1
|
Liu SF, Kucherenko MM, Sang P, Li Q, Yao J, Nambiar Veetil N, Gransar T, Alesutan I, Voelkl J, Salinas G, Grune J, Simmons S, Knosalla C, Kuebler WM. RUNX2 is stabilised by TAZ and drives pulmonary artery calcification and lung vascular remodelling in pulmonary hypertension due to left heart disease. Eur Respir J 2024; 64:2300844. [PMID: 39542509 DOI: 10.1183/13993003.00844-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/13/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Calcification is common in chronic vascular disease, yet its role in pulmonary hypertension due to left heart disease is unknown. Here, we probed for the role of runt-related transcription factor-2 (RUNX2), a master transcription factor in osteogenesis, and its regulation by the HIPPO pathway transcriptional coactivator with PDZ-binding motif (TAZ) in the osteogenic reprogramming of pulmonary artery smooth muscle cells and vascular calcification in patients with pulmonary hypertension due to left heart disease. We similarly examined its role using an established rat model of pulmonary hypertension due to left heart disease induced by supracoronary aortic banding. METHODS Pulmonary artery samples were collected from patients and rats with pulmonary hypertension due to left heart disease. Genome-wide RNA sequencing was performed, and pulmonary artery calcification assessed. Osteogenic signalling via TAZ and RUNX2 was delineated by protein biochemistry. In vivo, the therapeutic potential of RUNX2 or TAZ inhibition by CADD522 or verteporfin was tested in the rat model. RESULTS Gene ontology term analysis identified significant enrichment in ossification and osteoblast differentiation genes, including RUNX2, in pulmonary arteries of patients and lungs of rats with pulmonary hypertension due to left heart disease. Pulmonary artery calcification was evident in both patients and rats. Both TAZ and RUNX2 were upregulated and activated in pulmonary artery smooth muscle cells of patients and rats. Co-immunoprecipitation revealed a direct interaction of RUNX2 with TAZ in pulmonary artery smooth muscle cells. TAZ inhibition or knockdown decreased RUNX2 abundance due to accelerated RUNX2 protein degradation rather than reduced de novo synthesis. Inhibition of either TAZ or RUNX2 attenuated pulmonary artery calcification, distal lung vascular remodelling and pulmonary hypertension development in the rat model. CONCLUSION Pulmonary hypertension due to left heart disease is associated with pulmonary artery calcification that is driven by TAZ-dependent stabilisation of RUNX2, causing osteogenic reprogramming of pulmonary artery smooth muscle cells. The TAZ-RUNX2 axis may present a therapeutic target in pulmonary hypertension due to left heart disease.
Collapse
Affiliation(s)
- Shao-Fei Liu
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Mariya M Kucherenko
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Pengchao Sang
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Qiuhua Li
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Juquan Yao
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Netra Nambiar Veetil
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Tara Gransar
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Ioana Alesutan
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Voelkl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Salinas
- NGS - Integrative Genomics Core Unit (NIG), Institute of Pathology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jana Grune
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Christoph Knosalla
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors share the last authorship
| | - Wolfgang M Kuebler
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- DZL (German Centre for Lung Research), partner site Berlin, Berlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, ON, Canada
- Departements of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
- These authors share the last authorship
| |
Collapse
|
2
|
Zhang XZ, Jiang YL, Hu JG, Zhao LM, Chen QZ, Liang Y, Zhang Y, Lei XX, Wang R, Lei Y, Zhang QY, Li-Ling J, Xie HQ. Procyanidins-crosslinked small intestine submucosa: A bladder patch promotes smooth muscle regeneration and bladder function restoration in a rabbit model. Bioact Mater 2021; 6:1827-1838. [PMID: 33336114 PMCID: PMC7721664 DOI: 10.1016/j.bioactmat.2020.11.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 02/05/2023] Open
Abstract
Currently the standard surgical treatment for bladder defects is augmentation cystoplasty with autologous tissues, which has many side effects. Biomaterials such as small intestine submucosa (SIS) can provide an alternative scaffold for the repair as bladder patches. Previous studies have shown that SIS could enhance the capacity and compliance of the bladder, but its application is hindered by issues like limited smooth muscle regeneration and stone formation since the fast degradation and poor mechanical properties of the SIS. Procyanidins (PC), a natural bio-crosslinking agent, has shown anti-calcification, anti-inflammatory and anti-oxidation properties. More importantly, PC and SIS can crosslink through hydrogen bonds, which may endow the material with enhanced mechanical property and stabilized functionalities. In this study, various concentrations of PC-crosslinked SIS (PC-SIS) were prepared to repair the full-thickness bladder defects, with an aim to reduce complications and enhance bladder functions. In vitro assays showed that the crosslinking has conferred the biomaterial with superior mechanical property and anti-calcification property, ability to promote smooth muscle cell adhesion and upregulate functional genes expression. Using a rabbit model with bladder defects, we demonstrated that the PC-SIS scaffold can rapidly promote in situ tissue regrowth and regeneration, in particular smooth muscle remodeling and improvement of urinary functions. The PC-SIS scaffold has therefore provided a promising material for the reconstruction of a functional bladder.
