1
|
Ben Hassine A, Petit C, Thomas M, Mundweiler S, Guignandon A, Avril S. Gene expression modulation in human aortic smooth muscle cells under induced physiological mechanical stretch. Sci Rep 2024; 14:31147. [PMID: 39732782 DOI: 10.1038/s41598-024-82495-4] [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: 06/04/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
In this study, we investigated gene expression in vitro of human primary Aortic smooth muscle cells (AoSMCs) in response to 9% physiological dynamic stretch over a 4 to 72-h timeframe using RT-qPCR. AoSMC were derived from primary culture and were exposed to continuous cycles of stretch and relaxation at 1 Hz by a computer-controlled Flex Jr.™ Tension System. Unstretched control AoSMCs were simultaneously cultured in the same dishes. Our results revealed a rapid and significant upregulation of specific genes (COL1A1, FBN1, LAMA5, TGFBR1 and TGFBR2) within the initial 4 h for AoSMCs subjected to dynamic stretching, whilst control cells did not respond within the same 4 h. The upregulated genes were the ones associated with extracellular matrix (ECM) fibrillogenesis and regulation of traction forces. Interestingly, stretched cells maintained stable gene expression between 4 and 72 h, whilst control cells exhibited variations over time in the absence of mechanical cues. These findings shed light on the essential role played by pulsatile stretches in the regulation of gene expressions by AoSMCs and the intricate processes governing their mechanobiological function, paving the way for further investigations in cardiovascular health.
Collapse
Affiliation(s)
- Amira Ben Hassine
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Claudie Petit
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Mireille Thomas
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Stéphanie Mundweiler
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Alain Guignandon
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Stéphane Avril
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France.
| |
Collapse
|
2
|
Rinne A, Pluteanu F. Ca 2+ Signaling in Cardiovascular Fibroblasts. Biomolecules 2024; 14:1365. [PMID: 39595542 PMCID: PMC11592142 DOI: 10.3390/biom14111365] [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: 09/09/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Fibrogenesis is a physiological process required for wound healing and tissue repair. It is induced by activation of quiescent fibroblasts, which first proliferate and then change their phenotype into migratory, contractile myofibroblasts. Myofibroblasts secrete extracellular matrix proteins, such as collagen, to form a scar. Once the healing process is terminated, most myofibroblasts undergo apoptosis. However, in some tissues, such as the heart, myofibroblasts remain active and sensitive to neurohumoral factors and inflammatory mediators, which lead eventually to excessive organ fibrosis. Many cellular processes involved in fibroblast activation, including cell proliferation, protein secretion and cell contraction, are highly regulated by intracellular Ca2+ signals. This review summarizes current research on Ca2+ signaling pathways underlying fibroblast activation. We present receptor- and ion channel-mediated Ca2+ signaling pathways, discuss how localized Ca2+ signals of the cell nucleus may be involved in fibroblast activation and present Ca2+-sensitive transcription pathways relevant for fibroblast biology. When investigated, we highlight how the function of Ca2+-handling proteins changes during cardiac and pulmonary fibrosis. Many aspects of Ca2+ signaling remain unexplored in different types of cardiovascular fibroblasts in relation to pathologies, and a better understanding of Ca2+ signaling in fibroblasts will help to design targeted therapies against fibrosis.
Collapse
Affiliation(s)
- Andreas Rinne
- Department of Biophysics and Cellular Biotechnology, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania;
| | - Florentina Pluteanu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| |
Collapse
|
3
|
KIM H, LICHTENSTEIN AH, CORESH J, APPEL LJ, REBHOLZ CM. Serum protein responses to Dietary Approaches to Stop Hypertension (DASH) and DASH-Sodium trials and associations with blood pressure changes. J Hypertens 2024; 42:1823-1830. [PMID: 39196693 PMCID: PMC11849912 DOI: 10.1097/hjh.0000000000003828] [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: 08/30/2024]
Abstract
OBJECTIVES The Dietary Approaches to Stop Hypertension (DASH) diet reduces blood pressure, but the mechanisms underlying DASH diet-blood pressure relations are not well understood. Proteomic measures may provide insights into the pathophysiological mechanisms through which the DASH diet reduces blood pressure. METHODS The DASH (1994-1996) and DASH-Sodium (1997-1999) trials were multicenter, randomized-controlled feeding trials. Proteomic profiling was conducted in serum collected at the end of the feeding period (DASH, N = 215; DASH-Sodium, N = 390). Multivariable linear regression models were used to identify interactions between 71 DASH diet-related proteins and changes in systolic and diastolic blood pressure. Estimates were meta-analyzed across both trials. Elastic net models were used to identify proteins that predict changes in blood pressure. RESULTS Ten significant interactions were identified [systolic blood pressure: seven proteins; diastolic blood pressure: three proteins], which represented nine unique proteins. A high level of renin at the end of the feeding period was associated with greater reductions in diastolic blood pressure in individuals consuming the control than DASH diets. A high level of procollagen c-endopeptidase enhancer 1 (PCOLCE) and collagen triple helix repeat-containing protein 1 (CTHRC1) were associated with greater reductions in systolic blood pressure in individuals consuming the DASH than control diets, and with elevations in systolic blood pressure in individuals consuming the control diets (P for interaction for all tests < 0.05). Elastic net models identified six additional proteins that predicted change in blood pressure. CONCLUSIONS Several novel proteins were identified that may provide some insight into the relationship between the DASH diet and blood pressure.
Collapse
Affiliation(s)
- Hyunju KIM
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Alice H. LICHTENSTEIN
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts
| | - Josef CORESH
- New York University Grossman School of Medicine, New York, New York
| | - Lawrence J. APPEL
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Casey M. REBHOLZ
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Wang H, Martinez Yus M, Brady T, Choi R, Nandakumar K, Smith L, Jang R, Wodu BP, Almodiel JD, Stoddart L, Kim DH, Steppan J, Santhanam L. Sex differences and role of lysyl oxidase-like 2 in angiotensin II-induced hypertension in mice. Am J Physiol Heart Circ Physiol 2024; 327:H642-H659. [PMID: 39028284 PMCID: PMC11427116 DOI: 10.1152/ajpheart.00110.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Hypertension, a disease with known sexual dimorphism, accelerates aging-associated arterial stiffening, partly because of the activation of matrix remodeling caused by increased biomechanical load. In this study, we tested the effect of biological sex and the role of the matrix remodeling enzyme lysyl oxidase-like 2 (LOXL2) in hypertension-induced arterial stiffening. Hypertension was induced by angiotensin II (ANG II) infusion via osmotic minipumps in 12- to 14-wk-old male and female mice. Blood pressure and pulse wave velocity (PWV) were measured noninvasively. Wire myography and uniaxial tensile testing were used to test aortic vasoreactivity and mechanical properties. Aortic wall composition was examined by histology and Western blotting. Uniaxial stretch of cultured cells was used to evaluate the effect of biomechanical strain. LOXL2's catalytic function was examined using knockout and inhibition. ANG II infusion-induced hypertension in both genotypes and sexes. Wild-type (WT) males exhibited arterial stiffening in vivo and ex vivo. Aortic remodeling with increased wall thickness, intralamellar distance, higher LOXL2, and collagen I and IV content was noted in WT males. Female mice did not exhibit increased PWV despite the onset of hypertension. LOXL2 depletion improved vascular reactivity and mechanics in hypertensive males. LOXL2 depletion improved aortic mechanics but worsened hypercontractility in females. Hypertensive cyclic strain contributed to LOXL2 upregulation in the cell-derived matrix in vascular smooth muscle cells (VSMCs) but not endothelial cells. LOXL2's catalytic function facilitated VSMC alignment in response to biomechanical strain. In conclusion, in males, arterial stiffening in hypertension is driven both by VSMC response and matrix remodeling. Females are protected from PWV elevation in hypertension. LOXL2 depletion is protective in males with improved mechanical and functional aortic properties. VSMCs are the primary source of LOXL2 in the aorta, and hypertension increases LOXL2 processing and shifts to collagen I accumulation. Overall, LOXL2 depletion offers protection in young hypertensive males and females.NEW & NOTEWORTHY We examined the effect of sex on the evolution of angiotensin II (ANG II)-induced hypertension and the role of lysyl oxidase-like 2 (LOXL2), an enzyme that catalyzes matrix cross linking. While ANG II led to hypertension and worsening vascular reactivity in both sexes, aortic remodeling and stiffening occurred only in males. LOXL2 depletion improved outcomes in males but not females. Thus males and females exhibit a distinct etiology of hypertension and LOXL2 is an effective target in males.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Amino Acid Oxidoreductases/metabolism
- Amino Acid Oxidoreductases/genetics
- Angiotensin II
- Aorta/physiopathology
- Aorta/pathology
- Aorta/enzymology
- Aorta/drug effects
- Aorta/metabolism
- Cells, Cultured
- Disease Models, Animal
- Hypertension/chemically induced
- Hypertension/physiopathology
- Hypertension/enzymology
- Hypertension/metabolism
- Hypertension/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/physiopathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Sex Factors
- Vascular Remodeling
- Vascular Stiffness
Collapse
Affiliation(s)
- Huilei Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Marta Martinez Yus
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Travis Brady
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Rira Choi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Kavitha Nandakumar
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Logan Smith
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Rosie Jang
- Department of Molecular and Cellular Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Bulouere Princess Wodu
- Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Jose Diego Almodiel
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Laila Stoddart
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Mechanical Engineering, Johns Hopkins University, Whiting School of Engineering, Baltimore, Maryland, United States
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, Maryland, United States
| | - Jochen Steppan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Lakshmi Santhanam
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
5
|
Walsh HJ, Junejo RT, Lip GYH, Fisher JP. The effect of hypertension on cerebrovascular carbon dioxide reactivity in atrial fibrillation patients. Hypertens Res 2024; 47:1678-1687. [PMID: 38600276 PMCID: PMC11150149 DOI: 10.1038/s41440-024-01662-2] [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: 10/25/2023] [Revised: 03/07/2024] [Accepted: 03/17/2024] [Indexed: 04/12/2024]
Abstract
Atrial fibrillation (AF) and hypertension (HTN) are both associated with impaired cerebrovascular carbon dioxide reactivity (CVRCO2), an indicator of cerebral vasodilatory reserve. We hypothesised that CVRCO2 would be lower in patients with both AF and HTN (AF + HTN) compared to normotensive AF patients, due to an additive effect of AF and HTN on CVRCO2. Forty AF (68 ± 9 years) and fifty-seven AF + HTN (68 ± 8 years) patients underwent transcranial Doppler ultrasound measurement of middle cerebral artery blood velocity (MCA Vm) during stepped increases and decreases in end-tidal carbon dioxide (PETCO2). A cerebrovascular conductance index (CVCi) was calculated as the ratio of MCA Vm and mean arterial pressure (MAP). CVRCO2 was determined from the linear slope for MCA Vm and MCA CVCi vs PETCO2. Baseline MAP was higher in AF + HTN than AF (107 ± 9 vs. 98 ± 9 mmHg, respectively; p < 0.001), while MCA Vm was not different (AF + HTN:49.6 [44.1-69.0]; AF:51.7 [45.2-63.3] cm.s-1; p = 0.075), and CVCi was lower in AF + HTN (0.46 [0.42-0.57] vs. 0.54 [0.44-0.63] cm.s-1.mmHg-1; p < 0.001). MCA Vm CVRCO2 was not different (AF + HTN: 1.70 [1.47-2.19]; AF 1.74 [1.54-2.52] cm/s/mmHg-2; p = 0.221), while CVCi CVRCO2 was 13% lower in AF + HTN (0.013 ± 0.004 vs 0.015 ± 0.005 cm.s-1.mmHg-1; p = 0.047). Our results demonstrate blunted cerebral vasodilatory reserve (determined as MCA CVCi CVRCO2) in AF + HTN compared to AF alone. This may implicate HTN as a driver of further cerebrovascular dysfunction in AF that may be important for the development of AF-related cerebrovascular events and downstream cognitive decline. We demonstrated reduced cerebrovascular CO2 responsiveness in atrial fibrillation with hypertension (AF+HTN) vs. atrial fibrillation (AF). Furthermore, AF per se (as opposed to normal sinus rhythm) predicts reduced cerebrovascular CO2 responsiveness. Our findings suggest additional cerebrovascular dysfunction in AF+HTN vs. AF.
