05), a finding which persisted after propensity score matching. PA correlated negatively with IMT (r = -0.181), RRI (r = -0.374), cfPWV (r = -0.358), hsTnT (r = -0.238) and NT-pro-BNP (r = -0.318) (all p < .001). In multivariable analysis, the associations with RRI, cfPWV, hsTnT and NT-pro-BNP remained unchanged. PA values 6.0-6.5° for males and 5.2-5.8° for females were predictive of commonly used cutoffs. The combination of ΑCC/AHA ASCVD Score with PA outperformed either factor in predicting cfPWV, RRI for males and hsTnT, BNP for both genders. CONCLUSIONS PA exhibits independent correlations with various parameters pertinent to cardiovascular risk and may be useful for cardiovascular assessment.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- Department for Endocrinology, Diabetology, Metabolic diseases and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research(DZD), Munich-Neuherberg, Germany
| | - Lukas Schimpfle
- Department for Endocrinology, Diabetology, Metabolic diseases and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Zoltan Κender
- Department for Endocrinology, Diabetology, Metabolic diseases and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research(DZD), Munich-Neuherberg, Germany
| | - Alba Sulaj
- Department for Endocrinology, Diabetology, Metabolic diseases and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research(DZD), Munich-Neuherberg, Germany
| | - Ekaterina von Rauchhaupt
- Department for Endocrinology, Diabetology, Metabolic diseases and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research(DZD), Munich-Neuherberg, Germany
| | - Stephan Herzig
- German Center for Diabetes Research(DZD), Munich-Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Munich-Neuherberg, Germany
| | - Julia Szendroedi
- Department for Endocrinology, Diabetology, Metabolic diseases and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research(DZD), Munich-Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Stefan Kopf
- Department for Endocrinology, Diabetology, Metabolic diseases and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research(DZD), Munich-Neuherberg, Germany
| |
Collapse
|
5
|
Treadway J, Bielinski A, Zafiratos M, Polakowski J. Species comparison of compounds with known blood pressure effects in a vascular smooth muscle cell collagen contraction assay. J Pharmacol Toxicol Methods 2023; 123:107290. [PMID: 37442214 DOI: 10.1016/j.vascn.2023.107290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023]
Abstract
INTRODUCTION There is a great need for new approaches early in drug discovery that have the potential to improve clinical translation of compound-mediated cardiovascular effects. Current approaches frequently rely on in vivo animal models or in vitro tissue bath preparations, both of which are low throughput and costly. An in vitro surrogate screen for blood pressure using primary human cells may serve as a higher throughput method to quickly select compounds void of this secondary pharmacology and potentially improve late-stage drug development outcomes. METHODS In this study, we investigated 10 compounds with published in vivo blood pressure effects in a commercially available collagen contraction assay and evaluated rat, human, and canine (aortic) vascular smooth muscle cells (VSMCs). The aim of this study was to evaluate consistency between species and test their ability to predict the effects of known human vasodilators and constrictors. VSMCs were embedded at the same cell density in a collagen matrix which then floated freely in media containing test compounds. Collagen discs contracted faster than vehicle treated controls when incubated with a constrictor, and slower in the presence of a dilator. RESULTS Rat VSMCs responded as predicted of a VSMC-only culture to 9 out of 10 compounds. Human VSMCs responded as predicted to 8 out of 10 compounds, and canine VSMCs responded to 7 out of 10 compounds. DISCUSSION Our results suggest that rat VSMCs predict 90% of the effects of known vasoactive compounds in the collagen contraction assay while human and canine VSMCs were slightly less predictive (80% and 70%, respectively). Although blood pressure regulation is a multi-faceted and complex process, our data suggests the collagen smooth muscle contraction assay is useful as a qualitative early screen of compounds that act directly on smooth muscle cells of the arterial vasculature.
Collapse
Affiliation(s)
- Jessica Treadway
- Abbvie Inc, 1 North Waukegan Rd, North Chicago, IL 60064, United States of America.
| | - Aimee Bielinski
- Abbvie Inc, 1 North Waukegan Rd, North Chicago, IL 60064, United States of America
| | - Mark Zafiratos
- Abbvie Inc, 1 North Waukegan Rd, North Chicago, IL 60064, United States of America
| | - James Polakowski
- Abbvie Inc, 1 North Waukegan Rd, North Chicago, IL 60064, United States of America
| |
Collapse
|
6
|
Kilanowski-Doroh IM, McNally AB, Wong T, Visniauskas B, Blessinger SA, Imulinde Sugi A, Richard C, Diaz Z, Horton A, Natale CA, Ogola BO, Lindsey SH. Ovariectomy-Induced Arterial Stiffening Differs from Vascular Aging and is Reversed by GPER Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552881. [PMID: 37645992 PMCID: PMC10462036 DOI: 10.1101/2023.08.10.552881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Arterial stiffness is a cardiovascular risk factor and dramatically increases as women transition through menopause. The current study assessed whether a mouse model of menopause increases arterial stiffness in a similar manner to aging, and whether activation of the G protein-coupled estrogen receptor (GPER) could reverse stiffness. Female C57Bl/6J mice were ovariectomized (OVX) at 10 weeks of age or aged to 52 weeks, and some mice were treated with GPER agonists. OVX and aging increased pulse wave velocity to a similar extent independent of changes in blood pressure. Aging increased carotid wall thickness, while OVX increased material stiffness without altering vascular geometry. RNA-Seq analysis revealed that OVX downregulated smooth muscle contractile genes. The enantiomerically pure GPER agonist, LNS8801, reversed stiffness in OVX mice to a greater degree than the racemic agonist G-1. In summary, OVX and aging induced arterial stiffening via potentially different mechanisms. Aging was associated with inward remodeling while OVX induced material stiffness independent of geometry and a loss of the contractile phenotype. This study helps to further our understanding of the impact of menopause on vascular health and identifies LNS8801 as a potential therapy to counteract this detrimental process in women.
Collapse
|
7
|
Yang TC, Lu MH, Wang WJ, Chen JY. CEBPB/POU2F2 modulates endothelin 1 expression in prehypertensive SHR vascular smooth muscle cells. J Mol Endocrinol 2023; 71:e220178. [PMID: 36942826 PMCID: PMC10235928 DOI: 10.1530/jme-22-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/21/2023] [Indexed: 03/23/2023]
Abstract
The pathogenesis of hypertension is not fully understood; endothelin 1 (EDN1) is involved in developing essential hypertension. EDN1 can promote vascular smooth muscle cell (VSMC) proliferation or hypertrophy through autocrine and paracrine effects. Proliferating smooth muscle cells in the aorta are 'dedifferentiated' cells that cause increased arterial stiffness and remodeling. Male SHRs had higher aortic stiffness than normal control male WKY rats. Male SHR VSMCs expressed high levels of the EDN1 gene, but endothelial cells did not. Therefore, it is necessary to understand the molecular mechanism of enhanced EDN1 expression in SHR VSMCs. We identified POU2F2 and CEBPB as the main molecules that enhance EDN1 expression in male SHR VSMCs. A promoter activity analysis confirmed that the enhancer region of the Edn1 promoter in male SHR VSMCs was from -1309 to -1279 bp. POU2F2 and CEBPB exhibited an additive role in the enhancer region of the EdnET1 promoter. POU2F2 or CEBPB overexpression sufficiently increased EDN1 expression, and co-transfection with the CEBPB and POU2F2 expression plasmids had additive effects on the activity of the Edn1 promoter and EDN1 secretion level of male WKY VSMCs. In addition, the knockdown of POU2F2 also revealed that POU2F2 is necessary to enhance EDN1 expression in SHR VSMCs. The enhancer region of the Edn1 promoter is highly conserved in rats, mice, and humans. POU2F2 and CEBPB mRNA levels were significantly increased in remodeled human VMSCs. In conclusion, the novel regulation of POU2F2 and CEBPB in VSMCs will help us understand the pathogenesis of hypertension and support the development of future treatments for hypertension.
Collapse
Affiliation(s)
- Tien-Chun Yang
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hua Lu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Jie Wang
- Department of Entomology, University of California, Riverside, California, USA
| | - Jang-Yi Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
8
|
Pewowaruk RJ, Colebank MJ, Spronck B, Korcarz CE, Gepner AD. Biomechanics models predict increasing smooth muscle tone as a novel therapeutic target for central arterial dysfunction in hypertension. J Hypertens 2023; 41:572-579. [PMID: 36728092 PMCID: PMC9992290 DOI: 10.1097/hjh.0000000000003370] [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] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Vasodilation can paradoxically increase arterial stiffness in older, hypertensive adults. This study modeled increasing smooth muscle tone as a therapeutic strategy to improve central arterial dysfunction in hypertension using participant-specific simulations. METHODS Participant-specific models of the carotid artery were parameterized from vascular ultrasound measures of nitroglycerin-induced vasodilation in 18 hypertensive veterans. The acute changes in carotid artery mechanics were simulated for changes of ±2, ±4, and ±6% in smooth muscle tone and ±5, ±10, and ±15 mmHg in mean arterial pressure (MAP). The chronic carotid artery adaptations were simulated based on the hypothesis that the carotid artery will remodel wall-cross sectional area to maintain mechanical homeostasis. RESULTS A 6% increase in smooth muscle tone acutely decreased carotid pulse wave velocity from 6.89 ± 1.24 m/s to 5.83 ± 1.73 m/s, and a 15 mmHg decrease in MAP decreased carotid pulse wave velocity to 6.17 ± 1.23 m/s. A 6% increase in smooth muscle tone acutely decreased wall stress from 76.2 ± 12.3 to 64.2 ± 10.4 kPa, and a 15 mmHg decrease in MAP decreased wall stress to 60.6 ± 10.7 kPa. A 6% increase in smooth muscle tone chronically decreased wall cross-sectional area from 18.3 ± 5.4 to 15.2 ± 4.9 mm 2, and a 15 mmHg decrease in MAP decreased wall cross-sectional area to 14.3 ± 4.6 mm 2 . CONCLUSION In participant-specific simulation, increasing smooth muscle tone can have a stronger or equivalent effect on carotid artery mechanics compared with decreasing blood pressure. Increasing central arterial smooth muscle tone may be a novel therapeutic target to improve central arterial dysfunction in older, hypertensive adults and should be a focus of future research.
Collapse
Affiliation(s)
- Ryan J Pewowaruk
- William S. Middleton Memorial Veterans Hospital
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mitchel J Colebank
- Department of Biomedical Engineering, Irvine - Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, California, USA
| | - Bart Spronck
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Claudia E Korcarz
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Adam D Gepner
- William S. Middleton Memorial Veterans Hospital
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
9
|
Roberts E, Xu T, Assoian RK. Cell contractility and focal adhesion kinase control circumferential arterial stiffness. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:28-39. [PMID: 36222505 PMCID: PMC9782408 DOI: 10.1530/vb-22-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
Abstract
Arterial stiffening is a hallmark of aging and cardiovascular disease. While it is well established that vascular smooth muscle cells (SMCs) contribute to arterial stiffness by synthesizing and remodeling the arterial extracellular matrix, the direct contributions of SMC contractility and mechanosensors to arterial stiffness, and particularly the arterial response to pressure, remain less well understood despite being a long-standing question of biomedical importance. Here, we have examined this issue by combining the use of pressure myography of intact carotid arteries, pharmacologic inhibition of contractility, and genetic deletion of SMC focal adhesion kinase (FAK). Biaxial inflation-extension tests performed at physiological pressures showed that acute inhibition of cell contractility with blebbistatin or EGTA altered vessel geometry and preferentially reduced circumferential, as opposed to axial, arterial stiffness in wild-type mice. Similarly, genetic deletion of SMC FAK, which attenuated arterial contraction to KCl, reduced vessel wall thickness and circumferential arterial stiffness in response to pressure while having minimal effect on axial mechanics. Moreover, these effects of FAK deletion were lost by treating arteries with blebbistatin or by inhibiting myosin light-chain kinase. The expression of arterial fibrillar collagens, the integrity of arterial elastin, or markers of SMC differentiation were not affected by the deletion of SMC FAK. Our results connect cell contractility and SMC FAK to the regulation of arterial wall thickness and directionally specific arterial stiffening.