Collapse
Affiliation(s)
- Xiu-Zhen Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan-Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Jun-Gen Hu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Long-Mei Zhao
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Qiu-Zhu Chen
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan Liang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Zhang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiong-Xin Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Rui Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yi Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Jesse Li-Ling
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Meeremans M, Van de Walle GR, Van Vlierberghe S, De Schauwer C. The Lack of a Representative Tendinopathy Model Hampers Fundamental Mesenchymal Stem Cell Research. Front Cell Dev Biol 2021; 9:651164. [PMID: 34012963 PMCID: PMC8126669 DOI: 10.3389/fcell.2021.651164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Overuse tendon injuries are a major cause of musculoskeletal morbidity in both human and equine athletes, due to the cumulative degenerative damage. These injuries present significant challenges as the healing process often results in the formation of inferior scar tissue. The poor success with conventional therapy supports the need to search for novel treatments to restore functionality and regenerate tissue as close to native tendon as possible. Mesenchymal stem cell (MSC)-based strategies represent promising therapeutic tools for tendon repair in both human and veterinary medicine. The translation of tissue engineering strategies from basic research findings, however, into clinical use has been hampered by the limited understanding of the multifaceted MSC mechanisms of action. In vitro models serve as important biological tools to study cell behavior, bypassing the confounding factors associated with in vivo experiments. Controllable and reproducible in vitro conditions should be provided to study the MSC healing mechanisms in tendon injuries. Unfortunately, no physiologically representative tendinopathy models exist to date. A major shortcoming of most currently available in vitro tendon models is the lack of extracellular tendon matrix and vascular supply. These models often make use of synthetic biomaterials, which do not reflect the natural tendon composition. Alternatively, decellularized tendon has been applied, but it is challenging to obtain reproducible results due to its variable composition, less efficient cell seeding approaches and lack of cell encapsulation and vascularization. The current review will overview pros and cons associated with the use of different biomaterials and technologies enabling scaffold production. In addition, the characteristics of the ideal, state-of-the-art tendinopathy model will be discussed. Briefly, a representative in vitro tendinopathy model should be vascularized and mimic the hierarchical structure of the tendon matrix with elongated cells being organized in a parallel fashion and subjected to uniaxial stretching. Incorporation of mechanical stimulation, preferably uniaxial stretching may be a key element in order to obtain appropriate matrix alignment and create a pathophysiological model. Together, a thorough discussion on the current status and future directions for tendon models will enhance fundamental MSC research, accelerating translation of MSC therapies for tendon injuries from bench to bedside.
Collapse
Affiliation(s)
- Marguerite Meeremans
- Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Catharina De Schauwer
- Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
4
|
Chen J, Zhou Y, Liu S, Li C. Biomechanical signal communication in vascular smooth muscle cells. J Cell Commun Signal 2020; 14:357-376. [PMID: 32780323 DOI: 10.1007/s12079-020-00576-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Biomechanical stresses are closely associated with cardiovascular development and diseases. In vivo, vascular smooth muscle cells are constantly stimulated by biomechanical factors caused by increased blood pressure leading to the non-specific activation of cell transmembrane proteins. Thus, various intracellular signal molecules are simultaneously activated via signaling cascades, which are closely related to alterations in the differentiation, phenotype, inflammation, migration, pyroptosis, calcification, proliferation, and apoptosis of vascular smooth muscle cells. Meanwhile, mechanical stress-induced miRNAs and epigenetics modification on vascular smooth muscle cells play critical roles as well. Eventually, the overall pathophysiology of the cells is altered, resulting in the development of many major clinical diseases, including hypertension, atherosclerosis, grafted venous atherosclerosis, and aneurysm, among others. In this paper, important advances in mechanical signal communication in vascular smooth muscle cells are reviewed.