Collapse
Affiliation(s)
- Harvey J Walsh
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand
| | - Rehan T Junejo
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - James P Fisher
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
6
|
Noro J, Vilaça-Faria H, Reis RL, Pirraco RP. Extracellular matrix-derived materials for tissue engineering and regenerative medicine: A journey from isolation to characterization and application. Bioact Mater 2024; 34:494-519. [PMID: 38298755 PMCID: PMC10827697 DOI: 10.1016/j.bioactmat.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Biomaterial choice is an essential step during the development tissue engineering and regenerative medicine (TERM) applications. The selected biomaterial must present properties allowing the physiological-like recapitulation of several processes that lead to the reestablishment of homeostatic tissue or organ function. Biomaterials derived from the extracellular matrix (ECM) present many such properties and their use in the field has been steadily increasing. Considering this growing importance, it becomes imperative to provide a comprehensive overview of ECM biomaterials, encompassing their sourcing, processing, and integration into TERM applications. This review compiles the main strategies used to isolate and process ECM-derived biomaterials as well as different techniques used for its characterization, namely biochemical and chemical, physical, morphological, and biological. Lastly, some of their applications in the TERM field are explored and discussed.
Collapse
Affiliation(s)
- Jennifer Noro
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Helena Vilaça-Faria
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
7
|
Prapas S, Katsavrias K, Gaudino M, Puskas JD, Di Mauro M, Zografos P, Guarracini S, Linardakis I, Panagiotopoulos I, Di Marco M, Papandreopoulos S, Pomakidou S, Totaro A, Calafiore AM. Saphenous vein to the right coronary system from the right thoracic artery or the aorta. Long-term propensity-matched results of 2 groups. Eur J Cardiothorac Surg 2024; 65:ezae060. [PMID: 38400814 DOI: 10.1093/ejcts/ezae060] [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: 11/06/2023] [Revised: 01/09/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024] Open
Abstract
OBJECTIVES Since 2000, we anastomosed the saphenous vein graft to the right coronary artery system using the stump of the right internal thoracic artery as inflow. The long-term results of patients where the right coronary artery was grafted with the right internal thoracic artery or the ascending aorta as saphenous vein inflow has not been reported. METHODS From 2000 to 2018, 699 consecutive patients had right internal thoracic artery elongated with saphenous vein (I-graft group, n = 358, 51.2%) or saphenous vein from the aorta (Ao-graft group, n = 341, 48.8%) on right coronary artery system. Inclusion criteria were age ≤75 years, bilateral internal thoracic arteries as a Y graft on the left system (three-vessel disease, n = 603, 86.3%) or as a left internal thoracic artery on left anterior descending and right internal thoracic artery elongated with saphenous vein on the right coronary artery system (two-vessel disease, n = 96, 13.7%), only 1 saphenous vein per patient. Propensity-matching identified 272 patients per group. One-hundred and twenty-two patients underwent coronary computed tomographic angiography to asses grafts patency after a median follow-up of 88 (65-93) months. RESULTS In the paired samples, there was no difference in the early outcome. Ten-year survival and freedom from death, non-fatal acute myocardial infarction and repeat revascularization were higher in I-graft group: 90.6 [standard error (SE): 2.0] vs 78.2 (SE: 5.3), P = 0.0266, and 85.2 (SE: 2.4) vs 69.9 (SE: 5.3), P = 0.0179. Saphenous vein graft, at a long-time follow-up, showed a higher patency rate (81.6% (SE: 7.0) vs 50.7% (SE: 7.9), P < 0.0001) and a smaller internal lumen diameter (2.7, standard deviation: 0.4 vs 3.4, standard deviation: 0.6 mm, P < 0.0001) when right internal thoracic artery was the inflow. CONCLUSIONS Grafting the right coronary artery with saphenous vein may entail higher patency rate and better outcome when the inflow is the right internal thoracic artery than when is the ascending aorta. Prospective randomized data are needed to test this hypothesis.
Collapse
Affiliation(s)
- Sotirios Prapas
- 1st Department of Cardiac Surgery A, Henry Dunant Hospital, Athens, Greece
| | | | - Mario Gaudino
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - John D Puskas
- Department of Cardiovascular Surgery, Mount Sinai Hospital and Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michele Di Mauro
- Cardio-Thoracic Surgery Unit, Heart and Vascular Centre, Maastricht University Medical Centre (MUMC), Cardiovascular Research Institute Maastricht (CARIM), Maastricht, Netherlands
- Department of Cardiology, "Pierangeli" Hospital, Pescara, Italy
| | | | | | - Ioannis Linardakis
- 1st Department of Cardiac Surgery A, Henry Dunant Hospital, Athens, Greece
| | | | | | | | | | - Antonio Totaro
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | | |
Collapse
|
8
|
Xiao Y, Vazquez-Padron RI, Martinez L, Singer HA, Woltmann D, Salman LH. Role of platelet factor 4 in arteriovenous fistula maturation failure: What do we know so far? J Vasc Access 2024; 25:390-406. [PMID: 35751379 PMCID: PMC9974241 DOI: 10.1177/11297298221085458] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The rate of arteriovenous fistula (AVF) maturation failure remains unacceptably high despite continuous efforts on technique improvement and careful pre-surgery planning. In fact, half of all newly created AVFs are unable to be used for hemodialysis (HD) without a salvage procedure. While vascular stenosis in the venous limb of the access is the culprit, the underlying factors leading to vascular narrowing and AVF maturation failure are yet to be determined. We have recently demonstrated that AVF non-maturation is associated with post-operative medial fibrosis and fibrotic stenosis, and post-operative intimal hyperplasia (IH) exacerbates the situation. Multiple pathological processes and signaling pathways are underlying the stenotic remodeling of the AVF. Our group has recently indicated that a pro-inflammatory cytokine platelet factor 4 (PF4/CXCL4) is upregulated in veins that fail to mature after AVF creation. Platelet factor 4 is a fibrosis marker and can be detected in vascular stenosis tissue, suggesting that it may contribute to AVF maturation failure through stimulation of fibrosis and development of fibrotic stenosis. Here, we present an overview of the how PF4-mediated fibrosis determines AVF maturation failure.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Daniel Woltmann
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Loay H Salman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY, USA
| |
Collapse
|
9
|
Jiang QL, Xu JY, Yao QP, Jiang R, Xu Q, Zhang BT, Li T, Jiang J. Transfer RNA-derived small RNA tRF-Glu-CTC attenuates neointimal formation via inhibition of fibromodulin. Cell Mol Biol Lett 2024; 29:2. [PMID: 38172726 PMCID: PMC10763295 DOI: 10.1186/s11658-023-00523-z] [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: 08/02/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Neointimal hyperplasia is a pathological vascular remodeling caused by abnormal proliferation and migration of subintimal vascular smooth muscle cells (VSMCs) following intimal injury. There is increasing evidence that tRNA-derived small RNA (tsRNA) plays an important role in vascular remodeling. The purpose of this study is to search for tsRNAs signature of neointima formation and to explore their potential functions. The balloon injury model of rat common carotid artery was replicated to induce intimal hyperplasia, and the differentially expressed tsRNAs (DE-tsRNAs) in arteries with intimal hyperplasia were screened by small RNA sequencing and tsRNA library. A total of 24 DE-tsRNAs were found in the vessels with intimal hyperplasia by small RNA sequencing. In vitro, tRF-Glu-CTC inhibited the expression of fibromodulin (FMOD) in VSMCs, which is a negative modulator of TGF-β1 activity. tRF-Glu-CTC also increased VSMC proliferation and migration. In vivo experiments showed that inhibition of tRF-Glu-CTC expression after balloon injury of rat carotid artery can reduce the neointimal area. In conclusion, tRF-Glu-CTC expression is increased after vascular injury and inhibits FMOD expression in VSMCs, which influences neointima formation. On the other hand, reducing the expression of tRF-Glu-CTC after vascular injury may be a potential approach to prevent vascular stenosis.