Collapse
Affiliation(s)
- Emilia Roberts
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tina Xu
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Richard K Assoian
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Connecting Aortic Stiffness to Vascular Contraction: Does Sex Matter? Int J Mol Sci 2022; 23:ijms231911314. [PMID: 36232616 PMCID: PMC9569837 DOI: 10.3390/ijms231911314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
This study was designed to connect aortic stiffness to vascular contraction in young male and female Wistar rats. We hypothesized that female animals display reduced intrinsic media-layer stiffness, which associates with improved vascular function. Atomic force microscopy (AFM)-based nanoindentation analysis was used to derive stiffness (Young’s modulus) in biaxially (i.e., longitudinal and circumferential) unloaded aortic rings. Reactivity studies compatible with uniaxial loading (i.e., circumferential) were used to assess vascular responses to a selective α1 adrenergic receptor agonist in the presence or absence of extracellular calcium. Elastin and collagen levels were indirectly evaluated with fluorescence microscopy and a picrosirius red staining kit, respectively. We report that male and female Wistar rats display similar AFM-derived aortic media-layer stiffness, even though female animals withstand higher aortic intima-media thickness-to-diameter ratio than males. Female animals also present reduced phenylephrine-induced aortic force development in concentration-response and time-force curves. Specifically, we observed impaired force displacement in both parts of the contraction curve (Aphasic and Atonic) in experiments conducted with and without extracellular calcium. Additionally, collagen levels were lower in female animals without significant elastin content and fragmentation changes. In summary, sex-related functional differences in isolated aortas appear to be related to dissimilarities in the dynamics of vascular reactivity and extracellular matrix composition rather than a direct response to a shift in intrinsic media-layer stiffness.
Collapse
|
11
|
How do cells stiffen? Biochem J 2022; 479:1825-1842. [PMID: 36094371 DOI: 10.1042/bcj20210806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Cell stiffness is an important characteristic of cells and their response to external stimuli. In this review, we survey methods used to measure cell stiffness, summarize stimuli that alter cell stiffness, and discuss signaling pathways and mechanisms that control cell stiffness. Several pathological states are characterized by changes in cell stiffness, suggesting this property can serve as a potential diagnostic marker or therapeutic target. Therefore, we consider the effect of cell stiffness on signaling and growth processes required for homeostasis and dysfunction in healthy and pathological states. Specifically, the composition and structure of the cell membrane and cytoskeleton are major determinants of cell stiffness, and studies have identified signaling pathways that affect cytoskeletal dynamics both directly and by altered gene expression. We present the results of studies interrogating the effects of biophysical and biochemical stimuli on the cytoskeleton and other cellular components and how these factors determine the stiffness of both individual cells and multicellular structures. Overall, these studies represent an intersection of the fields of polymer physics, protein biochemistry, and mechanics, and identify specific mechanisms involved in mediating cell stiffness that can serve as therapeutic targets.
Collapse
|
12
|
He Y, Qin C, Sun Z, Liu Z, Chen Y, Meng K. Atomic force microscopy application to study of the biomechanical properties of the aortic intima in the context of early atherosclerosis. Microsc Res Tech 2022; 85:3411-3417. [PMID: 35804436 DOI: 10.1002/jemt.24196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/11/2022]
Abstract
Atherosclerosis is characterized by the infiltration of macrophages, accumulation of lipids, activation of endothelial cells and synthesis of extracellular matrix by vascular smooth muscle cells. However, there have been few atomic force microscopy (AFM) studies of the aortic intima in situ in the context of atherosclerosis. By employing a customized liquid cell for AFM, we investigated the aortic intima obtained from male C57BL/6 ApoE-deficient mice (ApoE-/- ) aged 14 weeks and male C57BL/6 ApoE-sufficient mice (ApoE+/+ ) aged between 18 and 26 weeks that were fed a high-fat and high-cholesterol diet for 4 weeks and performed force spectroscopy mapping of the biomechanical properties of the intima. In the aortas of ApoE-deficient mice, the intima became stiffer than that of ApoE-sufficient mice. In addition, the cytoskeleton of endothelial cells was enlarged, and extracellular matrix accumulated. The biomechanical properties of the aortic intima are altered in early atherogenesis, which may be induced by the enlargement of the endothelial cell cytoskeleton and the increased synthesis of extracellular matrix by activated smooth muscle cells.
Collapse
Affiliation(s)
- Yin He
- Emergency Department, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Chuanyu Qin
- Department of Cardiology of Second Affiliated Hospital of Qiqihaer Medical Collage, Qiqihar, Heilongjiang, People's Republic of China
| | - Zhifu Sun
- Otolaryngology Head and Neck Surgery, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| | - Zesen Liu
- Department of Cardiology of Luhe Hospital, Capital Medical University, Beijing, China
| | - Yan Chen
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Kang Meng
- Department of Cardiology, Capital Medical University Affiliated Anzhen Hospital, Beijing, China
| |
Collapse
|
13
|
Achner L, Klersy T, Fels B, Reinberger T, Schmidt CX, Groß N, Hille S, Müller OJ, Aherrahrou Z, Kusche-Vihrog K, Raasch W. AFM-based nanoindentation indicates an impaired cortical stiffness in the AAV-PCSK9 DY atherosclerosis mouse model. Pflugers Arch 2022; 474:993-1002. [PMID: 35648220 PMCID: PMC9393126 DOI: 10.1007/s00424-022-02710-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/22/2022] [Indexed: 12/23/2022]
Abstract
Investigating atherosclerosis and endothelial dysfunction has mainly become established in genetically modified ApoE−/− or LDL-R−/− mice transgenic models. A new AAV-PCSK9DYDY mouse model with no genetic modification has now been reported as an alternative atherosclerosis model. Here, we aimed to employ this AAV-PCSK9DY mouse model to quantify the mechanical stiffness of the endothelial surface, an accepted hallmark for endothelial dysfunction and forerunner for atherosclerosis. Ten-week-old male C57BL/6 N mice were injected with AAV-PCSK9DY (0.5, 1 or 5 × 1011 VG) or saline as controls and fed with Western diet (1.25% cholesterol) for 3 months. Total cholesterol (TC) and triglycerides (TG) were measured after 6 and 12 weeks. Aortic sections were used for atomic force microscopy (AFM) measurements or histological analysis using Oil-Red-O staining. Mechanical properties of in situ endothelial cells derived from ex vivo aorta preparations were quantified using AFM-based nanoindentation. Compared to controls, an increase in plasma TC and TG and extent of atherosclerosis was demonstrated in all groups of mice in a viral load-dependent manner. Cortical stiffness of controls was 1.305 pN/nm and increased (10%) in response to viral load (≥ 0.5 × 1011 VG) and positively correlated with the aortic plaque content and plasma TC and TG. For the first time, we show changes in the mechanical properties of the endothelial surface and thus the development of endothelial dysfunction in the AAV-PCSK9DY mouse model. Our results demonstrate that this model is highly suitable and represents a good alternative to the commonly used transgenic mouse models for studying atherosclerosis and other vascular pathologies.
Collapse
Affiliation(s)
- Leonie Achner
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Tobias Klersy
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Benedikt Fels
- Institute for Physiology, University Lübeck, Lübeck, Germany
| | - Tobias Reinberger
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Institute for Cardiogenetics, University Lübeck, Lübeck, Germany
| | - Cosima X Schmidt
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Natalie Groß
- Institute for Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Susanne Hille
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Department of Internal Medicine III, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Oliver J Müller
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Department of Internal Medicine III, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Zouhair Aherrahrou
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Institute for Cardiogenetics, University Lübeck, Lübeck, Germany
| | - Kristina Kusche-Vihrog
- Institute for Physiology, University Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany. .,CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany.
| |
Collapse
|
14
|
Pierce GL, Coutinho TA, DuBose LE, Donato AJ. Is It Good to Have a Stiff Aorta with Aging? Causes and Consequences. Physiology (Bethesda) 2022; 37:154-173. [PMID: 34779281 PMCID: PMC8977146 DOI: 10.1152/physiol.00035.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/09/2023] Open
Abstract
Aortic stiffness increases with advancing age, more than doubling during the human life span, and is a robust predictor of cardiovascular disease (CVD) clinical events independent of traditional risk factors. The aorta increases in diameter and length to accommodate growing body size and cardiac output in youth, but in middle and older age the aorta continues to remodel to a larger diameter, thinning the pool of permanent elastin fibers, increasing intramural wall stress and resulting in the transfer of load bearing onto stiffer collagen fibers. Whereas aortic stiffening in early middle age may be a compensatory mechanism to normalize intramural wall stress and therefore theoretically "good" early in the life span, the negative clinical consequences of accelerated aortic stiffening beyond middle age far outweigh any earlier physiological benefit. Indeed, aortic stiffness and the loss of the "windkessel effect" with advancing age result in elevated pulsatile pressure and flow in downstream microvasculature that is associated with subclinical damage to high-flow, low-resistance organs such as brain, kidney, retina, and heart. The mechanisms of aortic stiffness include alterations in extracellular matrix proteins (collagen deposition, elastin fragmentation), increased arterial tone (oxidative stress and inflammation-related reduced vasodilators and augmented vasoconstrictors; enhanced sympathetic activity), arterial calcification, vascular smooth muscle cell stiffness, and extracellular matrix glycosaminoglycans. Given the rapidly aging population of the United States, aortic stiffening will likely contribute to substantial CVD burden over the next 2-3 decades unless new therapeutic targets and interventions are identified to prevent the potential avalanche of clinical sequelae related to age-related aortic stiffness.