Collapse
Affiliation(s)
- Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Alesutan I, Voelkl J, Feger M, Kratschmar DV, Castor T, Mia S, Sacherer M, Viereck R, Borst O, Leibrock C, Gawaz M, Kuro-O M, Pilz S, Tomaschitz A, Odermatt A, Pieske B, Wagner CA, Lang F. Involvement Of Vascular Aldosterone Synthase In Phosphate-Induced Osteogenic Transformation Of Vascular Smooth Muscle Cells. Sci Rep 2017; 7:2059. [PMID: 28515448 PMCID: PMC5435689 DOI: 10.1038/s41598-017-01882-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/04/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification resulting from hyperphosphatemia is a major determinant of mortality in chronic kidney disease (CKD). Vascular calcification is driven by aldosterone-sensitive osteogenic transformation of vascular smooth muscle cells (VSMCs). We show that even in absence of exogenous aldosterone, silencing and pharmacological inhibition (spironolactone, eplerenone) of the mineralocorticoid receptor (MR) ameliorated phosphate-induced osteo-/chondrogenic transformation of primary human aortic smooth muscle cells (HAoSMCs). High phosphate concentrations up-regulated aldosterone synthase (CYP11B2) expression in HAoSMCs. Silencing and deficiency of CYP11B2 in VSMCs ameliorated phosphate-induced osteogenic reprogramming and calcification. Phosphate treatment was followed by nuclear export of APEX1, a CYP11B2 transcriptional repressor. APEX1 silencing up-regulated CYP11B2 expression and stimulated osteo-/chondrogenic transformation. APEX1 overexpression blunted the phosphate-induced osteo-/chondrogenic transformation and calcification of HAoSMCs. Cyp11b2 expression was higher in aortic tissue of hyperphosphatemic klotho-hypomorphic (kl/kl) mice than in wild-type mice. In adrenalectomized kl/kl mice, spironolactone treatment still significantly ameliorated aortic osteoinductive reprogramming. Our findings suggest that VSMCs express aldosterone synthase, which is up-regulated by phosphate-induced disruption of APEX1-dependent gene suppression. Vascular CYP11B2 may contribute to stimulation of VSMCs osteo-/chondrogenic transformation during hyperphosphatemia.
Collapse
Affiliation(s)
- Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Jakob Voelkl
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
| | - Martina Feger
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Denise V Kratschmar
- Department of Pharmaceutical Sciences, and the National Center for Excellence in Research NCCR Kidney.CH, University of Basel, Basel, Switzerland
| | - Tatsiana Castor
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Sobuj Mia
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Michael Sacherer
- Div. of Cardiology, Medical University of Graz and Ludwig Boltzmann Institute for Translational Heart Failure Research, Graz, Austria
| | - Robert Viereck
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | | | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Stefan Pilz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University of Graz, Graz, Austria
| | - Andreas Tomaschitz
- Div. of Cardiology, Medical University of Graz and Ludwig Boltzmann Institute for Translational Heart Failure Research, Graz, Austria
- Bad Gleichenberg Clinic, Bad Gleichenberg, Austria
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, and the National Center for Excellence in Research NCCR Kidney.CH, University of Basel, Basel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Department of Cardiology, University of Graz, Graz, Austria; Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, and the National Center for Excellence in Research NCCR Kidney, Zurich, Switzerland
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
6
|
Molostvov G, Hiemstra TF, Fletcher S, Bland R, Zehnder D. Arterial Expression of the Calcium-Sensing Receptor Is Maintained by Physiological Pulsation and Protects against Calcification. PLoS One 2015; 10:e0138833. [PMID: 26436544 PMCID: PMC4593585 DOI: 10.1371/journal.pone.0138833] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/03/2015] [Indexed: 12/19/2022] Open
Abstract
Vascular calcification (VC) is common in chronic kidney disease (CKD) and contributes to cardiovascular mortality. The calcium-sensing receptor (CaSR) is present in human artery, senses extracellular calcium and may directly modulate VC. Objective: to investigate the association between arterial cyclic strain, CaSR expression and VC. Methods and Results: human aortic smooth muscle cells (HAoSMC) were cultured under static or strained conditions, with exposure to CaSR agonists, the calcimimetic R568, and after CaSR silencing and over-expression. High extracellular calcium reduced CaSR expression and promoted osteochondrogenic transformation and calcium deposition. This was partially prevented by cyclic strain and exposure to R568. CaSR silencing enhanced calcification and osteochondrogenic transformation, whereas CaSR over-expression attenuated this procalcific response, demonstrating a central role for the CaSR in the response to cyclic strain and regulation of VC. In arterial explants from CKD patients (n = 11) and controls (n = 9), exposure to R568 did not significantly alter calcium deposition, osteochondrogenic markers or total artery calcium content. Conclusions: physiological mechanical strain is important for arterial homeostasis and may protect arteries from VC. The beneficial effects of cyclic strain may be mediated via the CaSR.