Collapse
Affiliation(s)
- Qi-Lan Jiang
- Department of Clinical Nutrition, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jia-Ying Xu
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China
| | - Qing-Ping Yao
- Institute of Mechanobiology and Medical Engineering, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Jiang
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qin Xu
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China
| | - Bo-Tao Zhang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
10
|
Wang T, Zhang S, Wang L, Su K, Tang Z, He H, Shi Y, Liu Y, Zheng M, Fu W, Hu S, Zhang X, Wu T. Local application of triamcinolone acetonide-conjugated chitosan membrane to prevent benign biliary stricture. Drug Deliv Transl Res 2022; 12:2895-2906. [PMID: 35426041 DOI: 10.1007/s13346-022-01153-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 12/16/2022]
Abstract
Benign biliary stricture (BBS) is the proliferation of fibrous tissue of the biliary tract caused by the biliary operation, bile duct stones, cholangitis, trauma, and other etiologies due to scar contracture. Recent therapeutic strategies to suppress stenosis are insufficient. Here, we developed a sustained-release membrane (SM) of triamcinolone acetonide (TA) with N-succinyl hydroxypropyl chitosan (TASM) for inhibiting fibroblast proliferation in vitro and bile duct hyperplasia in the rabbit model for benign biliary stricture formation. The TASM were successfully placed in 45 of 50 rabbits. Evaluation of subcutaneous stimulation and acute liver injury confirms the safety of TASM in vivo. Compared to the control group, the TASM can significantly inhibit the proliferation of scar muscle fibroblasts in vitro. ELISA and immunofluorescence showed TASM could increase bFGF level and inhibit expression of TGFβ1 and αSMA. Cholangiographic and histologic examinations demonstrated significantly decreased tissue hyperplasia in the TASM groups compared with the model group. The immunohistochemical staining showed that TASM could reduce the level of cytokine-induced scars and inhibit the proliferation of myofibroblasts. Taken together, the chitosan membrane chemically conjugated with TA can effectively inhibit the benign biliary stricture. Further clinical usage of this membrane may effectively reduce the occurrence of benign biliary stricture.
Collapse
Affiliation(s)
- Tao Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Shibo Zhang
- Department of Hepatopancreatobiliary Surgery, The First Hospital of QuJing, QuJing, 655000, Yunnan, China
| | - Lianmin Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Kun Su
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Zhiyi Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Haiyu He
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Yanmei Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Yaqiong Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Mengyao Zheng
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Wen Fu
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Sheng Hu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Xiaowen Zhang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Tao Wu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China.
| |
Collapse
|
11
|
van Asten JGM, Ristori T, Nolan DR, Lally C, Baaijens FPT, Sahlgren CM, Loerakker S. Computational analysis of the role of mechanosensitive Notch signaling in arterial adaptation to hypertension. J Mech Behav Biomed Mater 2022; 133:105325. [PMID: 35839633 PMCID: PMC7613661 DOI: 10.1016/j.jmbbm.2022.105325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/03/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022]
Abstract
Arteries grow and remodel in response to mechanical stimuli. Hypertension, for example, results in arterial wall thickening. Cell-cell Notch signaling between vascular smooth muscle cells (VSMCs) is known to be involved in this process, but the underlying mechanisms are still unclear. Here, we investigated whether Notch mechanosensitivity to strain may regulate arterial thickening in hypertension. We developed a multiscale computational framework by coupling a finite element model of arterial mechanics, including residual stress, to an agent-based model of mechanosensitive Notch signaling, to predict VSMC phenotypes as an indicator of growth and remodeling. Our simulations revealed that the sensitivity of Notch to strain at mean blood pressure may be a key mediator of arterial thickening in hypertensive arteries. Further simulations showed that loss of residual stress can have synergistic effects with hypertension, and that changes in the expression of Notch receptors, but not Jagged ligands, may be used to control arterial growth and remodeling and to intensify or counteract hypertensive thickening. Overall, we identify Notch mechanosensitivity as a potential mediator of vascular adaptation, and we present a computational framework that can facilitate the testing of new therapeutic and regenerative strategies.
Collapse
Affiliation(s)
- Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - David R Nolan
- School of Engineering and Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Caitríona Lally
- School of Engineering and Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Frank P T Baaijens
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
12
|
Dependency of deformation of cell nucleus on stretch direction of tissue: Relation to anisotropic response of aortic media to hypertension. J Mech Behav Biomed Mater 2022; 133:105326. [DOI: 10.1016/j.jmbbm.2022.105326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/07/2022] [Accepted: 06/18/2022] [Indexed: 11/19/2022]
|
13
|
Karakaya C, van Turnhout MC, Visser VL, Ristori T, Bouten CVC, Sahlgren CM, Loerakker S. Notch signaling regulates strain-mediated phenotypic switching of vascular smooth muscle cells. Front Cell Dev Biol 2022; 10:910503. [PMID: 36036000 PMCID: PMC9412035 DOI: 10.3389/fcell.2022.910503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/11/2022] [Indexed: 11/27/2022] Open
Abstract
Mechanical stimuli experienced by vascular smooth muscle cells (VSMCs) and mechanosensitive Notch signaling are important regulators of vascular growth and remodeling. However, the interplay between mechanical cues and Notch signaling, and its contribution to regulate the VSMC phenotype are still unclear. Here, we investigated the role of Notch signaling in regulating strain-mediated changes in VSMC phenotype. Synthetic and contractile VSMCs were cyclically stretched for 48 h to determine the temporal changes in phenotypic features. Different magnitudes of strain were applied to investigate its effect on Notch mechanosensitivity and the phenotypic regulation of VSMCs. In addition, Notch signaling was inhibited via DAPT treatment and activated with immobilized Jagged1 ligands to understand the role of Notch on strain-mediated phenotypic changes of VSMCs. Our data demonstrate that cyclic strain induces a decrease in Notch signaling along with a loss of VSMC contractile features. Accordingly, the activation of Notch signaling during cyclic stretching partially rescued the contractile features of VSMCs. These findings demonstrate that Notch signaling has an important role in regulating strain-mediated phenotypic switching of VSMCs.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mark C. van Turnhout
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Valery L. Visser
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Carlijn V. C. Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Cecilia M. Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- *Correspondence: Sandra Loerakker,
| |
Collapse
|
14
|
Mechanisms underlying the effects of caloric restriction on hypertension. Biochem Pharmacol 2022; 200:115035. [DOI: 10.1016/j.bcp.2022.115035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 11/20/2022]
|
15
|
Maselli D, Garoffolo G, Cassanmagnago GA, Vono R, Ruiter MS, Thomas AC, Madeddu P, Pesce M, Spinetti G. Mechanical Strain Induces Transcriptomic Reprogramming of Saphenous Vein Progenitors. Front Cardiovasc Med 2022; 9:884031. [PMID: 35711359 PMCID: PMC9197233 DOI: 10.3389/fcvm.2022.884031] [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: 03/02/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
Intimal hyperplasia is the leading cause of graft failure in aortocoronary bypass grafts performed using human saphenous vein (SV). The long-term consequences of the altered pulsatile stress on the cells that populate the vein wall remains elusive, particularly the effects on saphenous vein progenitors (SVPs), cells resident in the vein adventitia with a relatively wide differentiation capacity. In the present study, we performed global transcriptomic profiling of SVPs undergoing uniaxial cyclic strain in vitro. This type of mechanical stimulation is indeed involved in the pathology of the SV. Results showed a consistent stretch-dependent gene regulation in cyclically strained SVPs vs. controls, especially at 72 h. We also observed a robust mechanically related overexpression of Adhesion Molecule with Ig Like Domain 2 (AMIGO2), a cell surface type I transmembrane protein involved in cell adhesion. The overexpression of AMIGO2 in stretched SVPs was associated with the activation of the transforming growth factor β pathway and modulation of intercellular signaling, cell-cell, and cell-matrix interactions. Moreover, the increased number of cells expressing AMIGO2 detected in porcine SV adventitia using an in vivo arterialization model confirms the upregulation of AMIGO2 protein by the arterial-like environment. These results show that mechanical stress promotes SVPs' molecular phenotypic switching and increases their responsiveness to extracellular environment alterations, thus prompting the targeting of new molecular effectors to improve the outcome of bypass graft procedure.
Collapse
Affiliation(s)
- Davide Maselli
- IRCCS MultiMedica, Milan, Italy
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giada Andrea Cassanmagnago
- IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | - Matthijs S. Ruiter
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Anita C. Thomas
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gaia Spinetti
- IRCCS MultiMedica, Milan, Italy
- *Correspondence: Gaia Spinetti
| |
Collapse
|
16
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [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: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
17
|
Pozzo CFSD, Sielski MS, de Campos Vidal B, Werneck CC, Vicente CP. A collagen I derived matricryptin increases aorta vascular wall remodeling after induced thrombosis in mouse. Thromb Res 2021; 209:59-68. [PMID: 34871983 DOI: 10.1016/j.thromres.2021.11.021] [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: 06/27/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 11/27/2022]
Abstract
Matricryptins are collagen fragments proteolytically released from the extracellular matrix (ECM) with biological activity that can regulate several processes involved in ECM remodeling. Vessel wall matrix reorganization after lesion is important to the recovery of vascular function. This study aimed to analyze the effect of the peptide p1158/59 (Lindsey, 2015) on thrombosis, neointimal formation, and vascular remodeling of C57BL6 mice abdominal aorta. We used a FeCl3 induced vascular injury mice model and analyzed thrombus size, neointima formation, gelatinase activities in situ, re-endothelization, and collagen fibers organization on the arterial wall using polarization microscopy. As result, we observed that 2 days after injury the treatment with p1158/59 increased thrombus size and gelatinase activity, vascular lesion and it did not recover the endothelium loss induced by the chemical injury. We also observed that the peptide increased neointima growth and collagen birefringence, indicating collagen fibers reorganization. It also promoted increased re-endothelization and decreased activity of gelatinases 14 days after injury. Thus, we conclude that the peptide p1158/59 impaired the initial thrombosis recovery 2 days after injury but was able to induce vascular ECM remodeling after 14 days, improving vessel re-endothelization, collagen fibers deposition, and organization.
Collapse
Affiliation(s)
| | - Micheli Severo Sielski
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Benedicto de Campos Vidal
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Claudio C Werneck
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Cristina Pontes Vicente
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
18
|
Zhu J, Yang G. H 2S signaling and extracellular matrix remodeling in cardiovascular diseases: A tale of tense relationship. Nitric Oxide 2021; 116:14-26. [PMID: 34428564 DOI: 10.1016/j.niox.2021.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network that not only provides mechanical support but also transduces essential molecular signals in organ functions. ECM is constantly remodeled to control tissue homeostasis, responsible for cell adhesion, cell migration, cell-to-cell communication, and cell differentiation, etc. The dysregulation of ECM components contributes to various diseases, including cardiovascular diseases, fibrosis, cancer, and neurodegenerative diseases, etc. Aberrant ECM remodeling is initiated by various stress, such as oxidative stress, inflammation, ischemia, and mechanical stress, etc. Hydrogen sulfide (H2S) is a gasotransmitter that exhibits a wide variety of cytoprotective and physiological functions through its anti-oxidative and anti-inflammatory actions. Amounting research shows that H2S can attenuate aberrant ECM remodeling. In this review, we discussed the implications and mechanisms of H2S in the regulation of ECM remodeling in cardiovascular diseases, and highlighted the potential of H2S in the prevention and treatment of cardiovascular diseases through attenuating adverse ECM remodeling.