Collapse
Affiliation(s)
- Gary L Pierce
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa
| | - Thais A Coutinho
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Divisions of Cardiology and Cardiac Prevention and Rehabilitation, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Lyndsey E DuBose
- Division of Geriatrics, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony J Donato
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
- Department of Biochemistry, University of Utah, Salt Lake City, Utah
- Geriatric Research Education and Clinical Center, VA Salt Lake City, Salt Lake City, Utah
| |
Collapse
|
15
|
Tuttle T, Darios E, Watts SW, Roccabianca S. Aortic stiffness is lower when perivascular adipose tissue (PVAT) is included: a novel ex vivo mechanics study. Am J Physiol Heart Circ Physiol 2022; 322:H1003-H1013. [PMID: 35275760 DOI: 10.1152/ajpheart.00574.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Perivascular adipose tissue (PVAT) is increasingly recognized as an essential layer of the functional vasculature, being responsible for producing vasoactive substances and assisting arterial stress relaxation. Here we test the hypothesis that PVAT reduces aortic stiffness. Our model was the thoracic aorta of the male Sprague Dawley rat. Uniaxial mechanical tests for three groups of tissue were performed: aorta +PVAT (+PVAT), aorta - PVAT (-PVAT), and isolated PVAT (PVAT only). The output of the mechanical test is reported in the form of a Cauchy stress-stretch curve. This work presents a novel, physiologically relevant approach to measure mechanical stiffness ex vivo in isolated PVAT. Low-stress stiffness (), high-stress stiffness (), and the stress corresponding to a stretch of 1.2 () were measured as metrics of distensibility. The low-stress stiffness was largest in the -PVAT samples and smallest in PVAT only samples. Both the high-stress stiffness and the stress at 1.2 stretch were significantly higher in -PVAT samples when compared to +PVAT samples. Taken together these results suggest that -PVAT samples are stiffer (less distensible) both at low stress (not significant) as well as at high stress (significant) when compared to +PVAT samples. These conclusions are supported by the results of the continuum mechanics material model we also used to interpret the same experimental data. Thus, tissue stiffness is significantly lower when considering PVAT as part of the aortic wall. As such, PVAT should be considered as a target for improving vascular function in diseases with elevated aortic stiffness, including hypertension.
Collapse
Affiliation(s)
- Tyler Tuttle
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| | - Emma Darios
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Sara Roccabianca
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
16
|
Pewowaruk RJ, Gepner AD. Smooth muscle tone alters arterial stiffness: the importance of the extracellular matrix to vascular smooth muscle stiffness ratio. J Hypertens 2022; 40:512-519. [PMID: 34751172 PMCID: PMC8810580 DOI: 10.1097/hjh.0000000000003039] [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] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Recent studies show that vascular smooth muscle (VSM) is more important to elastic artery mechanics than previously believed. It remains unclear whether increased VSM tone increases or decreases arterial stiffness. METHODS AND RESULTS We developed a novel arterial mechanics model based on pressure-diameter relationships that incorporates the contributions of extracellular matrix (ECM) and VSM to arterial stiffness measures. This model is advantageous because it simple enough to use with limited clinical data but has biologically relevant parameters which include ECM stiffness, VSM stiffness, and VSM tone. The model was used to retrospectively analyze the effects of nitroglycerin-induced vasodilation in four clinical studies. Stiffness parameters were modeled for five arterial regions including both elastic and muscular arteries. The model describes complex experimental data with changing VSM tone and blood pressure. Our analysis found that when ECM is less stiff than VSM, increasing VSM tone increases arterial stiffness. The opposite is seen when ECM is stiffer than VSM, increasing VSM tone decreases stiffness. Our results also suggest that VSM tone is a compensatory mechanism for elevated ECM stiffness in hypertensive individuals. CONCLUSION Based on retrospective analysis of four clinical studies, we propose a simple hypothesis for the role of VSM tone on arterial stiffness: increased VSM tone increases arterial stiffness when VSM is stiffer than ECM and decreases arterial stiffness when ECM is stiffer than VSM. This hypothesis and the methods used in this study could have important implications for understanding arterial physiology in both hypertension and cardiovascular disease and deserve further exploration.
Collapse
Affiliation(s)
- Ryan J Pewowaruk
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Adam D Gepner
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| |
Collapse
|
17
|
Mammoto A, Matus K, Mammoto T. Extracellular Matrix in Aging Aorta. Front Cell Dev Biol 2022; 10:822561. [PMID: 35265616 PMCID: PMC8898904 DOI: 10.3389/fcell.2022.822561] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/07/2022] [Indexed: 12/11/2022] Open
Abstract
The aging population is booming all over the world and arterial aging causes various age-associated pathologies such as cardiovascular diseases (CVDs). The aorta is the largest elastic artery, and transforms pulsatile flow generated by the left ventricle into steady flow to maintain circulation in distal tissues and organs. Age-associated structural and functional changes in the aortic wall such as dilation, tortuousness, stiffening and losing elasticity hamper stable peripheral circulation, lead to tissue and organ dysfunctions in aged people. The extracellular matrix (ECM) is a three-dimensional network of macromolecules produced by resident cells. The composition and organization of key ECM components determine the structure-function relationships of the aorta and therefore maintaining their homeostasis is critical for a healthy performance. Age-associated remodeling of the ECM structural components, including fragmentation of elastic fibers and excessive deposition and crosslinking of collagens, is a hallmark of aging and leads to functional stiffening of the aorta. In this mini review, we discuss age-associated alterations of the ECM in the aortic wall and shed light on how understanding the mechanisms of aortic aging can lead to the development of efficient strategy for aortic pathologies and CVDs.
Collapse
Affiliation(s)
- Akiko Mammoto
- Department of Pediatrics, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Milwaukee, WI, United States
- *Correspondence: Akiko Mammoto, ; Tadanori Mammoto,
| | - Kienna Matus
- Department of Pediatrics, Milwaukee, WI, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
- *Correspondence: Akiko Mammoto, ; Tadanori Mammoto,
| |
Collapse
|
18
|
Ramirez-Perez FI, Cabral-Amador FJ, Whaley-Connell AT, Aroor AR, Morales-Quinones M, Woodford ML, Ghiarone T, Ferreira-Santos L, Jurrissen TJ, Manrique-Acevedo CM, Jia G, DeMarco VG, Padilla J, Martinez-Lemus LA, Lastra G. Cystamine reduces vascular stiffness in Western diet-fed female mice. Am J Physiol Heart Circ Physiol 2022; 322:H167-H180. [PMID: 34890280 PMCID: PMC8742720 DOI: 10.1152/ajpheart.00431.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Consumption of diets high in fat, sugar, and salt (Western diet, WD) is associated with accelerated arterial stiffening, a major independent risk factor for cardiovascular disease (CVD). Women with obesity are more prone to develop arterial stiffening leading to more frequent and severe CVD compared with men. As tissue transglutaminase (TG2) has been implicated in vascular stiffening, our goal herein was to determine the efficacy of cystamine, a nonspecific TG2 inhibitor, at reducing vascular stiffness in female mice chronically fed a WD. Three experimental groups of female mice were created. One was fed regular chow diet (CD) for 43 wk starting at 4 wk of age. The second was fed a WD for the same 43 wk, whereas a third cohort was fed WD, but also received cystamine (216 mg/kg/day) in the drinking water during the last 8 wk on the diet (WD + C). All vascular stiffness parameters assessed, including aortic pulse wave velocity and the incremental modulus of elasticity of isolated femoral and mesenteric arteries, were significantly increased in WD- versus CD-fed mice, and reduced in WD + C versus WD-fed mice. These changes coincided with respectively augmented and diminished vascular wall collagen and F-actin content, with no associated effect in blood pressure. In cultured human vascular smooth muscle cells, cystamine reduced TG2 activity, F-actin:G-actin ratio, collagen compaction capacity, and cellular stiffness. We conclude that cystamine treatment represents an effective approach to reduce vascular stiffness in female mice in the setting of WD consumption, likely because of its TG2 inhibitory capacity.NEW & NOTEWORTHY This study evaluates the novel role of transglutaminase 2 (TG2) inhibition to directly treat vascular stiffness. Our data demonstrate that cystamine, a nonspecific TG2 inhibitor, improves vascular stiffness induced by a diet rich in fat, fructose, and salt. This research suggests that TG2 inhibition might bear therapeutic potential to reduce the disproportionate burden of cardiovascular disease in females in conditions of chronic overnutrition.
Collapse
Affiliation(s)
- Francisco I. Ramirez-Perez
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Biomedical, Biological, and Chemical Engineering Department, University of Missouri, Columbia, Missouri
| | | | - Adam T. Whaley-Connell
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,4Division of Nephrology and Hypertension, Department of Medicine, University of Missouri, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | - Annayya R. Aroor
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | | | - Makenzie L. Woodford
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Thaysa Ghiarone
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Larissa Ferreira-Santos
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,6Instituto do Coracao, Hospital das Clínicas da Faculdade de
Medicina da Universidade de São Paulo, Faculdade de Medicina, Universidade
de São Paulo, São Paulo, Brazil
| | - Thomas J. Jurrissen
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,7Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Camila M. Manrique-Acevedo
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | - GuangHong Jia
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | - Vincent G. DeMarco
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,4Division of Nephrology and Hypertension, Department of Medicine, University of Missouri, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri,8Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,7Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Luis A. Martinez-Lemus
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Biomedical, Biological, and Chemical Engineering Department, University of Missouri, Columbia, Missouri,8Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Guido Lastra
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
19
|
Petit C, Karkhaneh Yousefi AA, Guilbot M, Barnier V, Avril S. AFM Stiffness Mapping in Human Aortic Smooth Muscle Cells. J Biomech Eng 2022; 144:1133331. [DOI: 10.1115/1.4053657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Indexed: 11/08/2022]
Abstract
Abstract
Aortic Smooth Muscle Cells (SMCs) play a vital role in maintaining mechanical homeostasis in the aorta. We recently found that SMCs of aneurysmal aortas apply larger traction forces than SMCs of healthy aortas. This result was explained by the significant increase of hypertrophic SMCs abundance in aneurysms. In the present study, we investigate whether the cytoskeleton stiffness of SMCs may also be altered in aneurysmal aortas. For that, we use Atomic Force Microscopy (AFM) nanoindentation with a specific mode that allows subcellular-resolution mapping of the local stiffness across a specified region of interest of the cell. Aortic SMCs from a commercial human lineage (AoSMCs, Lonza) and primary aneurysmal SMCs (AnevSMCs) are cultured in conditions promoting the development of their contractile apparatus, and seeded on hydrogels with stiffness properties of 12kPa and 25kPa. Results show that all SMC exhibit globally a lognormal stiffness distribution, with medians in the range 10-30 kPa. The mean of stiffness distributions is slightly higher in aneurysmal SMCs than in healthy cells (16 kPa versus 12 kPa) but the differences are not statistically significant due to the large dispersion of AFM nanoindentation stiffness. We conclude that the possible alterations previously found in aneurysmal SMCs do not affect significantly the AFM nanoindentation stiffness of their cytoskeleton.
Collapse
Affiliation(s)
- Claudie Petit
- Mines Saint-Etienne, Université de Lyon, INSERM, U 1059 SAINBIOSE, F - 42023 Saint-Etienne France
| | | | - Marine Guilbot
- Mines Saint-Etienne, Université de Lyon, INSERM, U 1059 SAINBIOSE, F - 42023 Saint-Etienne France
| | - Vincent Barnier
- Mines Saint-Etienne, Université de Lyon, CNRS, UMR 5307 LGF, F - 42023 Saint-Etienne France
| | - Stephane Avril
- Mines Saint-Etienne, Université de Lyon, INSERM, U 1059 SAINBIOSE, F - 42023 Saint-Etienne France
| |
Collapse
|
20
|
Vatner SF, Zhang J, Vyzas C, Mishra K, Graham RM, Vatner DE. Vascular Stiffness in Aging and Disease. Front Physiol 2021; 12:762437. [PMID: 34950048 PMCID: PMC8688960 DOI: 10.3389/fphys.2021.762437] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/26/2021] [Indexed: 01/01/2023] Open
Abstract
The goal of this review is to provide further understanding of increased vascular stiffness with aging, and how it contributes to the adverse effects of major human diseases. Differences in stiffness down the aortic tree are discussed, a topic requiring further research, because most prior work only examined one location in the aorta. It is also important to understand the divergent effects of increased aortic stiffness between males and females, principally due to the protective role of female sex hormones prior to menopause. Another goal is to review human and non-human primate data and contrast them with data in rodents. This is particularly important for understanding sex differences in vascular stiffness with aging as well as the changes in vascular stiffness before and after menopause in females, as this is controversial. This area of research necessitates studies in humans and non-human primates, since rodents do not go through menopause. The most important mechanism studied as a cause of age-related increases in vascular stiffness is an alteration in the vascular extracellular matrix resulting from an increase in collagen and decrease in elastin. However, there are other mechanisms mediating increased vascular stiffness, such as collagen and elastin disarray, calcium deposition, endothelial dysfunction, and the number of vascular smooth muscle cells (VSMCs). Populations with increased longevity, who live in areas called “Blue Zones,” are also discussed as they provide additional insights into mechanisms that protect against age-related increases in vascular stiffness. Such increases in vascular stiffness are important in mediating the adverse effects of major cardiovascular diseases, including atherosclerosis, hypertension and diabetes, but require further research into their mechanisms and treatment.