Collapse
MESH Headings
- Adult
- Aged
- Aorta/cytology
- Aorta/metabolism
- Calcium/metabolism
- Calcium/pharmacology
- Cells, Cultured
- Chondrogenesis/drug effects
- Core Binding Factor Alpha 1 Subunit/biosynthesis
- Core Binding Factor Alpha 1 Subunit/genetics
- Extracellular Matrix Proteins/biosynthesis
- Extracellular Matrix Proteins/genetics
- Female
- Gene Expression Regulation/drug effects
- Humans
- Male
- Middle Aged
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Osteoblasts/cytology
- Osteogenesis/drug effects
- Phenethylamines/pharmacology
- Phosphoproteins/biosynthesis
- Phosphoproteins/genetics
- Propylamines/pharmacology
- Pulsatile Flow/physiology
- Receptors, Calcium-Sensing/agonists
- Receptors, Calcium-Sensing/antagonists & inhibitors
- Receptors, Calcium-Sensing/genetics
- Receptors, Calcium-Sensing/physiology
- Recombinant Fusion Proteins/biosynthesis
- Stress, Mechanical
- Transfection
- Vascular Calcification/physiopathology
- Vascular Calcification/prevention & control
- Young Adult
Collapse
Affiliation(s)
- Guerman Molostvov
- The Clinical Sciences Research Laboratory, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Thomas F. Hiemstra
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Cambridge Clinical Trials Unit, Addenbrooke’s Hospital, Cambridge, United Kingdom
- * E-mail:
| | - Simon Fletcher
- Department of Nephrology, University Hospital Coventry and Warwickshire, Coventry, United Kingdom
| | - Rosemary Bland
- The Clinical Sciences Research Laboratory, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Daniel Zehnder
- The Clinical Sciences Research Laboratory, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Department of Nephrology, University Hospital Coventry and Warwickshire, Coventry, United Kingdom
| |
Collapse
|
7
|
Xu ZC, Zhang Q, Li H. Elastic large muscular vessel wall engineered with bone marrow‑derived cells under pulsatile stimulation in a bioreactor. Mol Med Rep 2015; 12:6005-12. [PMID: 26238613 DOI: 10.3892/mmr.2015.4147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 07/03/2015] [Indexed: 11/05/2022] Open
Abstract
Bone marrow‑derived cells (BMCs) have demonstrated their ability to differentiate into multiple cell lineages and may be a promising cell source for vascular tissue engineering. Although much progress has been made in the engineering of small blood vessels (<6 mm in diameter) with biodegradable materials such as polyglycolic acid (PGA), it remains a challenge to engineer large vessels (>6 mm in diameter) due to unsatisfactory biomechanical properties. The present study was to engineered an elastic large vessel wall (6 mm in diameter) using a PGA unwoven fibre scaffold covered with BMCs from canine humeri. The cell‑PGA sheet was then loaded into a bioreactor designed for the present study, with dynamic pulsatile culture conditions to mimic the physiological vessel environment. After four weeks of the pulsatile stimuli culture, an elastic vessel wall was formed. Histological analyses demonstrated that layers of smooth muscle‑like cells and well‑oriented collagenous fibres were evenly oriented in the dynamic group. By contrast, disorganised cells and randomly collagenous fibres were apparent in the static group. Furthermore, the engineered vessel wall in the dynamic group exhibited significantly improved biomechanical properties compared with those in static culture group. The approach developed in the present study was demonstrated to have promising potential to be used for the engineering of large vessel as well as other smooth muscle cell‑containing tissues, including bladder, urethral and intestinal tissues.
Collapse
Affiliation(s)
- Zhi Cheng Xu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, P.R. China
| | - Qun Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, P.R. China
| | - Hong Li
- Department of Life Information and Instrument Engineering, Hangzhou Electronic Science and Technology University, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
8
|
Ahn H, Ju YM, Takahashi H, Williams DF, Yoo JJ, Lee SJ, Okano T, Atala A. Engineered small diameter vascular grafts by combining cell sheet engineering and electrospinning technology. Acta Biomater 2015; 16:14-22. [PMID: 25641646 DOI: 10.1016/j.actbio.2015.01.030] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/16/2014] [Accepted: 01/08/2015] [Indexed: 12/11/2022]
Abstract
Tissue engineering offers an attractive approach to creating functional small-diameter (<5mm) blood vessels by combining autologous cells with a natural and/or synthetic scaffold under suitable culture conditions, which results in a tubular construct that can be implanted in vivo. We have previously developed a vascular scaffold fabricated by electrospinning poly(ε-caprolactone) (PCL) and type I collagen that mimics the structural and biomechanical properties of native vessels. In this study, we investigated whether a smooth muscle cell (SMC) sheet could be combined with the electrospun vascular scaffolds to produce a more mature smooth muscle layer as compared to the conventional cell seeding method. The pre-fabricated SMC sheet, wrapped around the vascular scaffold, provided high cell seeding efficiency (approx. 100%) and a mature smooth muscle layer that expressed strong cell-to-cell junction, connexin 43 (CX43), and contractile proteins, α smooth muscle actin (α-SMA) and myosin light chain kinase (MLCK). Moreover, bioreactor-associated preconditioning of the SMC sheet-combined vascular scaffold maintained high cell viability (95.9 ± 2.7%) and phenotypes and improved cellular infiltration and mechanical properties (35.7% of tensile strength, 47.5% of elasticity, and 113.2% of elongation at break).