Collapse
Affiliation(s)
- Jiechun Zhu
- School of Biological, Chemical & Forensic Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Guangdong Yang
- School of Biological, Chemical & Forensic Sciences, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada.
| |
Collapse
|
19
|
Tierney JW, Evans BC, Cheung-Flynn J, Wang B, Colazo JM, Polcz ME, Cook RS, Brophy CM, Duvall CL. Therapeutic MK2 inhibition blocks pathological vascular smooth muscle cell phenotype switch. JCI Insight 2021; 6:e142339. [PMID: 34622803 PMCID: PMC8525639 DOI: 10.1172/jci.insight.142339] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Vascular procedures, such as stenting, angioplasty, and bypass grafting, often fail due to intimal hyperplasia (IH), wherein contractile vascular smooth muscle cells (VSMCs) dedifferentiate to synthetic VSMCs, which are highly proliferative, migratory, and fibrotic. Previous studies suggest MAPK-activated protein kinase 2 (MK2) inhibition may limit VSMC proliferation and IH, although the molecular mechanism underlying the observation remains unclear. We demonstrated here that MK2 inhibition blocked the molecular program of contractile to synthetic dedifferentiation and mitigated IH development. Molecular markers of the VSMC contractile phenotype were sustained over time in culture in rat primary VSMCs treated with potent, long-lasting MK2 inhibitory peptide nanopolyplexes (MK2i-NPs), a result supported in human saphenous vein specimens cultured ex vivo. RNA-Seq of MK2i-NP-treated primary human VSMCs revealed programmatic switching toward a contractile VSMC gene expression profile, increasing expression of antiinflammatory and contractile-associated genes while lowering expression of proinflammatory, promigratory, and synthetic phenotype-associated genes. Finally, these results were confirmed using an in vivo rabbit vein graft model where brief, intraoperative treatment with MK2i-NPs decreased IH and synthetic phenotype markers while preserving contractile proteins. These results support further development of MK2i-NPs as a therapy for blocking VSMC phenotype switch and IH associated with cardiovascular procedures.
Collapse
Affiliation(s)
- J. William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Brian C. Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bo Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Monica E. Polcz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of General Surgery and
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Colleen M. Brophy
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
20
|
Role of Vascular Smooth Muscle Cell Phenotype Switching in Arteriogenesis. Int J Mol Sci 2021; 22:ijms221910585. [PMID: 34638923 PMCID: PMC8508942 DOI: 10.3390/ijms221910585] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Arteriogenesis is one of the primary physiological means by which the circulatory collateral system restores blood flow after significant arterial occlusion in peripheral arterial disease patients. Vascular smooth muscle cells (VSMCs) are the predominant cell type in collateral arteries and respond to altered blood flow and inflammatory conditions after an arterial occlusion by switching their phenotype between quiescent contractile and proliferative synthetic states. Maintaining the contractile state of VSMC is required for collateral vascular function to regulate blood vessel tone and blood flow during arteriogenesis, whereas synthetic SMCs are crucial in the growth and remodeling of the collateral media layer to establish more stable conduit arteries. Timely VSMC phenotype switching requires a set of coordinated actions of molecular and cellular mediators to result in an expansive remodeling of collaterals that restores the blood flow effectively into downstream ischemic tissues. This review overviews the role of VSMC phenotypic switching in the physiological arteriogenesis process and how the VSMC phenotype is affected by the primary triggers of arteriogenesis such as blood flow hemodynamic forces and inflammation. Better understanding the role of VSMC phenotype switching during arteriogenesis can identify novel therapeutic strategies to enhance revascularization in peripheral arterial disease.
Collapse
|
21
|
Dynamic Crosstalk between Vascular Smooth Muscle Cells and the Aged Extracellular Matrix. Int J Mol Sci 2021; 22:ijms221810175. [PMID: 34576337 PMCID: PMC8468233 DOI: 10.3390/ijms221810175] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/15/2023] Open
Abstract
Vascular aging is accompanied by the fragmentation of elastic fibers and collagen deposition, leading to reduced distensibility and increased vascular stiffness. A rigid artery facilitates elastin to degradation by MMPs, exposing vascular cells to greater mechanical stress and triggering signaling mechanisms that only exacerbate aging, creating a self-sustaining inflammatory environment that also promotes vascular calcification. In this review, we highlight the role of crosstalk between smooth muscle cells and the vascular extracellular matrix (ECM) and how aging promotes smooth muscle cell phenotypes that ultimately lead to mechanical impairment of aging arteries. Understanding the underlying mechanisms and the role of associated changes in ECM during aging may contribute to new approaches to prevent or delay arterial aging and the onset of cardiovascular diseases.
Collapse
|
22
|
Ragno M, Pianese L, Caulo M, Logullo F, Angelini M, Incensi A, Liguori R, Fortunato A, Federico A, Trojano L, Donadio V. Cutaneous Sensory and Autonomic Small Fiber Neuropathy in HTRA1-Related Cerebral Small Vessel Disease. J Neuropathol Exp Neurol 2021; 80:713-716. [PMID: 33313782 DOI: 10.1093/jnen/nlaa150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Michele Ragno
- Division of Neurology, Ospedale Madonna del Soccorso, ASUR Marche, San Benedetto del Tronto-Ascoli Piceno, Italy
| | - Luigi Pianese
- U.O.C. Patologia Clinica, ASUR MARCHE AV5, Ascoli Piceno, Italy
| | - Massimo Caulo
- Department of Neuroscience and Clinical Sciences, University of Chieti, Italy
| | | | - Mario Angelini
- U.O.C. Ematologia e Terapia Cellulare, ASUR MARCHE AV5, Ascoli Piceno, Italy
| | - Alex Incensi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | | | - Antonio Federico
- Department of Medicine, Surgery and Neurosciences, Medical School, University of Siena, Siena, Italy
| | - Luigi Trojano
- Department of Psychology, University of Campania 'Luigi Vanvitelli', Caserta, Italy
| | - Vincenzo Donadio
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| |
Collapse
|
23
|
Tropea T, Mandalà M. Caloric restriction enhances vascular tone of cerebral and mesenteric resistance arteries in aged rats. Mech Ageing Dev 2021; 197:111520. [PMID: 34129890 DOI: 10.1016/j.mad.2021.111520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 11/27/2022]
Abstract
Vascular changes of tone and biomechanical properties induced by ageing increase the risk for cardiovascular diseases. Caloric restriction (CR) has been shown to protect against cardiovascular diseases and improve endothelial dysfunction in cerebral resistance arteries. We hypothesise that CR will enhance vascular tone and structural properties of cerebral resistance arteries and exert comparable beneficial effects on the systemic vasculature of aged rat model. Eighteen-month-old male Sprague-Dawley rats were feed either ad libitum or restricted to 60 % of calorie consumption up to 24 months of age, when body weight (BW) measurements were taken and functional and structural properties of resistance arteries were assessed using a pressure myograph. In cerebral arteries, CR increased myogenic tone (p < 0.001) and distensibility (p < 0.01) in response to intraluminal pressure and concentration-dependent constriction to KCl (p < 0.001). In mesenteric arteries constriction in response to KCl was increased (p < 0.0001) and wall thickness reduced (p < 0.01) in CR rats. BW was reduced (p < 0.0001) in FR rats. Our findings demonstrate that CR improves vascular tone of resistance arteries regardless the type of stimulus and independently of the vascular bed. CR may be a beneficial dietary approach to prevent age-related vascular diseases.
Collapse
Affiliation(s)
- Teresa Tropea
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, United Kingdom; Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy
| | - Maurizio Mandalà
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy; Department of Obstetrics, Gynecology and Reproductive Science, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
24
|
Corrêa JWN, Boaro KR, Sene LB, Polidoro JZ, Salles TA, Martins FL, Bendhack LM, Girardi ACC. Antiproteinuric and Hyperkalemic Mechanisms Activated by Dual Versus Single Blockade of the RAS in Renovascular Hypertensive Rats. Front Physiol 2021; 12:656460. [PMID: 34177612 PMCID: PMC8221266 DOI: 10.3389/fphys.2021.656460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022] Open
Abstract
This study aimed to investigate the antiproteinuric and hyperkalemic mechanisms activated by dual renin-angiotensin system (RAS) blockade in renovascular hypertensive rats (2-kidney 1-clip model [2K-1C]). Six weeks after clipping the left renal artery or sham operation (2K), rats were treated with losartan, enalapril, or both drugs for two weeks. We found that 2K-1C rats displayed higher tail-cuff blood pressure (BP), increased non-clipped kidney Ang II concentration, and more pronounced urinary albumin excretion than 2K. BP was decreased by the treatment with either enalapril or losartan, and the combination of both drugs promoted an additional antihypertensive effect in 2K-1C rats. Renal Ang II content and albuminuria were reduced by either enalapril or losartan in monotherapy and restored to control levels by dual RAS blockade. Albuminuria in 2K-1C rats was accompanied by downregulation of the glomerular slit protein podocin, reduction of the endocytic receptors megalin and cubilin, and a marked decrease in the expression of the ClC-5 chloride channel, compared to 2K animals. Treatment with losartan and enalapril in monotherapy or combination increased the expression of podocin, cubilin, and ClC-5. However, only the combined therapy normalized podocin, cubilin, and ClC-5 protein abundance in the non-clipped kidney of 2K-1C rats. Renovascular hypertensive 2K-1C rats had a lower concentration of plasma potassium compared to 2K rats. Single RAS blockade normalized potassium plasma concentration, whereas 2K-1C rats treated with dual RAS blockade exhibited hyperkalemia. Hypokalemia in 2K-1C rats was accompanied by an increase in the cleaved activated forms of α-ENaC and γ-ENaC and the expression of β-ENaC. Combined RAS blockade but not monotherapy significantly reduced the expression of these ENaC subunits in 2K-1C rats. Indeed, double RAS blockade reduced the abundance of cleaved-α-ENaC to levels lower than those of 2K rats. Collectively, these results demonstrate that the antiproteinuric effect of dual RAS blockade in 2K-1C rats is associated with the restored abundance of podocin and cubilin, and ClC-5. Moreover, double RAS blockade-induced hyperkalemia may be due, at least partially, to an exaggerated downregulation of cleaved α-ENaC in the non-clipped kidney of renovascular hypertensive rats.