Collapse
Affiliation(s)
- Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, Rutgers University - New Jersey Medical School, Newark, NJ, United States
| | - Jie Zhang
- Department of Cell Biology and Molecular Medicine, Rutgers University - New Jersey Medical School, Newark, NJ, United States
| | - Christina Vyzas
- Department of Cell Biology and Molecular Medicine, Rutgers University - New Jersey Medical School, Newark, NJ, United States
| | - Kalee Mishra
- Department of Cell Biology and Molecular Medicine, Rutgers University - New Jersey Medical School, Newark, NJ, United States
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, University of New South Wales, Darlinghurst, NSW, Australia
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, Rutgers University - New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
21
|
González-Blázquez R, Alcalá M, Cárdenas-Rebollo JM, Viana M, Steckelings UM, Boisvert WA, Unger T, Fernández-Alfonso MS, Somoza B, Gil-Ortega M. AT2R stimulation with C21 prevents arterial stiffening and endothelial dysfunction in the abdominal aorta from mice fed a high-fat diet. Clin Sci (Lond) 2021; 135:2763-2780. [PMID: 34854902 DOI: 10.1042/cs20210971] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to evaluate the effect of Compound 21 (C21), a selective AT2R agonist, on the prevention of endothelial dysfunction, extracellular matrix (ECM) remodeling and arterial stiffness associated with diet-induced obesity (DIO). Five-week-old male C57BL/6J mice were fed a standard (Chow) or high-fat diet (HF) for 6 weeks. Half of the animals of each group were simultaneously treated with C21 (1 mg/kg/day, in the drinking water), generating four groups: Chow C, Chow C21, HF C, and HF C21. Vascular function and mechanical properties were determined in the abdominal aorta. To evaluate ECM remodeling, collagen deposition and TGF-β1 concentrations were determined in the abdominal aorta and the activity of metalloproteinases (MMP) 2 and 9 was analyzed in the plasma. Abdominal aortas from HF C mice showed endothelial dysfunction as well as enhanced contractile but reduced relaxant responses to Ang II. This effect was abrogated with C21 treatment by preserving NO availability. A left-shift in the tension-stretch relationship, paralleled by an augmented β-index (marker of intrinsic arterial stiffness), and enhanced collagen deposition and MMP-2/-9 activities were also detected in HF mice. However, when treated with C21, HF mice exhibited lower TGF-β1 levels in abdominal aortas together with reduced MMP activities and collagen deposition compared with HF C mice. In conclusion, these data demonstrate that AT2R stimulation by C21 in obesity preserves NO availability and prevents unhealthy vascular remodeling, thus protecting the abdominal aorta in HF mice against the development of endothelial dysfunction, ECM remodeling and arterial stiffness.
Collapse
Affiliation(s)
- Raquel González-Blázquez
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Martín Alcalá
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, 28925, Madrid, Spain
| | - José Miguel Cárdenas-Rebollo
- Departamento de Matemática Aplicada y Estadística. Facultad de Ciencias Económicas y Empresariales. Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Marta Viana
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, 28925, Madrid, Spain
| | - Ulrike Muscha Steckelings
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, BSB311, Honolulu, HI 96813, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Str., Kazan 420008, Russia
| | - Thomas Unger
- CARIM - School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - María S Fernández-Alfonso
- Instituto Pluridisciplinar, Unidad de Cartografía Cerebral, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| |
Collapse
|
22
|
Kamyab R, Namdar H, Torbati M, Ghojazadeh M, Araj-Khodaei M, Fazljou SMB. Medicinal Plants in the Treatment of Hypertension: A Review. Adv Pharm Bull 2021; 11:601-617. [PMID: 34888207 PMCID: PMC8642800 DOI: 10.34172/apb.2021.090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 11/09/2022] Open
Abstract
Traditional medicine is a comprehensive term for ancient, culture-bound health care practices that existed before the use of science in health matters and has been used for centuries. Medicinal plants are used to treat patients with cardiovascular diseases, which may occur due to ailments of the heart and blood vessels and comprise heart attacks, cerebrovascular diseases, hypertension, and heart failure. Hypertension causes difficulty in the functioning of the heart and is involved in atherosclerosis, raising the risk of heart attack and stroke. Many drugs are available for managing these diseases, though common antihypertensive drugs are generally accompanied by many side effects. Medicinal herbs have several active substances with pharmacological and prophylactic properties that can be used in the treatment of hypertension. This review presents an overview of some medicinal plants that have been shown to have hypotensive or antihypertensive properties.
Collapse
Affiliation(s)
- Raha Kamyab
- Department of Persian Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Namdar
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadali Torbati
- Department of Food Science and Technology, Faculty of Nutrition, Tabriz University of Medical Science, Tabriz, Iran
| | - Morteza Ghojazadeh
- Research Center for Evidence Based Medicine (RCEBM), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Araj-Khodaei
- Department of Persian Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
23
|
Längst N, Adler J, Schweigert O, Kleusberg F, Cruz Santos M, Knauer A, Sausbier M, Zeller T, Ruth P, Lukowski R. Cyclic GMP-Dependent Regulation of Vascular Tone and Blood Pressure Involves Cysteine-Rich LIM-Only Protein 4 (CRP4). Int J Mol Sci 2021; 22:9925. [PMID: 34576086 PMCID: PMC8466836 DOI: 10.3390/ijms22189925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/09/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023] Open
Abstract
The cysteine-rich LIM-only protein 4 (CRP4), a LIM-domain and zinc finger containing adapter protein, has been implicated as a downstream effector of the second messenger 3',5'-cyclic guanosine monophosphate (cGMP) pathway in multiple cell types, including vascular smooth muscle cells (VSMCs). VSMCs and nitric oxide (NO)-induced cGMP signaling through cGMP-dependent protein kinase type I (cGKI) play fundamental roles in the physiological regulation of vascular tone and arterial blood pressure (BP). However, it remains unclear whether the vasorelaxant actions attributed to the NO/cGMP axis require CRP4. This study uses mice with a targeted deletion of the CRP4 gene (CRP4 KO) to elucidate whether cGMP-elevating agents, which are well known for their vasorelaxant properties, affect vessel tone, and thus, BP through CRP4. Cinaciguat, a NO- and heme-independent activator of the NO-sensitive (soluble) guanylyl cyclase (NO-GC) and NO-releasing agents, relaxed both CRP4-proficient and -deficient aortic ring segments pre-contracted with prostaglandin F2α. However, the magnitude of relaxation was slightly, but significantly, increased in vessels lacking CRP4. Accordingly, CRP4 KO mice presented with hypotonia at baseline, as well as a greater drop in systolic BP in response to the acute administration of cinaciguat, sodium nitroprusside, and carbachol. Mechanistically, loss of CRP4 in VSMCs reduced the Ca2+-sensitivity of the contractile apparatus, possibly involving regulatory proteins, such as myosin phosphatase targeting subunit 1 (MYPT1) and the regulatory light chain of myosin (RLC). In conclusion, the present findings confirm that the adapter protein CRP4 interacts with the NO-GC/cGMP/cGKI pathway in the vasculature. CRP4 seems to be part of a negative feedback loop that eventually fine-tunes the NO-GC/cGMP axis in VSMCs to increase myofilament Ca2+ desensitization and thereby the maximal vasorelaxant effects attained by (selected) cGMP-elevating agents.
Collapse
Affiliation(s)
- Natalie Längst
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Julia Adler
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Olga Schweigert
- Cardiovascular Systems Medicine and Molecular Translation, University Center of Cardiovascular Science, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (O.S.); (T.Z.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Felicia Kleusberg
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Amelie Knauer
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Matthias Sausbier
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Tanja Zeller
- Cardiovascular Systems Medicine and Molecular Translation, University Center of Cardiovascular Science, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (O.S.); (T.Z.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| |
Collapse
|
24
|
Kajuluri LP, Singh K, Morgan KG. Vascular aging, the vascular cytoskeleton and aortic stiffness. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:186-197. [PMID: 34414394 PMCID: PMC8372409 DOI: 10.37349/emed.2021.00041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vascular aging, aortic stiffness and hypertension are mechanistically interrelated. The perspective presented here will focus mainly on the molecular mechanisms of age-associated increases in the stiffness of the vascular smooth muscle cell (VSMC). This review will highlight the mechanisms by which the VSMC contributes to disorders of vascular aging. Distinct functional sub-components of the vascular cell and the molecular mechanisms of the protein-protein interactions, signaling mechanisms and intracellular trafficking processes in the setting of the aging aorta will be detailed.
Collapse
Affiliation(s)
| | - Kuldeep Singh
- Department of Health Sciences, Boston University, Boston, MA 02215, USA.,CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India
| | - Kathleen G Morgan
- Department of Health Sciences, Boston University, Boston, MA 02215, USA
| |
Collapse
|
25
|
Gade JL, Thore CJ, Sonesson B, Stålhand J. In vivo parameter identification in arteries considering multiple levels of smooth muscle activity. Biomech Model Mechanobiol 2021; 20:1547-1559. [PMID: 33934232 PMCID: PMC8298368 DOI: 10.1007/s10237-021-01462-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/16/2021] [Indexed: 11/25/2022]
Abstract
In this paper an existing in vivo parameter identification method for arteries is extended to account for smooth muscle activity. Within this method a continuum-mechanical model, whose parameters relate to the mechanical properties of the artery, is fit to clinical data by solving a minimization problem. Including smooth muscle activity in the model increases the number of parameters. This may lead to overparameterization, implying that several parameter combinations solve the minimization problem equally well and it is therefore not possible to determine which set of parameters represents the mechanical properties of the artery best. To prevent overparameterization the model is fit to clinical data measured at different levels of smooth muscle activity. Three conditions are considered for the human abdominal aorta: basal during rest; constricted, induced by lower-body negative pressure; and dilated, induced by physical exercise. By fitting the model to these three arterial conditions simultaneously a unique set of model parameters is identified and the model prediction agrees well with the clinical data.