Collapse
|
9
|
Potter CMF, Lao KH, Zeng L, Xu Q. Role of biomechanical forces in stem cell vascular lineage differentiation. Arterioscler Thromb Vasc Biol 2014; 34:2184-90. [PMID: 25012135 DOI: 10.1161/atvbaha.114.303423] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mechanical forces have long been known to play a role in the maintenance of vascular homeostasis in the mature animal and in developmental regulation in the fetus. More recently, it has been shown that stem cells play a role in vascular repair and remodeling in response to biomechanical stress. Laminar shear stress can directly activate growth factor receptors on stem/progenitor cells, initiating signaling pathways leading toward endothelial cell differentiation. Cyclic strain can stimulate stem cell differentiation toward smooth muscle lineages through different mechanisms. In vivo, blood flow in the coronary artery is significantly altered after stenting, leading to changes in biomechanical forces on the vessel wall. This disruption may activate stem cell differentiation into a variety of cells and cause delayed re-endothelialization. Based on progress in the research field, the present review aims to explore the role of mechanical forces in stem cell differentiation both in vivo and in vitro and to examine what this means for the application of stem cells in the clinic, in tissue engineering, and for the management of aberrant stem cell contribution to disease.
Collapse
Affiliation(s)
- Claire M F Potter
- From the Cardiovascular Division, King's College London, London, United Kingdom
| | - Ka Hou Lao
- From the Cardiovascular Division, King's College London, London, United Kingdom
| | - Lingfang Zeng
- From the Cardiovascular Division, King's College London, London, United Kingdom
| | - Qingbo Xu
- From the Cardiovascular Division, King's College London, London, United Kingdom.
| |
Collapse
|
10
|
Venkataraman L, Bashur CA, Ramamurthi A. Impact of cyclic stretch on induced elastogenesis within collagenous conduits. Tissue Eng Part A 2014; 20:1403-15. [PMID: 24313750 DOI: 10.1089/ten.tea.2013.0294] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In vitro tissue engineering of vascular conduits requires a synergy between several external factors, including biochemical supplementation and mechanotranductive stimulation. The goal of this study was to improve adult human vascular smooth muscle cell orientation and elastic matrix synthesis within 3D tubular collagen gel constructs. We used a combination of elastogenic factors (EFs) previously tested in our lab, along with cyclic circumferential strains at low amplitude (2.5%) delivered at a range of frequencies (0.5, 1.5, and 3 Hz). After 21 days of culture, the constructs were analyzed for elastic matrix outcomes, activity of matrix metalloproteinases (MMPs)-2 and -9, cell densities and phenotype, and mechanical properties of constructs. While cell densities remained unaffected by the addition of stretch, contractile phenotypic markers were elevated in all stretched constructs relative to control. Constructs cultured with EFs stretched at 1.5 Hz exhibited the maximum elastin mRNA expression and total matrix elastin (over sixfold vs. the static EFs control). MMP-2 content was comparable in all treatment conditions, but MMP-9 levels were elevated at the higher frequencies (1.5 and 3 Hz). Minimal circumferential orientation was achieved and the mechanical properties remained comparable among the treatment conditions. Overall, constructs treated with EFs and stretched at 1.5 Hz exhibited the most elastogenic outcomes.