Collapse
Affiliation(s)
- José Wilson N Corrêa
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil.,Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Amazonas, Manaus, Brazil
| | - Karoline R Boaro
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Letícia B Sene
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Juliano Z Polidoro
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Thiago A Salles
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Flavia L Martins
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Lusiane M Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Adriana C C Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
25
|
Garoffolo G, Pesce M. Vascular dysfunction and pathology: focus on mechanical forces. VASCULAR BIOLOGY 2021; 3:R69-R75. [PMID: 34291191 PMCID: PMC8284946 DOI: 10.1530/vb-21-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/09/2021] [Indexed: 11/08/2022]
Abstract
The role of mechanical forces is emerging as a new player in the pathophysiologic programming of the cardiovascular system. The ability of the cells to 'sense' mechanical forces does not relate only to perception of movement or flow, as intended traditionally, but also to the biophysical properties of the extracellular matrix, the geometry of the tissues, and the force distribution inside them. This is also supported by the finding that cells can actively translate mechanical cues into discrete gene expression and epigenetic programming. In the present review, we will contextualize these new concepts in the vascular pathologic programming.
Collapse
Affiliation(s)
- Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, Milan, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, Milan, Italy
| |
Collapse
|
26
|
Al-Maslamani NA, Khilan AA, Horn HF. Design of a 3D printed, motorized, uniaxial cell stretcher for microscopic and biochemical analysis of mechanotransduction. Biol Open 2021; 10:bio057778. [PMID: 33563607 PMCID: PMC7888744 DOI: 10.1242/bio.057778] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/13/2021] [Indexed: 12/14/2022] Open
Abstract
Cells respond to mechanical cues from their environment through a process of mechanosensing and mechanotransduction. Cell stretching devices are important tools to study the molecular pathways responsible for cellular responses to mechanobiological processes. We describe the development and testing of a uniaxial cell stretcher that has applications for microscopic as well as biochemical analyses. By combining simple fabrication techniques with adjustable control parameters, the stretcher is designed to fit a variety of experimental needs. The stretcher can be used for static and cyclic stretching. As a proof of principle, we visualize stretch induced deformation of cell nuclei via incremental static stretch, and changes in IEX1 expression via cyclic stretching. This stretcher is easily modified to meet experimental needs, inexpensive to build, and should be readily accessible for most laboratories with access to 3D printing.
Collapse
Affiliation(s)
- Noor A Al-Maslamani
- Biological and Biomedical Sciences Division, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| | - Abdulghani A Khilan
- Biological and Biomedical Sciences Division, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| | - Henning F Horn
- Biological and Biomedical Sciences Division, College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| |
Collapse
|
27
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
28
|
Latorre M, Spronck B, Humphrey JD. Complementary roles of mechanotransduction and inflammation in vascular homeostasis. Proc Math Phys Eng Sci 2021; 477:20200622. [PMID: 33642928 PMCID: PMC7897647 DOI: 10.1098/rspa.2020.0622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022] Open
Abstract
Arteries are exposed to relentless pulsatile haemodynamic loads, but via mechanical homeostasis they tend to maintain near optimal structure, properties and function over long periods in maturity in health. Numerous insults can compromise such homeostatic tendencies, however, resulting in maladaptations or disease. Chronic inflammation can be counted among the detrimental insults experienced by arteries, yet inflammation can also play important homeostatic roles. In this paper, we present a new theoretical model of complementary mechanobiological and immunobiological control of vascular geometry and composition, and thus properties and function. We motivate and illustrate the model using data for aortic remodelling in a common mouse model of induced hypertension. Predictions match the available data well, noting a need for increased data for further parameter refinement. The overall approach and conclusions are general, however, and help to unify two previously disparate literatures, thus leading to deeper insight into the separate and overlapping roles of mechanobiology and immunobiology in vascular health and disease.
Collapse
Affiliation(s)
- Marcos Latorre
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA,Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA,e-mail:
| |
Collapse
|
29
|
Humphrey JD. Mechanisms of Vascular Remodeling in Hypertension. Am J Hypertens 2020; 34:432-441. [PMID: 33245319 PMCID: PMC8140657 DOI: 10.1093/ajh/hpaa195] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Hypertension is both a cause and a consequence of central artery stiffening, which in turn is an initiator and indicator of myriad disease conditions and thus all-cause mortality. Such stiffening results from a remodeling of the arterial wall that is driven by mechanical stimuli and mediated by inflammatory signals, which together lead to differential gene expression and concomitant changes in extracellular matrix composition and organization. This review focuses on biomechanical mechanisms by which central arteries remodel in hypertension within the context of homeostasis-what promotes it, what prevents it. It is suggested that the vasoactive capacity of the wall and inflammatory burden strongly influence the ability of homeostatic mechanisms to adapt the arterial wall to high blood pressure or not. Maladaptation, often reflected by inflammation-driven adventitial fibrosis, not just excessive intimal-medial thickening, significantly diminishes central artery function and disturbs hemodynamics, ultimately compromising end organ perfusion and thus driving the associated morbidity and mortality. It is thus suggested that there is a need for increased attention to controlling both smooth muscle phenotype and inflammation in hypertensive remodeling of central arteries, with future studies of the often adaptive response of medium-sized muscular arteries promising to provide additional guidance.
Collapse
Affiliation(s)
- Jay D Humphrey
- Department of Biomedical Engineering, Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA,Correspondence: Jay D. Humphrey ()
| |
Collapse
|
30
|
Davaapil H, Shetty DK, Sinha S. Aortic "Disease-in-a-Dish": Mechanistic Insights and Drug Development Using iPSC-Based Disease Modeling. Front Cell Dev Biol 2020; 8:550504. [PMID: 33195187 PMCID: PMC7655792 DOI: 10.3389/fcell.2020.550504] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/08/2020] [Indexed: 12/24/2022] Open
Abstract
Thoracic aortic diseases, whether sporadic or due to a genetic disorder such as Marfan syndrome, lack effective medical therapies, with limited translation of treatments that are highly successful in mouse models into the clinic. Patient-derived induced pluripotent stem cells (iPSCs) offer the opportunity to establish new human models of aortic diseases. Here we review the power and potential of these systems to identify cellular and molecular mechanisms underlying disease and discuss recent advances, such as gene editing, and smooth muscle cell embryonic lineage. In particular, we discuss the practical aspects of vascular smooth muscle cell derivation and characterization, and provide our personal insights into the challenges and limitations of this approach. Future applications, such as genotype-phenotype association, drug screening, and precision medicine are discussed. We propose that iPSC-derived aortic disease models could guide future clinical trials via “clinical-trials-in-a-dish”, thus paving the way for new and improved therapies for patients.
Collapse
Affiliation(s)
- Hongorzul Davaapil
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| | - Deeti K Shetty
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| |
Collapse
|
31
|
Jafarihaghighi F, Ardjmand M, Mirzadeh A, Hassani MS, Parizi SS. Current challenges and future trends in manufacturing small diameter artificial vascular grafts in bioreactors. Cell Tissue Bank 2020; 21:377-403. [PMID: 32415569 DOI: 10.1007/s10561-020-09837-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/09/2020] [Indexed: 01/17/2023]
Abstract
Cardiovascular diseases are a leading cause of death. Vascular surgery is mainly used to solve this problem. However, the generation of a functional and suitable substitute for small diameter (< 6 mm) displacement is challengeable. Moreover, synthetic prostheses, made of polyethylene terephthalate and extended polytetrafluoroethylene show have shown insufficient performance. Therefore, the challenges dominating the use of autografts have prevented their efficient use. Tissue engineering is highlighted in regenerative medicine perhaps in aiming to address the issue of end-stage organ failure. While organs and complex tissues require the vascular supply to support the graft survival and render the bioartificial organ role, vascular tissue engineering has shown to be a hopeful method for cell implantation by the production of tissues in vitro. Bioreactors are a salient point in vascular tissue engineering due to the capability for reproducible and controlled variations showing a new horizon in blood vessel substitution. This review strives to display the overview of current concepts in the development of small-diameter by using bioreactors. In this work, we show a critical look at different factors for developing small-diameter and give suggestions for future studies.
Collapse
Affiliation(s)
- Farid Jafarihaghighi
- Department of Chemical Engineering, South Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mehdi Ardjmand
- Department of Chemical Engineering, South Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Abolfazl Mirzadeh
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Mohammad Salar Hassani
- Department of Chemical Engineering, South Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Shahriar Salemi Parizi
- Department of Chemical Engineering, South Tehran Branch, Islamic Azad University, Tehran, Iran
- Young Researchers and Elite Club, South Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
32
|
Garoffolo G, Ruiter MS, Piola M, Brioschi M, Thomas AC, Agrifoglio M, Polvani G, Coppadoro L, Zoli S, Saccu C, Spinetti G, Banfi C, Fiore GB, Madeddu P, Soncini M, Pesce M. Coronary artery mechanics induces human saphenous vein remodelling via recruitment of adventitial myofibroblast-like cells mediated by Thrombospondin-1. Am J Cancer Res 2020; 10:2597-2611. [PMID: 32194822 PMCID: PMC7052885 DOI: 10.7150/thno.40595] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/22/2019] [Indexed: 12/27/2022] Open
Abstract
Rationale: Despite the preferred application of arterial conduits, the greater saphenous vein (SV) remains indispensable for coronary bypass grafting (CABG), especially in multi-vessel coronary artery disease (CAD). The objective of the present work was to address the role of mechanical forces in the activation of maladaptive vein bypass remodeling, a process determining progressive occlusion and recurrence of ischemic heart disease. Methods: We employed a custom bioreactor to mimic the coronary shear and wall mechanics in human SV vascular conduits and reproduce experimentally the biomechanical conditions of coronary grafting and analyzed vein remodeling process by histology, histochemistry and immunofluorescence. We also subjected vein-derived cells to cyclic uniaxial mechanical stimulation in culture, followed by phenotypic and molecular characterization using RNA and proteomic methods. We finally validated our results in vitro and using a model of SV carotid interposition in pigs. Results: Exposure to pulsatile flow determined a remodeling process of the vascular wall involving reduction in media thickness. Smooth muscle cells (SMCs) underwent conversion from contractile to synthetic phenotype. A time-dependent increase in proliferating cells expressing mesenchymal (CD44) and early SMC (SM22α) markers, apparently recruited from the SV adventitia, was observed especially in CABG-stimulated vessels. Mechanically stimulated SMCs underwent transition from contractile to synthetic phenotype. MALDI-TOF-based secretome analysis revealed a consistent release of Thrombospondin-1 (TSP-1), a matricellular protein involved in TGF-β-dependent signaling. TSP-1 had a direct chemotactic effect on SV adventitia resident progenitors (SVPs); this effects was inhibited by blocking TSP-1 receptor CD47. The involvement of TSP-1 in adventitial progenitor cells differentiation and graft intima hyperplasia was finally contextualized in the TGF-β-dependent pathway, and validated in a saphenous vein into carotid interposition pig model. Conclusions: Our results provide the evidence of a matricellular mechanism involved in the human vein arterialization process controlled by alterations in tissue mechanics, and open the way to novel potential strategies to block VGD progression based on targeting cell mechanosensing-related effectors.