Collapse
Affiliation(s)
- Jan-Lucas Gade
- Department of Management and Engineering, Division of Solid Mechanics, Linköping University, Linköping, Sweden.
| | - Carl-Johan Thore
- Department of Management and Engineering, Division of Solid Mechanics, Linköping University, Linköping, Sweden
| | - Björn Sonesson
- Department of Cardiothoracic and Vascular Surgery, Skåne University Hospital, Malmö, Sweden
| | - Jonas Stålhand
- Department of Management and Engineering, Division of Solid Mechanics, Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Ogola BO, Clark GL, Abshire CM, Harris NR, Gentry KL, Gunda SS, Kilanowski-Doroh I, Wong TJ, Visniauskas B, Lawrence DJ, Zimmerman MA, Bayer CL, Groban L, Miller KS, Lindsey SH. Sex and the G Protein-Coupled Estrogen Receptor Impact Vascular Stiffness. Hypertension 2021; 78:e1-e14. [PMID: 34024124 PMCID: PMC8192475 DOI: 10.1161/hypertensionaha.120.16915] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Benard O. Ogola
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | - Gabrielle L. Clark
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | - Caleb M. Abshire
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | | | - Kaylee L. Gentry
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | - Shreya S. Gunda
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | | | - Tristen J. Wong
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | | | - Dylan J. Lawrence
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | | | - Carolyn L. Bayer
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kristin S. Miller
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | - Sarah H. Lindsey
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| |
Collapse
|
27
|
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: 4.7] [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
|
28
|
Varela R, Rauschert I, Romanelli G, Alberro A, Benech JC. Hyperglycemia and hyperlipidemia can induce morphophysiological changes in rat cardiac cell line. Biochem Biophys Rep 2021; 26:100983. [PMID: 33912691 PMCID: PMC8063753 DOI: 10.1016/j.bbrep.2021.100983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/04/2021] [Accepted: 03/08/2021] [Indexed: 01/04/2023] Open
Abstract
H9c2 cardiac cells were incubated under the control condition and at different hyperglycemic and hyperlipidemic media, and the following parameters were determined and quantified: a) cell death, b) type of cell death, and c) changes in cell length, width and height. Of all the proven media, the one that showed the greatest differences compared to the control was the medium glucose (G) 33 mM + 500 μM palmitic acid. This condition was called the hyperglycemic and hyperlipidemic condition (HHC). Incubation of H9c2 cells in HHC promoted 5.2 times greater total cell death when compared to the control. Of the total death ofthe HHC cells, 38.6% was late apoptotic and 8.3% early apoptotic. HHC also changes cell morphology. The reordering of the actin cytoskeleton and cell stiffness was also studied in control and HHC cells. The actin cytoskeleton was quantified and the number and distance of actin bundles were not the same in the control as under HHC. Young's modulus images show a map of cell stiffness. Cells incubated in HHC with the reordered actin cytoskeleton were stiffer than those incubated in control. The region of greatest stiffness was the peripheral zone of HHC cells (where the number of actin bundles was higher and the distance between them smaller). Our results suggest a correlation between the reordering of the actin cytoskeleton and cell stiffness. Thus, our study showed that HHC can promote morphophysiological changes in rat cardiac cells confirming that gluco-and lipotoxicity may play a central role in the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Rocío Varela
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Inés Rauschert
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay.,Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Gerardo Romanelli
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Andrés Alberro
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Juan C Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay.,Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| |
Collapse
|
29
|
Abstract
Significance: The vascular extracellular matrix (ECM) not only provides mechanical stability but also manipulates vascular cell behaviors, which are crucial for vascular function and homeostasis. ECM remodeling, which alters vascular wall mechanical properties and exposes vascular cells to bioactive molecules, is involved in the development and progression of hypertension. Recent Advances: This brief review summarized the dynamic changes in ECM components and their modification and degradation during hypertension and after antihypertensive treatment. We also discussed how alterations in the ECM amount, assembly, mechanical properties, and degradation fragment generation provide input into the pathological process of hypertension. Critical Issues: Although the relevance between ECM remodeling and hypertension has been recognized, the underlying mechanism by which ECM remodeling initiates the development of hypertension remains unclear. Therefore, the modulation of ECM remodeling on arterial stiffness and hypertension in genetically modified rodent models is summarized in this review. The circulating biomarkers based on ECM metabolism and therapeutic strategies targeting ECM disorders in hypertension are also introduced. Future Directions: Further research will provide more comprehensive understanding of ECM remodeling in hypertension by the application of matridomic and degradomic approaches. The better understanding of mechanisms underlying vascular ECM remodeling may provide novel potential therapeutic strategies for preventing and treating hypertension. Antioxid. Redox Signal. 34, 765-783.
Collapse
Affiliation(s)
- Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
30
|
HIF2A gain-of-function mutation modulates the stiffness of smooth muscle cells and compromises vascular mechanics. iScience 2021; 24:102246. [PMID: 33796838 PMCID: PMC7995528 DOI: 10.1016/j.isci.2021.102246] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/26/2020] [Accepted: 02/25/2021] [Indexed: 02/08/2023] Open
Abstract
Heterozygous gain-of-function (GOF) mutations of hypoxia-inducible factor 2α (HIF2A), a key hypoxia-sensing regulator, are associated with erythrocytosis, thrombosis, and vascular complications that account for morbidity and mortality of patients. We demonstrated that the vascular pathology of HIF2A GOF mutations is independent of erythrocytosis. We generated HIF2A GOF-induced pluripotent stem cells (iPSCs) and differentiated them into endothelial cells (ECs) and smooth muscle cells (SMCs). Unexpectedly, HIF2A-SMCs, but not HIF2A-ECs, were phenotypically aberrant, more contractile, stiffer, and overexpressed endothelin 1 (EDN1), myosin heavy chain, elastin, and fibrillin. EDN1 inhibition and knockdown of EDN1-receptors both reduced HIF2-SMC stiffness. Hif2A GOF heterozygous mice displayed pulmonary hypertension, had SMCs with more disorganized stress fibers and higher stiffness in their pulmonary arterial smooth muscle cells, and had more deformable pulmonary arteries compared with wild-type mice. Our findings suggest that targeting these vascular aberrations could benefit patients with HIF2A GOF and conditions of augmented hypoxia signaling. HIF2-SMCs are stiffer than WT-SMCs and differ in contractile SMC marker expression HIF2-SMCs and WT-SMCs differ in EDN1 production and ECM composition HIF- 2α induces EDN1; EDNI subsequently induces SMC stiffening Hif2A GOF mouse arterial SMCs have more disorganized stress fibers and are stiffer
Collapse
|
31
|
McCallinhart PE, Scandling BW, Trask AJ. Coronary remodeling and biomechanics: Are we going with the flow in 2020? Am J Physiol Heart Circ Physiol 2020; 320:H584-H592. [PMID: 33185115 DOI: 10.1152/ajpheart.00634.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Under normal conditions, coronary blood flow (CBF) provides critical blood supply to the myocardium so that it can appropriately meet the metabolic demands of the body. Dogmatically, there exist several known regulators and modulators of CBF that include local metabolites and neurohormonal factors that can influence the function of the coronary circulation. In disease states such as diabetes and myocardial ischemia, these regulators are impaired or shifted such that CBF is reduced. Although functional considerations have been and continued to be well studied, more recent evidence builds upon established studies that collectively suggest that the relative roles of coronary structure, biomechanics, and the influence of cardiac biomechanics via extravascular compression may also play a significant role in dictating CBF. In this mini review, we discuss these regulators of CBF under normal and pathophysiological conditions and their potential influence on the control of CBF.
Collapse
Affiliation(s)
- Patricia E McCallinhart
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
| | - Benjamin W Scandling
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio.,Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Aaron J Trask
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
32
|
DAPT, a potent Notch inhibitor regresses actively growing abdominal aortic aneurysm via divergent pathways. Clin Sci (Lond) 2020; 134:1555-1572. [PMID: 32490531 DOI: 10.1042/cs20200456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a localized pathological dilation of the aorta exceeding the normal diameter (∼20 mm) by more than 50% of its original size (≥30 mm), accounting for approximately 150000-200000 deaths worldwide per year. We previously reported that Notch inhibition does not decrease the size of pre-established AAA at late stage of the disease. Here, we examined whether a potent pharmacologic inhibitor of Notch signaling (DAPT (N-[N-(3,5-Difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester)), regresses an actively growing AAA. In a mouse model of an aneurysm (Apoe-/- mice; n=44); DAPT (n=17) or vehicle (n=17) was randomly administered at day 14 of angiotensin II (AngII; 1 µg/min/kg), three times a week and mice were killed on day 42. Progressive increase in aortic stiffness and maximal intraluminal diameter (MILD) was observed in the AngII + vehicle group, which was significantly prevented by DAPT (P<0.01). The regression of aneurysm with DAPT was associated with reduced F4/80+Cd68+ (cluster of differentiation 68) inflammatory macrophages. DAPT improved structural integrity of aorta by reducing collagen fibrils abnormality and restoring their diameter. Mechanistically, C-C chemokine receptor type 7 (Ccr7)+F4/80- dendritic cells (DCs), implicated in the regression of aneurysm, were increased in the aorta of DAPT-treated mice. In the macrophages stimulated with AngII or lipopolysaccharide (LPS), DAPT reverted the expression of pro-inflammatory genes Il6 and Il12 back to baseline within 6 h compared with vehicle (P<0.05). DAPT also significantly increased the expression of anti-inflammatory genes, including c-Myc, Egr2, and Arg1 at 12-24 h in the LPS-stimulated macrophages (P<0.05). Overall, these regressive effects of Notch signaling inhibitor emphasize its therapeutic implications to prevent the progression of active AAAs.
Collapse
|
33
|
Sanyour HJ, Rickel AP, Hong Z. The interplay of membrane cholesterol and substrate on vascular smooth muscle biomechanics. CURRENT TOPICS IN MEMBRANES 2020; 86:279-299. [PMID: 33837696 PMCID: PMC8041049 DOI: 10.1016/bs.ctm.2020.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
Cardiovascular disease (CVD) remains the primary cause of death worldwide. Specifically, atherosclerosis is a CVD characterized as a slow progressing chronic inflammatory disease. During atherosclerosis, vascular walls accumulate cholesterol and cause fatty streak formation. The progressive changes in vascular wall stiffness exert alternating mechanical cues on vascular smooth muscle cells (VSMCs). The detachment of VSMCs in the media layer of the vessel and migration toward the intima is a critical step in atherosclerosis. VSMC phenotypic switching is a complicated process that modifies VSMC structure and biomechanical function. These changes affect the expression and function of cell adhesion molecules, thus impacting VSMC migration. Accumulating evidence has shown cholesterol is capable of regulating cellular migration, proliferation, and spreading. However, the interaction and coordinated effects of both cellular cholesterol and the extracellular matrix (ECM) stiffness/composition on VSMC biomechanics remains to be elucidated.
Collapse
Affiliation(s)
- Hanna J Sanyour
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States
| | - Alex P Rickel
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States
| | - Zhongkui Hong
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States.
| |
Collapse
|
34
|
Al-Attraqchi OHA, Deb PK, Al-Attraqchi NHA. Review of the Phytochemistry and Pharmacological Properties of Valeriana officinalis. CURRENT TRADITIONAL MEDICINE 2020. [DOI: 10.2174/2215083805666190314112755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extracts of Valeriana officinalis (V. officinalis) are among the most widely used herbal medications worldwide. The roots of this medicinal plant have been shown to be rich in a diverse set of phytochemicals that are biologically active. Various classes of chemical compounds are present in the roots of V. officinalis including, terpenes valepotriates and lignans. The high amount of bioactive chemical components in the extracts of this plant gives it potential medicinal applications in different pathological conditions such as sleep disorders, cardiovascular diseases, anxiety and depression. The most common medicinal use of V. officinalis extract is for the treatment of insomnia and improving the quality of sleep. Mild sedative effects of the extract, as well as the absence of any significant side effects, make it a popular alternative to the synthetic sedative drugs which are usually associated with undesirable adverse effects. In this review, the bioactive chemical components of V. officinalis and their pharmacological activities are highlighted. Also, the medicinal applications of valerian (V. officinalis) extracts and the possible mechanisms of action for the activities are discussed.