Collapse
|
11
|
Park KS, Lee EG, Son Y. Uniaxial cyclic strain stimulates cell proliferation and secretion of interleukin-6 and vascular endothelial growth factor of human dermal fibroblasts seeded on chitosan scaffolds. J Biomed Mater Res A 2013; 102:2268-76. [PMID: 23894088 DOI: 10.1002/jbm.a.34881] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/25/2013] [Indexed: 12/22/2022]
Abstract
Human dermal fibroblasts were inoculated into chitosan sponge scaffolds coated with type I collagen and it might be developed as a dermal substitute and/or dressing material. The application of 14% uniaxial cyclic strain to the cellular scaffolds affected the characteristics of the seeded human dermal fibroblasts. Cyclic strain enhanced cellular proliferation, the activity of metalloproteinase-2, and the expression of extracellular matrix proteins such as fibronectin. Moreover, cyclic strain increased the expression of vascular endothelial growth factor (VEGF) and interleukin (IL)-6, which are critical to wound healing. Even under static culture (strain, 0%) following 14% cyclic strain, the expression of VEGF and IL-6, which had increased under 14% strain, was amplified or maintained for at least 3 days. Uniaxial cyclic strain may enhance the wound-healing potential of human dermal fibroblasts seeded on chitosan scaffolds through the changes in the cellular characteristics of the fibroblasts when the cellular scaffold is transplanted into skin wounds, especially chronic wounds such as diabetic wounds.
Collapse
Affiliation(s)
- Ki-Sook Park
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Kiheung-ku, Yong In, 441-701, Korea
| | | | | |
Collapse
|
12
|
Liang MS, Koobatian M, Lei P, Swartz DD, Andreadis ST. Differential and synergistic effects of mechanical stimulation and growth factor presentation on vascular wall function. Biomaterials 2013; 34:7281-91. [PMID: 23810080 DOI: 10.1016/j.biomaterials.2013.05.073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 05/27/2013] [Indexed: 01/04/2023]
Abstract
We investigated the hypothesis that immobilizing TGF-β1 within fibrin hydrogels may act in synergy with cyclic mechanical stimulation to enhance the properties of vascular grafts. To this end, we engineered a fusion TGF-β1 protein that can covalently anchor to fibrin during polymerization upon the action of factor XIII. We also developed a 24-well based bioreactor in which vascular constructs can be mechanically stimulated by distending the silastic mandrel in the middle of each well. TGF-β1 was either conjugated to fibrin or supplied in the culture medium and the fibrin-based constructs were cultured statically for a week followed by cyclic distention for another week. The tissues were examined for myogenic differentiation, vascular reactivity, mechanical properties and ECM content. Our results showed that some aspects of vascular function were differentially affected by growth factor presentation vs. pulsatile force application, while others were synergistically enhanced by both. Overall, this two-prong biomimetic approach improved ECM secretion, vascular reactivity and mechanical properties of vascular constructs. These findings may be applied in other tissue engineering applications such as cartilage, tendon or cardiac regeneration where growth factors TGF-β1 and mechano-stimulation play critical roles.
Collapse
Affiliation(s)
- Mao-Shih Liang
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | | | | | | | | |
Collapse
|
13
|
Ferdous Z, Jo H, Nerem RM. Strain Magnitude-Dependent Calcific Marker Expression in Valvular and Vascular Cells. Cells Tissues Organs 2013; 197:372-83. [DOI: 10.1159/000347007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2013] [Indexed: 11/19/2022] Open
|
14
|
Huang G, Wang L, Wang S, Han Y, Wu J, Zhang Q, Xu F, Lu TJ. Engineering three-dimensional cell mechanical microenvironment with hydrogels. Biofabrication 2012; 4:042001. [PMID: 23164720 DOI: 10.1088/1758-5082/4/4/042001] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cell mechanical microenvironment (CMM) significantly affects cell behaviors such as spreading, migration, proliferation and differentiation. However, most studies on cell response to mechanical stimulation are based on two-dimensional (2D) planar substrates, which cannot mimic native three-dimensional (3D) CMM. Accumulating evidence has shown that there is a significant difference in cell behavior in 2D and 3D microenvironments. Among the materials used for engineering 3D CMM, hydrogels have gained increasing attention due to their tunable properties (e.g. chemical and mechanical properties). In this paper, we provide an overview of recent advances in engineering hydrogel-based 3D CMM. Effects of mechanical cues (e.g. hydrogel stiffness and externally induced stress/strain in hydrogels) on cell behaviors are described. A variety of approaches to load mechanical stimuli in 3D hydrogel-based constructs are also discussed.