Collapse
|
33
|
Robbins AB, Freed AD, Moreno MR. Characterizing the non-linear mechanical behavior of native and biomimetic engineered tissues in 1D with physically meaningful parameters. J Mech Behav Biomed Mater 2020; 102:103509. [DOI: 10.1016/j.jmbbm.2019.103509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 01/06/2023]
|
34
|
Familial Aortopathies - State of the Art Review. Heart Lung Circ 2019; 29:607-618. [PMID: 32067919 DOI: 10.1016/j.hlc.2019.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/18/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
Aortopathies are conditions that result in aortic dilatation, aneurysm formation and dissection. Familial aortopathies (perhaps better known as heritable thoracic aortic aneurysm and dissection, h-TAAD, as not all have a positive family history) are recognised to have an underlying genetic cause and affect the aorta, predisposing it to the above pathologies. These conditions can also affect the extra-aortic vasculature, particularly large elastic arteries and other body systems. Mutations in a number of genes have been associated with h-TAAD. However, not all affected families have a pathogenic gene variant identified-highlighting the importance of a three-generational family history and the likely role of both environmental factors and future gene discoveries in furthering knowledge. Survival has improved over the last few decades, essentially due to surgical intervention. The benefit of identifying affected individuals depends upon a regular surveillance program and timely referral for surgery before complications such as dissection. Further research is required to appreciate fully the effects of individual gene variants and improve evidence for prophylactic medical therapy, as well as to understand the effect of h-TAAD on quality of life and life choices, particularly around exercise and pregnancy, for affected individuals. This will be complemented by laboratory-based research that seeks to understand the tissue pathways that underlie development of arterial pathology, ideally providing targets for novel medical therapies and a means of non-invasively identifying individuals at increased vascular risk to reduce dissection, which remains a devastating life-threatening event.
Collapse
|
35
|
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: 182] [Impact Index Per Article: 30.3] [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
|
36
|
Comparative study of variations in mechanical stress and strain of human blood vessels: mechanical reference for vascular cell mechano-biology. Biomech Model Mechanobiol 2019; 19:519-531. [PMID: 31494790 DOI: 10.1007/s10237-019-01226-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 08/31/2019] [Indexed: 10/26/2022]
Abstract
The diseases of human blood vessels are closely associated with local mechanical variations. A better understanding of the quantitative correlation in mechanical environment between the current mechano-biological studies and vascular physiological or pathological conditions in vivo is crucial for evaluating numerous existing results and exploring new factors for disease discovery. In this study, six representative human blood vessels with known experimental measurements were selected, and their stress and strain variations in vessel walls under different blood pressures were analyzed based on nonlinear elastic theory. The results suggest that conventional mechano-biological experiments seeking the different biological expressions of cells at high/low mechanical loadings are ambiguous as references for studying vascular diseases, because distinct "site-specific" characteristics appear in different vessels. The present results demonstrate that the inner surface of the vessel wall does not always suffer the most severe stretch under high blood pressures comparing to the outer surface. Higher tension on the outer surface of aortas supports the hypothesis of the outside-in inflammation dominated by aortic adventitial fibroblasts. These results indicate that cellular studies at different mechanical niches should be "disease-specific" as well. The present results demonstrate considerable stress gradients across the wall thickness, which indicate micro-scale mechanical variations existing around the vascular cells, and imply that the physiological or pathological changes are not static processes confined within isolated regions, but are coupled with dynamic cell behaviors such as migration. The results suggest that the stress gradients, as well as the mechanical stresses and strains, are key factors constituting the mechanical niches, which may shed new light on "factor-specific" experiments of vascular cell mechano-biology.
Collapse
|
37
|
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: 1.8] [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
|
38
|
Zeng XL, Sun L, Zheng HQ, Wang GL, Du YH, Lv XF, Ma MM, Guan YY. Smooth muscle-specific TMEM16A expression protects against angiotensin II-induced cerebrovascular remodeling via suppressing extracellular matrix deposition. J Mol Cell Cardiol 2019; 134:131-143. [PMID: 31301303 DOI: 10.1016/j.yjmcc.2019.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022]
Abstract
Cerebrovascular remodeling is the leading factor for stroke and characterized by increased extracellular matrix deposition, migration and proliferation of vascular smooth muscle cells, and inhibition of their apoptosis. TMEM16A is an important component of Ca2+-activated Cl- channels. Previously, we showed that downregulation of TMEM16A in the basilar artery was negatively correlated with cerebrovascular remodeling during hypertension. However, it is unclear whether TMEM16A participates in angiotensin II (Ang II)-induced vascular remodeling in mice that have TMEM16A gene modification. In this study, we generated a transgenic mouse that overexpresses TMEM16A specifically in vascular smooth muscle cells. We observed that vascular remodeling in the basilar artery during Ang II-induced hypertension was significantly suppressed upon vascular smooth muscle-specific overexpression of TMEM16A relative to control mice. Specifically, we observed a large reduction in the deposition of fibronectin and collagen I. The expression of matrix metalloproteinases (MMP-2, MMP-9, and MMP-14), and tissue inhibitors of metalloproteinases (TIMP-1 and TIMP-2) were upregulated in the basilar artery during Ang II-induced hypertension, but this was suppressed upon overexpression of TMEM16A in blood vessels. Furthermore, TMEM16A overexpression alleviated the overactivity of the canonical TGF-β1/Smad3, and non-canonical TGF-β1/ERK and JNK pathways in the basilar artery during Ang II-induced hypertension. These in vivo results were similar to the results derived in vitro with basilar artery smooth muscle cells stimulated by Ang II. Moreover, we observed that the inhibitory effect of TMEM16A on MMPs was mediated by decreasing the activation of WNK1, which is a Cl--sensitive serine/threonine kinase. In conclusion, this study demonstrates that TMEM16A protects against cerebrovascular remodeling during hypertension by suppressing extracellular matrix deposition. We also showed that TMEM16A exerts this effect by reducing the expression of MMPs via inhibiting WNK1, and decreasing the subsequent activities of TGF-β1/Smad3, ERK, and JNK. Accordingly, our results suggest that TMEM16A may serve as a novel therapeutic target for vascular remodeling.
Collapse
Affiliation(s)
- Xue-Lin Zeng
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Lu Sun
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Hua-Qing Zheng
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guan-Lei Wang
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Yan-Hua Du
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Xiao-Fei Lv
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Ming-Ming Ma
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Yong-Yuan Guan
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
39
|
Bax NAM, Duim SN, Kruithof BPT, Smits AM, Bouten CVC, Goumans MJ. In vivo and in vitro Approaches Reveal Novel Insight Into the Ability of Epicardium-Derived Cells to Create Their Own Extracellular Environment. Front Cardiovasc Med 2019; 6:81. [PMID: 31275946 PMCID: PMC6594358 DOI: 10.3389/fcvm.2019.00081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
Human epicardium-derived cells (hEPDCs) transplanted in the NOD-SCID mouse heart after myocardial infarction (MI) are known to improve cardiac function, most likely orchestrated by paracrine mechanisms that limit adverse remodeling. It is not yet known, however, if hEPDCs contribute to preservation of cardiac function via the secretion of matrix proteins and/or matrix proteases to reduce scar formation. This study describes the ability of hEPDCs to produce human collagen type I after transplantation into the infarct border zone, thereby creating their own extracellular environment. As the in vivo environment is too complex to investigate the mechanisms involved, we use an in vitro set-up, mimicking biophysical and biochemical cues from the myocardial tissue to unravel hEPDC-induced matrix remodeling. The in vivo contribution of hEPDCs to the cardiac extracellular matrix (ECM) was assessed in a historical dataset of the NOD-SCID murine model of experimentally induced MI and cell transplantation. Analysis showed that within 48 h after transplantation, hEPDCs produce human collagen type I. The build-up of the human collagen microenvironment was reversed within 6 weeks. To understand the hEPDCs response to the pathologic cardiac microenvironment, we studied the influence of cyclic straining and/or transforming growth beta (TGFβ) signaling in vitro. We revealed that 48 h of cyclic straining induced collagen type I production via the TGFβ/ALK5 signaling pathway. The in vitro approach enables further unraveling of the hEPDCs ability to secrete matrix proteins and matrix proteases and the potential to create and remodel the cardiac matrix in response to injury.