Collapse
|
35
|
Leopold JA, Maron BA, Loscalzo J. The application of big data to cardiovascular disease: paths to precision medicine. J Clin Invest 2020; 130:29-38. [PMID: 31895052 DOI: 10.1172/jci129203] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Advanced phenotyping of cardiovascular diseases has evolved with the application of high-resolution omics screening to populations enrolled in large-scale observational and clinical trials. This strategy has revealed that considerable heterogeneity exists at the genotype, endophenotype, and clinical phenotype levels in cardiovascular diseases, a feature of the most common diseases that has not been elucidated by conventional reductionism. In this discussion, we address genomic context and (endo)phenotypic heterogeneity, and examine commonly encountered cardiovascular diseases to illustrate the genotypic underpinnings of (endo)phenotypic diversity. We highlight the existing challenges in cardiovascular disease genotyping and phenotyping that can be addressed by the integration of big data and interpreted using novel analytical methodologies (network analysis). Precision cardiovascular medicine will only be broadly applied to cardiovascular patients once this comprehensive data set is subjected to unique, integrative analytical strategies that accommodate molecular and clinical heterogeneity rather than ignore or reduce it.
Collapse
|
36
|
Nabati M, Namazi SS, Yazdani J, Sharif Nia H. Relation Between Aortic Stiffness Index and Distensibility with Age in Hypertensive Patients. Int J Gen Med 2020; 13:297-303. [PMID: 32606894 PMCID: PMC7296550 DOI: 10.2147/ijgm.s253357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022] Open
Abstract
Background Systolic and diastolic blood pressure is associated with physiologic changes of aortic wall and left ventricular structure. We aimed to evaluate aortic stiffness index and distensibility, global longitudinal strain (GLS), post systolic index (PSI) in hypertensive patients and compare these parameters with normotensive subjects. Patients and Methods Eighty-two patients (42 hypertensive compared with 40 normotensive subjects) with preserved left ventricular ejection fraction and without significant coronary artery disease were enrolled in the study. Systolic and diastolic blood pressure was measured by automated BP measurement system. Aortic stiffness index and distensibility, GLS and PSI were measured by transthoracic echocardiography and compared in both study groups. Results Aortic stiffness index (0.097 vs 0.069) and E/e´ (8.16 vs 6.56) were significantly higher in hypertensive patients, respectively (p<0.05). Aortic distensibility (cm2/dyn) (0.28 vs 0.42) and e´ (cm/s) (8.25 vs 9.52) were significantly lower in hypertensive patients than normotensive subjects (p<0.05). PSI and GLS were not significantly different between both study groups. Aortic stiffness index and distensibility had significant correlation with age in normotensive subjects while this correlation was not statistically significant in hypertensive patients. Conclusion Hypertension is associated with diastolic dysfunction and abnormal aortic wall compliance. Age-related aortic wall changes can present early in hypertensive patients.
Collapse
Affiliation(s)
- Maryam Nabati
- Department of Cardiology, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Shojaeddin Namazi
- Student Research Committee, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Jamshid Yazdani
- Department of Biostatics, Faculty of Health, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Sharif Nia
- Department of Nursing, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
37
|
Morales-Quinones M, Ramirez-Perez FI, Foote CA, Ghiarone T, Ferreira-Santos L, Bloksgaard M, Spencer N, Kimchi ET, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. LIMK (LIM Kinase) Inhibition Prevents Vasoconstriction- and Hypertension-Induced Arterial Stiffening and Remodeling. Hypertension 2020; 76:393-403. [PMID: 32594801 DOI: 10.1161/hypertensionaha.120.15203] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased arterial stiffness and vascular remodeling precede and are consequences of hypertension. They also contribute to the development and progression of life-threatening cardiovascular diseases. Yet, there are currently no agents specifically aimed at preventing or treating arterial stiffening and remodeling. Previous research indicates that vascular smooth muscle actin polymerization participates in the initial stages of arterial stiffening and remodeling and that LIMK (LIM kinase) promotes F-actin formation and stabilization via cofilin phosphorylation and consequent inactivation. Herein, we hypothesize that LIMK inhibition is able to prevent vasoconstriction- and hypertension-associated arterial stiffening and inward remodeling. We found that small visceral arteries isolated from hypertensive subjects are stiffer and have greater cofilin phosphorylation than those from nonhypertensives. We also show that LIMK inhibition prevents arterial stiffening and inward remodeling in isolated human small visceral arteries exposed to prolonged vasoconstriction. Using cultured vascular smooth muscle cells, we determined that LIMK inhibition prevents vasoconstrictor agonists from increasing cofilin phosphorylation, F-actin volume, and cell cortex stiffness. We further show that localized LIMK inhibition prevents arteriolar inward remodeling in hypertensive mice. This indicates that hypertension is associated with increased vascular smooth muscle cofilin phosphorylation, cytoskeletal stress fiber formation, and heightened arterial stiffness. Our data further suggest that pharmacological inhibition of LIMK prevents vasoconstriction-induced arterial stiffening, in part, via reductions in vascular smooth muscle F-actin content and cellular stiffness. Accordingly, LIMK inhibition should represent a promising therapeutic means to stop the progression of arterial stiffening and remodeling in hypertension.
Collapse
Affiliation(s)
- Mariana Morales-Quinones
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Francisco I Ramirez-Perez
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Christopher A Foote
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Thaysa Ghiarone
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Larissa Ferreira-Santos
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Instituto do Coração (InCor), Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, Brazil (L.F.-S.)
| | - Maria Bloksgaard
- Department of Molecular Medicine, University of Southern Denmark, Odense (M.B.)
| | | | - Eric T Kimchi
- Department of Surgery (E.T.K.), University of Missouri, Columbia, MO.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (E.T.K., C.M.-A.)
| | - Camila Manrique-Acevedo
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Medicine, Division of Endocrinology, Diabetes and Metabolism (C.M.-A.), University of Missouri, Columbia, MO.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (E.T.K., C.M.-A.)
| | - Jaume Padilla
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Nutrition and Exercise Physiology (J.P.), University of Missouri, Columbia, MO
| | - Luis A Martinez-Lemus
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Medical Pharmacology and Physiology (L.A.M.-L.), University of Missouri, Columbia, MO
| |
Collapse
|
38
|
Tribological Characteristics of Human Vascular Smooth Muscle Cells: The Implication of Disease State on Friction. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.biotri.2020.100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
39
|
Hwang YJ, Cho DH. Activation of AMPK/proteasome/MLCK degradation signaling axis by telmisartan inhibits VSMC contractility and vessel contraction. Biochem Biophys Res Commun 2020; 524:853-860. [DOI: 10.1016/j.bbrc.2020.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/03/2020] [Indexed: 10/25/2022]
|
40
|
Sanyour HJ, Li N, Rickel AP, Torres HM, Anderson RH, Miles MR, Childs JD, Francis KR, Tao J, Hong Z. Statin-mediated cholesterol depletion exerts coordinated effects on the alterations in rat vascular smooth muscle cell biomechanics and migration. J Physiol 2020; 598:1505-1522. [PMID: 32083311 DOI: 10.1113/jp279528] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
KEY POINTS This study demonstrates and evaluates the changes in rat vascular smooth muscle cell biomechanics following statin-mediated cholesterol depletion. Evidence is presented to show correlated changes in migration and adhesion of vascular smooth muscle cells to extracellular matrix proteins fibronectin and collagen. Concurrently, integrin α5 expression was enhanced but not integrin α2. Atomic force microscopy analysis provides compelling evidence of coordinated reduction in vascular smooth muscle cell stiffness and actin cytoskeletal orientation in response to statin-mediated cholesterol depletion. Proof is provided that statin-mediated cholesterol depletion remodels total vascular smooth muscle cell cytoskeletal orientation that may additionally participate in altering ex vivo aortic vessel function. It is concluded that statin-mediated cholesterol depletion may coordinate vascular smooth muscle cell migration and adhesion to different extracellular matrix proteins and regulate cellular stiffness and cytoskeletal orientation, thus impacting the biomechanics of the cell. ABSTRACT Not only does cholesterol induce an inflammatory response and deposits in foam cells at the atherosclerotic plaque, it also regulates cellular mechanics, proliferation and migration in atherosclerosis progression. Statins are HMG-CoA reductase inhibitors that are known to inhibit cellular cholesterol biosynthesis and are clinically prescribed to patients with hypercholesterolemia or related cardiovascular conditions. Nonetheless, the effect of statin-mediated cholesterol management on cellular biomechanics is not fully understood. In this study, we aimed to assess the effect of fluvastatin-mediated cholesterol management on primary rat vascular smooth muscle cell (VSMC) biomechanics. Real-time measurement of cell adhesion, stiffness, and imaging were performed using atomic force microscopy (AFM). Cellular migration on extra cellular matrix (ECM) protein surfaces was studied by time-lapse imaging. The effect of changes in VSMC biomechanics on aortic function was assessed using an ex vivo myograph system. Fluvastatin-mediated cholesterol depletion (-27.8%) lowered VSMC migration distance on a fibronectin (FN)-coated surface (-14.8%) but not on a type 1 collagen (COL1)-coated surface. VSMC adhesion force to FN (+33%) and integrin α5 expression were enhanced but COL1 adhesion and integrin α2 expression were unchanged upon cholesterol depletion. In addition, VSMC stiffness (-46.6%) and ex vivo aortic ring contraction force (-40.1%) were lowered and VSMC actin cytoskeletal orientation was reduced (-24.5%) following statin-mediated cholesterol depletion. Altogether, it is concluded that statin-mediated cholesterol depletion may coordinate VSMC migration and adhesion to different ECM proteins and regulate cellular stiffness and cytoskeletal orientation, thus impacting the biomechanics of the cell and aortic function.
Collapse
Affiliation(s)
- Hanna J Sanyour
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Na Li
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Alex P Rickel
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Haydee M Torres
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, 57007, USA
| | - Ruthellen H Anderson
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Miranda R Miles
- BioSNTR, Sioux Falls, SD, 57107, USA.,Mechanical Engineering Department, South Dakota State University, Brookings, SD, 57007, USA
| | - Josh D Childs
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Kevin R Francis
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Jianning Tao
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.,Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, 57007, USA
| | - Zhongkui Hong
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| |
Collapse
|
41
|
Nwabuo CC, Vasan RS. Pathophysiology of Hypertensive Heart Disease: Beyond Left Ventricular Hypertrophy. Curr Hypertens Rep 2020; 22:11. [PMID: 32016791 DOI: 10.1007/s11906-020-1017-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Given that the life expectancy and the burden of hypertension are projected to increase over the next decade, hypertensive heart disease (HHD) may be expected to play an even more central role in the pathophysiology of cardiovascular disease (CVD). A broader understanding of the features and underlying mechanisms that constitute HHD therefore is of paramount importance. RECENT FINDINGS HHD is a condition that arises as a result of elevated blood pressure and constitutes a key underlying mechanism for cardiovascular morbidity and mortality. Historically, studies investigating HHD have primarily focused on left ventricular (LV) hypertrophy (LVH), but it is increasingly apparent that HHD encompasses a range of target-organ damage beyond LVH, including other cardiovascular structural and functional adaptations that may occur separately or concomitantly. HHD is characterized by micro- and macroscopic myocardial alterations, structural phenotypic adaptations, and functional changes that include cardiac fibrosis, and the remodeling of the atria and ventricles and the arterial system. In this review, we summarize the structural and functional alterations in the cardiac and vascular system that constitute HHD and underscore their underlying pathophysiology.