Collapse
Affiliation(s)
- Guoyou Huang
- Biomedical Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
McDonald SJ, Dooley PC, McDonald AC, Schuijers JA, Ward AR, Grills BL. Transient expression of myofibroblast-like cells in rat rib fracture callus. Acta Orthop 2012; 83:93-8. [PMID: 22248170 PMCID: PMC3278664 DOI: 10.3109/17453674.2011.652891] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE We have previously shown that early fracture callus of rat rib has viscoelastic and contractile properties resembling those of smooth muscle. The cells responsible for this contractility have been hypothesized to be myofibroblast-like in nature. In soft-tissue healing, force generated by contraction of myofibroblasts promotes healing. Accordingly, we tried to identify myofibroblast-like cells in early fibrous callus. ANIMALS AND METHODS Calluses from rat rib fractures were removed 7, 14, and 21 days after fracture and unfractured ribs acted as controls. All tissues were analyzed using qPCR and immunohistochemistry. We analyzed expression of smooth muscle- and myofibroblast-associated genes and proteins including alpha smooth muscle actin (αSMA), non-muscle myosin, fibronectin extra domain A variant (EDA-fibronectin), OB-cadherin, connexin-43, basic calponin (h1CaP), and h-caldesmon. RESULTS In calluses at 7 days post-fracture, there were statistically significant increases in expression of αSMA mRNA (2.5 fold), h1CaP mRNA (2.1 fold), EDA-fibronectin mRNA (14 fold), and connexin-43 mRNA (1.8 fold) compared to unfractured ribs, and by 21 days post-fracture mRNA expression in calluses had decreased to levels approaching those in unfractured rib. Immunohistochemistry of 7 day fibrous callus localized calponin, EDA-fibronectin and co-immunolabeling of OB-cadherin and αSMA (thus confirming a myofibroblastic phenotype) within various cell populations. INTERPRETATION This study provides further evidence that early rat rib callus is not only smooth muscle-like in nature but also contains a notable population of cells that have a distinct myofibroblastic phenotype. The presence of these cells indicates that in vivo contraction of early callus is a mechanism that may occur in fractures so as to facilitate healing, as it does in soft tissue wound repair.
Collapse
Affiliation(s)
- Stuart J McDonald
- Tissue and Cell Biology Group, Musculoskeletal Research Centre, La Trobe University, Victoria, Australia
| | - Philip C Dooley
- Tissue and Cell Biology Group, Musculoskeletal Research Centre, La Trobe University, Victoria, Australia
| | - Aaron C McDonald
- Tissue and Cell Biology Group, Musculoskeletal Research Centre, La Trobe University, Victoria, Australia
| | - Johannes A Schuijers
- Tissue and Cell Biology Group, Musculoskeletal Research Centre, La Trobe University, Victoria, Australia
| | - Alex R Ward
- Tissue and Cell Biology Group, Musculoskeletal Research Centre, La Trobe University, Victoria, Australia
| | - Brian L Grills
- Tissue and Cell Biology Group, Musculoskeletal Research Centre, La Trobe University, Victoria, Australia
| |
Collapse
|
16
|
Leroux-Berger M, Queguiner I, Maciel TT, Ho A, Relaix F, Kempf H. Pathologic calcification of adult vascular smooth muscle cells differs on their crest or mesodermal embryonic origin. J Bone Miner Res 2011; 26:1543-53. [PMID: 21425330 DOI: 10.1002/jbmr.382] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Vascular calcifications can occur in the elderly and in patients suffering from various diseases. Interestingly, depending on the pathology, different regions of the arterial system can be affected. Embryonic observations have clearly indicated that vascular smooth muscle cell (VSMC) origin is notably heterogeneous. For instance, in the aorta, VSMCs colonizing the aortic arch region derive from cardiac neural crest cells, whereas those populating the descending aorta derive from the mesoderm. We examined here whether the embryonic origin of aortic VSMCs would correlate with their ability to mineralize. Under hyperphosphatemic conditions that induce vascular calcifications, we performed ex vivo aortic explant cultures as well as in vitro VSMC cultures from wild-type mice. Our data showed that VSMC embryonic origin affects their ability to mineralize. Indeed, the aortic arch media made up of VSMCs of neural crest origin calcifies significantly earlier than the descending aorta composed of VSMCs, which are mesoderm-derived. Similar results were obtained with cultured VSMCs harvested from both aortic regions. We also demonstrated that in a mouse model deficient in matrix Gla protein, a potent calcification inhibitor, developing extensive and spontaneous medial calcifications of the aorta, lesions initiate in the aortic arch. Subsequently, calcifications progress outside the aortic arch region and ultimately spread all over the entire arterial tree, including the descending aorta. Altogether, our results support an unsuspected correlation between VSMCs of embryonic origin and the timing of appearance of calcifications.