Collapse
Affiliation(s)
- Noortje A M Bax
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Sjoerd N Duim
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Boudewijn P T Kruithof
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Anke M Smits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Marie José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
40
|
Li N, He Y, Yang G, Yu Q, Li M. Role of TRPC1 channels in pressure-mediated activation of airway remodeling. Respir Res 2019; 20:91. [PMID: 31092255 PMCID: PMC6518742 DOI: 10.1186/s12931-019-1050-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Bronchoconstriction and cough, a characteristic of the asthmatic response, leads to development of compressive stresses in the airway wall. We hypothesized that progressively pathological high mechanical stress could act on mechanosensitive cation channels, such as transient receptor potential channel 1 (TRPC1) and then contributes to airway remodeling. METHODS We imitate the pathological airway pressure in vitro using cyclic stretch at 10 and 15% elongation. Ca2+ imaging was applied to measure the activity of TRPC1 after bronchial epithelial cells exposed to cyclic stretch for 0, 0.5, 1, 1.5, 2, 2.5 h. To further clarify the function of channnel TRPC1 in the process of mechano-transduction in airway remodeling, the experiment in vivo was implemented. The TRPC1 siRNA and budesonide were applied separately to asthmatic models. The morphological changes were measured by HE and Massion method. The expression levels of TRPC1 were evaluated by real-time PCR, western blot and immunohistochemistry. The protein expression level of IL-13, TGF-β1 and MMP-9 in BALF were measured by ELISA. RESULTS The result showed that cyclic stretch for 15% elongation at 1.5 h could maximize the activity of TRPC1 channel. This influx in Ca2+ was blocked by TRPC1 siRNA. Higher TRPC1 expression was observed in the bronchial epithelial layer of ovalbumin induced asthmatic models. The knockdown of TRPC1 with TRPC1 siRNA was associated with a hampered airway remodeling process, such as decreased bronchial wall thickness and smooth muscle hypertrophy/hyperplasia, a decreased ECM deposition area and inflammation infiltration around airway wall. Meantime, expression of IL-13, TGF-β1 and MMP-9 in OVA+TRPC1 siRNA also showed reduced level. TRPC1 intervention treatment showed similar anti-remodeling therapeutic effect with budesonide. CONCLUSIONS These results demonstrate that most TRPC1 channels expressed in bronchial epithelial cells mediate the mechanotransduction mechanism. TRPC1 inducing abnormal Ca2+ signal mediates receptor-stimulated and mechanical stimulus-induced airway remodeling. The inhibition of TRPC1 channel could produce similar therapeutic effect as glucocortisteroid to curb the development of asthmatic airway remodeling.
Collapse
Affiliation(s)
- Na Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Ye He
- Department of Geriatrics, Sichuan Provincial People’s Hospital, Sichuan Academy of Medical Science, Chengdu, Sichuan Province 610072 People’s Republic of China
| | - Gang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Qian Yu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Minchao Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| |
Collapse
|
41
|
Qiu C, Chen M, Yao J, Sun X, Xu J, Zhang R, Wang X, Li G, Qian S. Mechanical Strain Induces Distinct Human Scleral Fibroblast Lineages: Differential Roles in Cell Proliferation, Apoptosis, Migration, and Differentiation. Invest Ophthalmol Vis Sci 2019; 59:2401-2410. [PMID: 29847646 DOI: 10.1167/iovs.18-23855] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to explore the effect of mechanical strain on human scleral fibroblasts (HSFs) and compare cell behaviors of HSFs from distinct regions. Methods Primary HSFs were cultivated using a digestive protocol. Cells were seeded on collagen I-coated Bioflex plates, and a FX-5000 tension system was used to perform biaxial mechanical strain in vitro. We applied 10%, 0.5-Hz mechanical strain. Cell behaviors of peripapillary and periphery HSFs were compared after the strain. Edu imaging, Cell Counting Kit-8 assay, and cell cycle flow cytometry were conducted to analyze cell proliferation ability. For cell apoptosis, flow cytometry of Annexin V/propidium iodide, caspase 3 activity, and Western blot were performed. Immunofluorescence, real-time PCR, and Western blot were used to investigate cell differentiation. A migration assay was also performed. Results Under the mechanical strain of 10%, 0.5 Hz for 24 hours, the proliferation ability and cell apoptosis of peripapillary HSFs did not have a significant change. The expression of alpha-smooth muscle actin (α-SMA) slightly decreased. However, increased cell proliferation, attenuated cell apoptosis and more expression of α-SMA were shown in the periphery HSFs under the same condition. The migration rate was also increased for periphery HSFs, whereas it kept almost the same for peripapillary HSFs under 10%, 0.5-Hz strain for 8 hours. Conclusions Mechanical strain affected the cell behaviors of HSFs. The different performance of cells from distinct regions may suggest familial linages of HSFs, probably induced by mechanical strain.
Collapse
Affiliation(s)
- Chen Qiu
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Minjie Chen
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Yao
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinghuai Sun
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Jianjiang Xu
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Zhang
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Xin Wang
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gang Li
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shaohong Qian
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Rezvani-Sharif A, Tafazzoli-Shadpour M, Avolio A. Progressive changes of elastic moduli of arterial wall and atherosclerotic plaque components during plaque development in human coronary arteries. Med Biol Eng Comput 2018; 57:731-740. [PMID: 30374700 DOI: 10.1007/s11517-018-1910-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022]
Abstract
Stiffness of the arterial wall and atherosclerotic plaque components is a determinant of the stress field within plaques, which has been suggested to be an indicator of plaque vulnerability. The diversity and inhomogeneous structure of atherosclerotic lesions complicate the characterization of plaque components. In the present study, stiffness of the arterial wall and atherosclerotic plaque components in human coronary arteries was examined in early and developed atherosclerotic lesions. The force-spectroscopy mode of the atomic force microscope and histological examination were used for determination of elastic moduli at specified locations within samples. Fibrous cap (E = 14.1 ± 3.8 kPa) showed lower stiffness than the fibrous tissue beneath the lipid pool (E = 17.6 ± 3.2 kPa). Calcification zones (E = 96.1 ± 18.8 kPa) and lipid pools (E = 2.7 ± 1.8 kPa) were the stiffest and softest components of atherosclerotic lesions, respectively. The increase of media stiffness (%44.8) and reduction of the elastic modulus of the internal elastic lamina (%28.9) was observed in coronary arteries. Moreover, significant differences were observed between the stiffness of medial layer in diseased parts and free-plaque segments in incomplete plaques of coronary arteries. Our results can be used for better understanding of remodeling mechanisms of the arterial wall with plaque development. Graphical abstract Stiffness alteration of the arterial wall and atherosclerotic plaque components with plaque development in coronary arteries.
Collapse
Affiliation(s)
- Alireza Rezvani-Sharif
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Hafez Street, Tehran, Iran.,Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Alberto Avolio
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Hafez Street, Tehran, Iran
| |
Collapse
|
43
|
Iffrig E, Wilson JS, Zhong X, Oshinski JN. Demonstration of circumferential heterogeneity in displacement and strain in the abdominal aortic wall by spiral cine DENSE MRI. J Magn Reson Imaging 2018; 49:731-743. [PMID: 30295345 DOI: 10.1002/jmri.26304] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/30/2018] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Knowledge of tissue properties of the abdominal aorta can improve understanding of vascular disease and guide interventional approaches. Existing MRI methods to quantify aortic wall displacement and strain are unable to discern circumferential heterogeneity. PURPOSE To assess regional variation in abdominal aortic wall displacement and strain as a function of circumferential position using spiral cine displacement encoding with stimulated echoes (DENSE). STUDY TYPE Prospective. POPULATION Cardiovascular disease-free men (n = 8) and women (n = 9) ages 30-42. SEQUENCES Prospective electrocardiogram (ECG)-gated and navigator echo-gated spiral, cine 2D DENSE and retrospective ECG-gated phase contrast MR (PCMR) sequences at 3T. ASSESSMENT In-plane displacement values of the aortic wall acquired with DENSE were used to determine radial and circumferential aortic wall motion. A quadrilateral-based 2D strain calculation method was implemented to determine strain from the displacement field. Peak displacement and its radial and circumferential contributions as well as peak circumferential strain were compared among eight circumferential wall segments. Distensibility was calculated using PCMR and compared with homogenized circumferential strain. STATISTICAL TESTS To account for repeated measurements in volunteers, linear mixed models for mean sector values were created for displacement magnitude, circumferential displacement, radial displacement, and circumferential strain. Comparisons were made between sectors. Calculated distensibility and homogenized circumferential strain were compared using Bland-Altman analysis. Statistical significance was defined as P < 0.05. RESULTS Displacement was highest in the anterior wall (1.5 ± 0.7 mm) and was primarily in the radial as compared with circumferential direction (1.04 ± 0.05 mm vs. 0.81 ± 0.42 mm). Circumferential strain was highest in the lateral walls (left 0.16 ± 0.05 and right 0.21 ± 0.12) with homogenized circumferential strain of 0.14 ± 0.05. DATA CONCLUSION DENSE imaging in the abdominal aortic wall demonstrated that the anterior aortic wall exhibits the greatest displacement, while the lateral wall experiences the largest circumferential strain. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2019;49:731-743.
Collapse
Affiliation(s)
- Elizabeth Iffrig
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - John S Wilson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia
| | - Xiadong Zhong
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia
| | - John N Oshinski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia
| |
Collapse
|
44
|
Prim DA, Menon V, Hasanian S, Carter L, Shazly T, Potts JD, Eberth JF. Perfusion Tissue Culture Initiates Differential Remodeling of Internal Thoracic Arteries, Radial Arteries, and Saphenous Veins. J Vasc Res 2018; 55:255-267. [PMID: 30179877 DOI: 10.1159/000492484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 07/23/2018] [Indexed: 01/26/2023] Open
Abstract
Adaptive remodeling processes are essential to the maintenance and viability of coronary artery bypass grafts where clinical outcomes depend strongly on the tissue source. In this investigation, we utilized an ex vivo perfusion bioreactor to culture porcine analogs of common human bypass grafts: the internal thoracic artery (ITA), the radial artery (RA), and the great saphenous vein (GSV), and then evaluated samples acutely (6 h) and chronically (7 days) under in situ or coronary-like perfusion conditions. Although morphologically similar, primary cells harvested from the ITA illustrated lower intimal and medial, but not adventitial, cell proliferation rates than those from the RA or GSV. Basal gene expression levels were similar in all vessels, with only COL3A1, SERPINE1, FN1, and TGFB1 being differentially expressed prior to culture; however, over half of all genes were affected nominally by the culturing process. When exposed to coronary-like conditions, RAs and GSVs experienced pathological remodeling not present in ITAs or when vessels were studied in situ. Many of the remodeling genes perturbed at 6 h were restored after 7 days (COL3A1, FN1, MMP2, and TIMP1) while others (SERPINE1, TGFB1, and VCAM1) were not. The findings elucidate the potential mechanisms of graft failure and highlight strategies to encourage healthy ex vivo pregraft conditioning.