Collapse
Affiliation(s)
| | - Ramachandran S Vasan
- Framingham Heart Study, 73 Mt. Wayte Avenue, Suite 2, Framingham, MA, 01702, USA. .,Departments of Epidemiology and Biostatistics, Boston University School of Public Health, Boston, MA, USA. .,Department of Medicine, Sections of Preventive Medicine and Epidemiology, and Cardiovascular Medicine, Boston University Schools of Medicine, Boston, MA, USA.
| |
Collapse
|
42
|
Mangum KD, Freeman EJ, Magin JC, Taylor JM, Mack CP. Transcriptional and posttranscriptional regulation of the SMC-selective blood pressure-associated gene, ARHGAP42. Am J Physiol Heart Circ Physiol 2020; 318:H413-H424. [PMID: 31886719 PMCID: PMC7052622 DOI: 10.1152/ajpheart.00143.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 12/19/2022]
Abstract
We previously showed that ARHGAP42 is a smooth muscle cell (SMC)-selective, RhoA-specific GTPase activating protein that regulates blood pressure and that a minor allele single nucleotide variation within a DNAse hypersensitive regulatory element in intron1 (Int1DHS) increased ARHGAP42 expression by promoting serum response factor binding. The goal of the current study was to identify additional transcriptional and posttranscriptional mechanisms that control ARHGAP42 expression. Using deletion/mutation, gel shift, and chromatin immunoprecipitation experiments, we showed that recombination signal binding protein for immunoglobulin κ-J region (RBPJ) and TEA domain family member 1 (TEAD1) binding to a conserved core region was required for full IntDHS transcriptional activity. Importantly, overexpression of the notch intracellular domain (NICD) or plating SMCs on recombinant jagged-1 increased IntDHS activity and endogenous ARHGAP42 expression while siRNA-mediated knockdown of TEAD1 inhibited ARHGAP42 mRNA levels. Re-chromatin immunoprecipitation experiments indicated that RBPJ and TEAD1 were bound to the Int1DHS enhancer at the same time, and coimmunoprecipitation assays indicated that these factors interacted physically. Our results also suggest TEAD1 and RBPJ bound cooperatively to the Int1DHS and that the presence of TEAD1 promoted the recruitment of NICD by RBPJ. Finally, we showed that ARHGAP42 expression was inhibited by micro-RNA 505 (miR505) which interacted with the ARHGAP42 3'-untranslated region (UTR) to facilitate its degradation and by AK124326, a long noncoding RNA that overlaps with the ARHGAP42 transcription start site on the opposite DNA strand. Since siRNA-mediated depletion of AK124326 was associated with increased H3K9 acetylation and RNA Pol-II binding at the ARHGAP42 gene, it is likely that AK124326 inhibits ARHGAP42 transcription.NEW & NOTEWORTHY First, RBPJ and TEAD1 converge at an intronic enhancer to regulate ARHGAP42 expression in SMCs. Second, TEAD1 and RBPJ interact physically and bind cooperatively to the ARHGAP42 enhancer. Third, miR505 interacts with the ARHGAP42 3'-UTR to facilitate its degradation. Finally, LncRNA, AK124326, inhibits ARHGAP42 transcription.
Collapse
Affiliation(s)
- Kevin D Mangum
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Emily J Freeman
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Justin C Magin
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Joan M Taylor
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Christopher P Mack
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| |
Collapse
|
43
|
Ghaedi E, Foshati S, Ziaei R, Beigrezaei S, Kord-Varkaneh H, Ghavami A, Miraghajani M. Effects of phytosterols supplementation on blood pressure: A systematic review and meta-analysis. Clin Nutr 2019; 39:2702-2710. [PMID: 31902603 DOI: 10.1016/j.clnu.2019.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 02/01/2023]
Abstract
Several reports have indicated a positive effect of phytosterols on blood pressure (BP), nevertheless these findings have been controversial. Therefore, a systematic review and meta-analysis of randomized controlled trials (RCTs) was aimed to investigate the effects of phytosterol supplementation on BP. An online search was carried out in PubMed, Scopus, ISI Web of Science, Cochrane library and Google Scholar up to May 2019. Weighted Mean difference (WMD) with 95% confidence intervals (CIs) were calculated using a fixed-effects model. The present meta-analysis of 19 RCTs showed that supplementation with phytosterols can decrease both systolic BP (WMD: -1.55 mmHg, 95% CI: -2.67 to -0.42, p = 0.007) and diastolic BP (WMD: -0.84 mmHg, 95% CI: -1.60 to -0.08, p = 0.03). Dose-response analysis revealed that phytosterol intake change SBP significantly based on treatment dose in nonlinear fashion. Subgroup analysis based on duration showed a significant effect of phytosterol on SBP and DBP in subsets of <12 weeks. In addition, a significant effect of phytosterol was observed in dosage of ≥2000 mg for SBP and <2000 mg for DBP. Based on current findings supplementation with phytosterol may be a beneficial adjuvant therapy in hypertensive patients as well as a complementary preventive option in prehypertensive and normotensive individuals. However, this issue is still open and requires further investigation in future studies.
Collapse
Affiliation(s)
- Ehsan Ghaedi
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran; Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Foshati
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rahele Ziaei
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sara Beigrezaei
- Nutrition and Food Security Research Center, Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamed Kord-Varkaneh
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abed Ghavami
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Miraghajani
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, UK.
| |
Collapse
|
44
|
Sharma N, Dev R, Belenchia AM, Aroor AR, Whaley-Connell A, Pulakat L, Hans CP. Deficiency of IL12p40 (Interleukin 12 p40) Promotes Ang II (Angiotensin II)-Induced Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2019; 39:212-223. [PMID: 30580570 DOI: 10.1161/atvbaha.118.311969] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Objective- Abdominal aortic aneurysm is caused by the accumulation of inflammatory cells in the aortic wall. Our recent studies demonstrated that inhibition of Notch signaling attenuates abdominal aortic aneurysm formation by shifting the macrophage balance towards anti-inflammatory (M2) phenotype. Using IL12p40-/- (interleukin 12 p40) mice, we investigated the effects of M2-predominant macrophages on the development of abdominal aortic aneurysm. Approach and Results- Male (8-10 week-old) wild-type and IL12p40-/- mice (n=15) on C57BL/6 background were infused with Ang II (angiotensin II, 1000 ng/kg per minute) by implanting osmotic pumps subcutaneously for 28 days. In the IL12p40-/- mice, Ang II significantly increased the maximal intraluminal diameter (9/15) as determined by transabdominal ultrasound imaging. In addition, IL12p40-deletion significantly increased aortic stiffness in response to Ang II as measured by pulse wave velocity and atomic force microscopy. Histologically, IL12p40-/- mice exhibited increased maximal external diameter of aorta and aortic lesions associated with collagen deposition and increased elastin fragmentation compared with wild-type mice infused with Ang II. Mechanistically, IL12p40 deficiency by siRNA (small interfering RNA) augmented the Tgfβ2-mediated Mmp2 expression in wild-type bone marrow-derived macrophages without affecting the expression of Mmp9. No such effects of IL12p40 deficiency on MMP2/MMP9 was observed in human aortic smooth muscle cells or fibroblasts. Depletion of macrophages in IL12p40-/- mice by clodronate liposomes significantly decreased the maximal external diameter of aorta and aortic stiffness in response to Ang II as determined by imaging and atomic force microscopy. Conclusions- IL12p40 depletion promotes the development of abdominal aortic aneurysm, in part, by facilitating recruitment of M2-like macrophages and potentiating aortic stiffness and fibrosis mediated by Tgfβ2.
Collapse
Affiliation(s)
- Neekun Sharma
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Rishabh Dev
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Anthony M Belenchia
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Annayya R Aroor
- Department of Medical Pharmacology and Physiology (A.R.A., C.P.H.), University of Missouri, Columbia
| | - Adam Whaley-Connell
- Harry S. Truman Memorial Veterans' Hospital (A.W.-C.), University of Missouri, Columbia
| | - Lakshmi Pulakat
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Chetan P Hans
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Department of Medical Pharmacology and Physiology (A.R.A., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| |
Collapse
|
45
|
Jaminon A, Reesink K, Kroon A, Schurgers L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. Int J Mol Sci 2019; 20:E5694. [PMID: 31739395 PMCID: PMC6888164 DOI: 10.3390/ijms20225694] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Arterial remodeling refers to the structural and functional changes of the vessel wall that occur in response to disease, injury, or aging. Vascular smooth muscle cells (VSMC) play a pivotal role in regulating the remodeling processes of the vessel wall. Phenotypic switching of VSMC involves oxidative stress-induced extracellular vesicle release, driving calcification processes. The VSMC phenotype is relevant to plaque initiation, development and stability, whereas, in the media, the VSMC phenotype is important in maintaining tissue elasticity, wall stress homeostasis and vessel stiffness. Clinically, assessment of arterial remodeling is a challenge; particularly distinguishing intimal and medial involvement, and their contributions to vessel wall remodeling. The limitations pertain to imaging resolution and sensitivity, so methodological development is focused on improving those. Moreover, the integration of data across the microscopic (i.e., cell-tissue) and macroscopic (i.e., vessel-system) scale for correct interpretation is innately challenging, because of the multiple biophysical and biochemical factors involved. In the present review, we describe the arterial remodeling processes that govern arterial stiffening, atherosclerosis and calcification, with a particular focus on VSMC phenotypic switching. Additionally, we review clinically applicable methodologies to assess arterial remodeling and the latest developments in these, seeking to unravel the ubiquitous corroborator of vascular pathology that calcification appears to be.