Collapse
Affiliation(s)
- David A Prim
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA
| | - Vinal Menon
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | - Shahd Hasanian
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA
| | - Laurel Carter
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | - Tarek Shazly
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA.,Mechanical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA
| | - Jay D Potts
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA.,Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | - John F Eberth
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, .,Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina,
| |
Collapse
|
45
|
Rezvani-Sharif A, Tafazzoli-Shadpour M, Avolio A. Mechanical Characterization of the Lamellar Structure of Human Abdominal Aorta in the Development of Atherosclerosis: An Atomic Force Microscopy Study. Cardiovasc Eng Technol 2018; 10:181-192. [PMID: 30006817 DOI: 10.1007/s13239-018-0370-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/08/2018] [Indexed: 12/16/2022]
Abstract
Atherosclerosis is a major risk factor for cardiovascular disease. However, mechanisms of interaction of atherosclerotic plaque development and local stiffness of the lamellar structure of the arterial wall are not well established. In the current study, the local Young's modulus of the wall and plaque components were determined for three different groups of healthy, mildly diseased and advanced atherosclerotic human abdominal aortas. Histological staining was performed to highlight the atherosclerotic plaque components and lamellar structure of the aortic media, consisting of concentric layers of elastin and interlamellar zones. The force spectroscopy mode of the atomic force microscopy was utilized to determine Young's moduli of aortic wall lamellae and plaque components at the micron level. The high variability of Young's moduli (E) at different locations of the atherosclerotic plaque such as the fibrous cap (E = 15.5± 2.6 kPa), calcification zone (E = 103.7±19.5 kPa), and lipid pool (E = 3.5±1.2 kPa) were observed. Reduction of elastin lamellae stiffness (18.6%), as well as stiffening of interlamellar zones (50%), were detected in the diseased portion of the medial layer of abdominal aortic wall compared to the healthy artery. Additionally, significant differences in the stiffness of both elastin lamellae and interlamellar zones were observed between the diseased wall and disease-free wall in incomplete plaques. Our results elucidate the alternation of the stiffness of different lamellae in the human abdominal aortic wall with atherosclerotic plaque development and may provide new insight on the remodeling of the aortic wall during the progression of atherosclerosis.
Collapse
Affiliation(s)
- Alireza Rezvani-Sharif
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.,Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | | | - Alberto Avolio
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
46
|
Bersi MR, Khosravi R, Wujciak AJ, Harrison DG, Humphrey JD. Differential cell-matrix mechanoadaptations and inflammation drive regional propensities to aortic fibrosis, aneurysm or dissection in hypertension. J R Soc Interface 2018; 14:rsif.2017.0327. [PMID: 29118111 DOI: 10.1098/rsif.2017.0327] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/17/2017] [Indexed: 01/01/2023] Open
Abstract
The embryonic lineage of intramural cells, microstructural organization of the extracellular matrix, local luminal and wall geometry, and haemodynamic loads vary along the length of the aorta. Yet, it remains unclear why certain diseases manifest differentially along the aorta. Toward this end, myriad animal models provide insight into diverse disease conditions-including fibrosis, aneurysm and dissection-but inherent differences across models impede general interpretations. We examined region-specific cellular, matrix, and biomechanical changes in a single experimental model of hypertension and atherosclerosis, which commonly coexist. Our findings suggest that (i) intramural cells within the ascending aorta are unable to maintain the intrinsic material stiffness of the wall, which ultimately drives aneurysmal dilatation, (ii) a mechanical stress-initiated, inflammation-driven remodelling within the descending aorta results in excessive fibrosis, and (iii) a transient loss of adventitial collagen within the suprarenal aorta contributes to dissection propensity. Smooth muscle contractility helps to control wall stress in the infrarenal aorta, which maintains mechanical properties near homeostatic levels despite elevated blood pressure. This early mechanoadaptation of the infrarenal aorta does not preclude subsequent acceleration of neointimal formation, however. Because region-specific conditions may be interdependent, as, for example, diffuse central arterial stiffening can increase cyclic haemodynamic loads on an aneurysm that is developing proximally, there is a clear need for more systematic assessments of aortic disease progression, not simply a singular focus on a particular region or condition.
Collapse
Affiliation(s)
- M R Bersi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - R Khosravi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - A J Wujciak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - D G Harrison
- Department of Medicine, Vanderbilt University, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA .,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
47
|
Xie X, Percipalle P. Elevated transforming growth factor β signaling activation in β-actin-knockout mouse embryonic fibroblasts enhances myofibroblast features. J Cell Physiol 2018; 233:8884-8895. [PMID: 29851084 PMCID: PMC6220129 DOI: 10.1002/jcp.26808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/30/2018] [Indexed: 01/08/2023]
Abstract
Signaling by the transforming growth factor‐β (TGF‐β) is an essential pathway regulating a variety of cellular events. TGF‐β is produced as a latent protein complex and is required to be activated before activating the receptor. The mechanical force at the cell surface is believed to be a mechanism for latent TGF‐β activation. Using β‐actin null mouse embryonic fibroblasts as a model, in which actin cytoskeleton and cell‐surface biophysical features are dramatically altered, we reveal increased TGF‐β1 activation and the upregulation of TGF‐β target genes. In β‐actin null cells, we show evidence that the enhanced TGF‐β signaling relies on the active utilization of latent TGF‐β1 in the cell culture medium. TGF‐β signaling activation contributes to the elevated reactive oxygen species production, which is likely mediated by the upregulation of Nox4. The previously observed myofibroblast phenotype of β‐actin null cells is inhibited by TGF‐β signaling inhibition, while the expression of actin cytoskeleton genes and angiogenic phenotype are not affected. Together, our study shows a scenario that the alteration of the actin cytoskeleton and the consequent changes in cellular biophysical features lead to changes in cell signaling process such as TGF‐β activation, which in turn contributes to the enhanced myofibroblast phenotype.
Collapse
Affiliation(s)
- Xin Xie
- Biology Program, Science Division, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Biology Program, Science Division, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
48
|
Ruiter MS, Pesce M. Mechanotransduction in Coronary Vein Graft Disease. Front Cardiovasc Med 2018; 5:20. [PMID: 29594150 PMCID: PMC5861212 DOI: 10.3389/fcvm.2018.00020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/22/2018] [Indexed: 12/19/2022] Open
Abstract
Autologous saphenous veins are the most commonly used conduits in revascularization of the ischemic heart by coronary artery bypass graft surgery, but are subject to vein graft failure. The current mini review aims to provide an overview of the role of mechanotransduction signalling underlying vein graft failure to further our understanding of the disease progression and to improve future clinical treatment. Firstly, limitation of damage during vein harvest and engraftment can improve outcome. In addition, cell cycle inhibition, stimulation of Nur77 and external grafting could form interesting therapeutic options. Moreover, the Hippo pathway, with the YAP/TAZ complex as the main effector, is emerging as an important node controlling conversion of mechanical signals into cellular responses. This includes endothelial cell inflammation, smooth muscle cell proliferation/migration, and monocyte attachment/infiltration. The combined effects of expression levels and nuclear/cytoplasmic translocation make YAP/TAZ interesting novel targets in the prevention and treatment of vein graft disease. Pharmacological, molecular and/or mechanical conditioning of saphenous vein segments between harvest and grafting may potentiate targeted and specific treatment to improve long-term outcome.
Collapse
Affiliation(s)
- Matthijs Steven Ruiter
- Cardiovascular Tissue Engineering Unit, Centro Cardiologico Monzino (IRCCS), Milan, Italy
| | - Maurizio Pesce
- Cardiovascular Tissue Engineering Unit, Centro Cardiologico Monzino (IRCCS), Milan, Italy
| |
Collapse
|
49
|
Minami K, Hayashi T, Sato K, Nakahara T. Development of micro mechanical device having two-dimensional array of micro chambers for cell stretching. Biomed Microdevices 2018; 20:10. [PMID: 29305659 DOI: 10.1007/s10544-017-0256-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This paper presents a novel cell stretching micro device having two-dimensional array of micro chambers. It enables an in situ time-lapse observation of stretched cell by using an optical microscope with high measurement efficiency. The presented device consists of a cell culture dish and the array of micro chambers made of silicone elastomer and extension structures made of photocurable resin, and is fabricated with MEMS technology. The fabrication process of the thin micro chamber array combines photoresist mold and lift-off process based on conventional photolithography. The fabricated device has 134micro chambers in 5μm or less thickness. It was demonstrated that the fabricated micro device could be used to make in-situ time-lapse observation of cell responses to stretching under optical microscopy. In addition, the influence of the chamber thickness to the quality of the microscope image observed was evaluated. It is confirmed that the proposed device having two-dimensional array of the thin micro chambers makes it possible to observe cell response for stretch stimuli with high quality and efficiency.
Collapse
Affiliation(s)
- K Minami
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, 2-6-1 Tokiwadai, Ube, Yamaguchi, 755-8611, Japan.
| | - T Hayashi
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, 2-6-1 Tokiwadai, Ube, Yamaguchi, 755-8611, Japan
| | - K Sato
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1 Minami Josanjima, Tokushima, Tokushima, 770-8506, Japan
| | - T Nakahara
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, 2-6-1 Tokiwadai, Ube, Yamaguchi, 755-8611, Japan
| |
Collapse
|
50
|
Serralheiro P, Soares A, Costa Almeida CM, Verde I. TGF-β1 in Vascular Wall Pathology: Unraveling Chronic Venous Insufficiency Pathophysiology. Int J Mol Sci 2017; 18:E2534. [PMID: 29186866 PMCID: PMC5751137 DOI: 10.3390/ijms18122534] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022] Open
Abstract
Chronic venous insufficiency and varicose veins occur commonly in affluent countries and are a socioeconomic burden. However, there remains a relative lack of knowledge about venous pathophysiology. Various theories have been suggested, yet the molecular sequence of events is poorly understood. Transforming growth factor-beta one (TGF-β1) is a highly complex polypeptide with multifunctional properties that has an active role during embryonic development, in adult organ physiology and in the pathophysiology of major diseases, including cancer and various autoimmune, fibrotic and cardiovascular diseases. Therefore, an emphasis on understanding its signaling pathways (and possible disruptions) will be an essential requirement for a better comprehension and management of specific diseases. This review aims at shedding more light on venous pathophysiology by describing the TGF-β1 structure, function, activation and signaling, and providing an overview of how this growth factor and disturbances in its signaling pathway may contribute to specific pathological processes concerning the vessel wall which, in turn, may have a role in chronic venous insufficiency.
Collapse
Affiliation(s)
- Pedro Serralheiro
- Norfolk and Norwich University Hospital, Colney Ln, Norwich NR47UY, UK.
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal.
| | - Andreia Soares
- Norfolk and Norwich University Hospital, Colney Ln, Norwich NR47UY, UK.
| | - Carlos M Costa Almeida
- Department of General Surgery (C), Coimbra University Hospital Centre, Portugal; Faculty of Medicine, University of Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal.
| | - Ignacio Verde
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal.
| |
Collapse
|