Collapse
Affiliation(s)
- Armand Jaminon
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Koen Reesink
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Abraham Kroon
- Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
46
|
Wang Z, Fu Z, Yang Y, Xing W, Zhang X, Wang J, Li Y, Yuan L, Gao F. A novel methodology for rat aortic pulse wave velocity assessment by Doppler ultrasound: validation against invasive measurements. Am J Physiol Heart Circ Physiol 2019; 317:H1376-H1387. [PMID: 31702970 DOI: 10.1152/ajpheart.00382.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is still lack of a simple, accurate, and noninvasive method for rat aortic pulse wave velocity (PWV) measurement, especially the transit distance cannot be accurately measured. Thus, we aimed to derive an equation for aortic transit distance as a function of the nose-to-rump length (L) and to test the hypothesis that aortic PWV measured by new equation combined with Doppler ultrasound (the "equation method") may have stronger correlation with invasive measurements than traditional "body surface method." Two-hundred male Sprague-Dawley (SD) rats (age ranged 5-24 wk) were included in protocol 1, and the aortic transit distances were measured postmortem. In protocol 2, heart-femoral PWV and carotid-femoral PWV were measured by equation method (hfPWVE, cfPWVE) and also by traditional body surface method (hfPWVS, cfPWVS) in another 30 young and 28 old rats. These measurements were then validated against invasively measured hfPWVI and cfPWVI from the same animal. Protocol 1 showed that the heart-femoral transit distance could be calculated by 0.6086 × L - 1.6523, and the carotid-femoral transit distance by 0.4614 × L + 1.8335. In protocol 2, in young rats, the Pearson r between hfPWVE, cfPWVE, hfPWVS, and cfPWVS and their corresponding invasive measurement were 0.8962, 0.8509, 0.8387, and 0.7828, respectively (all P < 0.0001). In the old group, the results were 0.8718, 0.7999, 0.8330, and 0.7112, respectively (all P < 0.0001). The hfPWVE and cfPWVE showed better agreement with hfPWVI and cfPWVI and lower intra- and interobserver variability compared with hfPWVS and cfPWVS in both groups. These findings demonstrate that this novel methodology provides a simple and reliable method for rat noninvasive aortic PWV measurement.NEW & NOTEWORTHY First, when measuring aortic PWV in SD rat models, the heart-femoral transit distance can be estimated by 0.6086 × L - 1.6523, and the carotid-femoral distance transit distance can be estimated by 0.4614 × L + 1.8335, where L (in mm) is nose-to-rump length. Second, this novel methodology for aortic PWV measurement was validated with a closer correlation with the invasive measurements than traditional approach in young and old rats. Third, this study provides a simple and reliable method for rat noninvasive aortic PWV measurement.
Collapse
Affiliation(s)
- Zhen Wang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Zihao Fu
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Yong Yang
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenjuan Xing
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Jiaping Wang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Yongzhi Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Lijun Yuan
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Feng Gao
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
47
|
Lv N, Li C, Liu X, Qi C, Wang Z. miR-34b Alleviates High Glucose-Induced Inflammation and Apoptosis in Human HK-2 Cells via IL-6R/JAK2/STAT3 Signaling Pathway. Med Sci Monit 2019; 25:8142-8151. [PMID: 31665127 PMCID: PMC6842269 DOI: 10.12659/msm.917128] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background It is well established that inflammation and apoptosis of renal tubular epithelial cells caused by hyperglycemia contribute to the development of diabetic nephropathy (DN). Although microRNAs (miRNAs) are known to have roles in inflammation-related disorders, the exact role of miR-34b in DN has not been defined, and the regulatory mechanism has been unclear. This study aimed to clarify the role of miR-34b in DN pathogenesis. Material/Methods Expression of miR-34b, IL-6R, and other key factors of inflammation, apoptosis (TNF-α, IL-1β, IL-6, caspase-3) in high glucose (HG)-induced HK-2 cells were measured by real-time PCR, Western blot, and flow cytometric cell apoptosis assays. We used luciferase reporter assay to detect the target of miR-34b. Moreover, the targeting gene of miR-34b and its downstream JAK2/STAT3 signaling pathway were explored. Results It was demonstrated that miR-34b overexpression inhibited apoptosis and expression levels of TNF-α, IL-1β, IL-6, and caspase-3 in HG-treated HK-2 cells. We also found that IL-6R is a direct target of miR-34b, which could rescue inflammation and apoptosis in HG-treated HK-2 cells transfected with miR-34b mimic. Furthermore, we showed that overexpression of miR-34b inhibited the IL-6R/JAK2/STAT3 signaling pathway in HG-treated HK-2 cells. Conclusions Our data suggest that overexpression of miR-34b improves inflammation and ameliorates apoptosis in HG-induced HK-2 cells via the IL-6R/JAK2/STAT3 pathway, indicating that miR-34b could be a promising therapeutic target in DN.
Collapse
Affiliation(s)
- Na Lv
- Department of Endocrinology, Dongying People's Hospital, Dongying, Shangdong, China (mainland)
| | - Chunqing Li
- Department of Endocrinology, Dongying People's Hospital, Dongying, Shangdong, China (mainland)
| | - Xin Liu
- Department of Endocrinology, Dongying People's Hospital, Dongying, Shangdong, China (mainland)
| | - Caihui Qi
- Department of Endocrinology, Dongying People's Hospital, Dongying, Shangdong, China (mainland)
| | - Zhenqing Wang
- Department of Endocrinology, Dongying People's Hospital, Dongying, Shangdong, China (mainland)
| |
Collapse
|
48
|
Sharma N, Dev R, Ruiz-Rosado JDD, Partida-Sanchez S, Guerau-de-Arellano M, Dhakal P, Kuivaniemi H, Hans CP. Pharmacological inhibition of Notch signaling regresses pre-established abdominal aortic aneurysm. Sci Rep 2019; 9:13458. [PMID: 31530833 PMCID: PMC6748927 DOI: 10.1038/s41598-019-49682-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by transmural infiltration of myeloid cells at the vascular injury site. Previously, we reported preventive effects of Notch deficiency on the development of AAA by reduction of infiltrating myeloid cells. In this study, we examined if Notch inhibition attenuates the progression of pre-established AAA and potential implications. Pharmacological Notch inhibitor (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-(S)-phenylglycine t-butyl ester; DAPT) was administered subcutaneously three times a week starting at day 28 of angiotensin II (AngII) infusion. Progressive increase in pulse wave velocity (PWV), maximal intra-luminal diameter (MILD) and maximal external aortic diameter (MEAD) were observed at day 56 of the AngII. DAPT prevented such increase in MILD, PWV and MEAD (P < 0.01). Histologically, the aortae of DAPT-treated Apoe-/- mice had significant reduction in inflammatory response and elastin fragmentation. Naked collagen microfibrils and weaker banded structure observed in the aortae of Apoe-/- mice in response to AngII, were substantially diminished by DAPT. A significant decrease in the proteolytic activity in the aneurysmal tissues and vascular smooth muscle cells (vSMCs) was observed with DAPT (P < 0.01). In human and mouse AAA tissues, increased immunoreactivity of activated Notch signaling correlated strongly with CD38 expression (R2 = 0.61). Collectively, we propose inhibition of Notch signaling as a potential therapeutic target for AAA progression.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/metabolism
- Angiotensin II/adverse effects
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/diagnostic imaging
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/metabolism
- Cells, Cultured
- Collagen/metabolism
- Cytokines/metabolism
- Dipeptides/pharmacology
- Disease Models, Animal
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Gene Expression Regulation/drug effects
- Humans
- Male
- Membrane Glycoproteins/metabolism
- Mice
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Receptors, Notch/antagonists & inhibitors
- Receptors, Notch/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Neekun Sharma
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA
| | - Rishabh Dev
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA
| | - Juan de Dios Ruiz-Rosado
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Medical Laboratory Science Division, The Ohio State University, Columbus, OH, USA
| | - Pramod Dhakal
- Animal Science Research Center, University of Missouri, Columbia, USA
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Chetan P Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA.
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA.
- Medical Pharmacology and Physiology, University of Missouri, Columbia, USA.
| |
Collapse
|
49
|
Spronck B, Humphrey JD. Arterial Stiffness: Different Metrics, Different Meanings. J Biomech Eng 2019; 141:091004. [PMID: 30985880 PMCID: PMC6808013 DOI: 10.1115/1.4043486] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/25/2019] [Indexed: 12/18/2022]
Abstract
Findings from basic science and clinical studies agree that arterial stiffness is fundamental to both the mechanobiology and the biomechanics that dictate vascular health and disease. There is, therefore, an appropriately growing literature on arterial stiffness. Perusal of the literature reveals, however, that many different methods and metrics are used to quantify arterial stiffness, and reported values often differ by orders of magnitude and have different meanings. Without clear definitions and an understanding of possible inter-relations therein, it is increasingly difficult to integrate results from the literature to glean true understanding. In this paper, we briefly review methods that are used to infer values of arterial stiffness that span studies on isolated cells, excised intact vessels, and clinical assessments. We highlight similarities and differences and identify a single theoretical approach that can be used across scales and applications and thus could help to unify future results. We conclude by emphasizing the need to move toward a synthesis of many disparate reports, for only in this way will we be able to move from our current fragmented understanding to a true appreciation of how vascular cells maintain, remodel, or repair the arteries that are fundamental to cardiovascular properties and function.
Collapse
Affiliation(s)
- B. Spronck
- Department of Biomedical Engineering,
Yale University,
New Haven, CT 06520
| | - J. D. Humphrey
- Fellow ASME
Department of Biomedical Engineering,
Yale University,
New Haven, CT 06520;
Vascular Biology and Therapeutics Program,
Yale School of Medicine,
New Haven, CT 06520
e-mail:
| |
Collapse
|
50
|
Farrell K, Simmers P, Mahajan G, Boytard L, Camardo A, Joshi J, Ramamurthi A, Pinet F, Kothapalli CR. Alterations in phenotype and gene expression of adult human aneurysmal smooth muscle cells by exogenous nitric oxide. Exp Cell Res 2019; 384:111589. [PMID: 31473210 DOI: 10.1016/j.yexcr.2019.111589] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/24/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022]
Abstract
Abdominal aortic aneurysms (AAA) are characterized by matrix remodeling, elastin degradation, absence of nitric oxide (NO) signaling, and inflammation, influencing smooth muscle cell (SMC) phenotype and gene expression. Little is known about the biomolecular release and intrinsic biomechanics of human AAA-SMCs. NO delivery could be an attractive therapeutic strategy to restore lost functionality of AAA-SMCs by inhibiting inflammation and cell stiffening. We aim to establish the differences in phenotype and gene expression of adult human AAA-SMCs from healthy SMCs. Based on our previous study which showed benefits of optimal NO dosage delivered via S-Nitrosoglutathione (GSNO) to healthy aortic SMCs, we tested whether such benefits would occur in AAA-SMCs. The mRNA expression of three genes involved in matrix degradation (ACE, ADAMTS5 and ADAMTS8) was significantly downregulated in AAA-SMCs. Total protein and glycosaminoglycans synthesis were higher in AAA-SMCs than healthy-SMCs (p < 0.05 for AAA-vs. healthy- SMC cultures) and was enhanced by GSNO and 3D cultures (p < 0.05 for 3D vs. 2D cultures; p < 0.05 for GSNO vs. non-GSNO cases). Elastin gene expression, synthesis and deposition, desmosine crosslinker levels, and lysyl oxidase (LOX) functional activity were lower, while cell proliferation, iNOS, LOX and fibrillin-1 gene expressions were higher in AAA-SMCs (p < 0.05 between respective cases), with differential benefits from GSNO exposure. GSNO and 3D cultures reduced MMPs -2, -9, and increased TIMP-1 release in AAA-SMC cultures (p < 0.05 for GSNO vs. non-GSNO cultures). AAA-SMCs were inherently stiffer and had smoother surface than healthy SMCs (p < 0.01 in both cases), but GSNO reduced stiffness (~25%; p < 0.01) and increased roughness (p < 0.05) of both cell types. In conclusion, exogenously-delivered NO offers an attractive strategy by providing therapeutic benefits to AAA-SMCs.
Collapse
Affiliation(s)
- Kurt Farrell
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Phillip Simmers
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Gautam Mahajan
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Ludovic Boytard
- University of Lille, Inserm U1167, Institut Pasteur de Lille, France
| | - Andrew Camardo
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, 44141, USA
| | - Jyotsna Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Anand Ramamurthi
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, 44141, USA
| | - Florence Pinet
- University of Lille, Inserm U1167, Institut Pasteur de Lille, France
| | - Chandrasekhar R Kothapalli
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA.
| |
Collapse
|