Collapse
MESH Headings
- Aging/drug effects
- Aging/pathology
- Alkaline Phosphatase/genetics
- Alkaline Phosphatase/metabolism
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Calcinosis/embryology
- Calcinosis/metabolism
- Calcinosis/pathology
- Calcium-Binding Proteins/deficiency
- Calcium-Binding Proteins/metabolism
- Cells, Cultured
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/metabolism
- Kinetics
- Mesoderm/drug effects
- Mesoderm/embryology
- Mesoderm/pathology
- Mice
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neural Crest/drug effects
- Neural Crest/embryology
- Neural Crest/pathology
- Phosphates/pharmacology
- Matrix Gla Protein
Collapse
Affiliation(s)
- Margot Leroux-Berger
- Laboratoire Angiogenèse Embryonnaire et Pathologique, INSERM U833, Paris, France
| | | | | | | | | | | |
Collapse
|
17
|
McDonald SJ, Dooley PC, McDonald AC, Djouma E, Schuijers JA, Ward AR, Grills BL. α(1) adrenergic receptor agonist, phenylephrine, actively contracts early rat rib fracture callus ex vivo. J Orthop Res 2011; 29:740-5. [PMID: 21437954 DOI: 10.1002/jor.21302] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 10/08/2010] [Indexed: 02/04/2023]
Abstract
Early, soft fracture callus that links fracture ends together is smooth muscle-like in nature. We aimed to determine if early fracture callus could be induced to contract and relax ex vivo by similar pathways to smooth muscle, that is, contraction via α(1) adrenergic receptor (α(1) AR) activation with phenylephrine (PE) and relaxation via β(2) adrenergic receptor (β(2) AR) stimulation with terbutaline. A sensitive force transducer quantified 7 day rat rib fracture callus responses in modified Krebs-Henseliet (KH) solutions. Unfractured ribs along with 7, 14, and 21 day fracture calluses were analyzed for both α(1) AR and β(2) AR gene expression using qPCR, whilst 7 day fracture callus was examined via immunohistochemistry for both α(1) AR and β(2) AR- immunoreactivity. In 7 day callus, PE (10(-6) M) significantly induced an increase in force that was greater than passive force generated in calcium-free KH (n = 8, mean 51% increase, 95% CI: 26-76%). PE-induced contractions in calluses were attenuated by the α(1) AR antagonist, prazosin (10(-6) M; n = 7, mean 5% increase, 95% CI: 2-11%). Terbutaline did not relax callus. Gene expression of α(1) ARs was constant throughout fracture healing; however, β(2) AR expression was down-regulated at 7 days compared to unfractured rib (p < 0.01). Furthermore, osteoprogenitor cells of early fibrous callus displayed considerable α(1) AR-like immunoreactivity but not β(2) AR-like immunoreactivity. Here, we demonstrate for the first time that early fracture callus can be pharmacologically induced to contract. We propose that increased concentrations of α(1) AR agonists such as noradrenaline may tonically contract callus in vivo to promote osteogenesis.
Collapse
Affiliation(s)
- Stuart J McDonald
- Musculoskeletal Research Centre, School of Human Biosciences, La Trobe University, Victoria 3086, Australia
| | | | | | | | | | | | | |
Collapse
|
18
|
Ferdous Z, Jo H, Nerem RM. Differences in valvular and vascular cell responses to strain in osteogenic media. Biomaterials 2011; 32:2885-93. [PMID: 21284997 PMCID: PMC3042538 DOI: 10.1016/j.biomaterials.2011.01.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 01/12/2011] [Indexed: 10/18/2022]
Abstract
Calcification is the primary cause of failure of bioprosthetic and tissue-engineered vascular and valvular grafts. We used tissue-engineered collagen gels containing human aortic smooth muscle cells (HASMC) and human aortic valvular interstitial cells (HAVIC) as a model to investigate cell-mediated differences in early markers of calcification. The HASMCs and HAVICs were isolated from non-sclerotic human tissues. After 21 days of culture in either regular or osteogenic media with or without 10% cyclic strain at 1 Hz, the collagen gels were assessed for DNA content, collagen I, matrix metalloproteinase (MMP)-2 and glycosaminoglycan (GAG) content. The collagen gels containing HASMCs contained significantly greater amounts of collagen I and GAG compared to HAVICs. Although strain increased MMP-2 activity for both cell types, this trend was significant (p ≤ 0.05) only for HAVICs. Cultured gels were also assessed for osteogenic markers calcium content, alkaline phosphatase (ALP), and Runx2 and were present at greater amounts in gels containing HASMCs than HAVICs. Calcium content, Runx2 expression, and ALP activity were also modulated by mechanical strain. The results indicate that cell-mediated differences exist between the vascular and valvular calcification processes. Further investigation is necessary for improved understanding and to detect biomarkers for early detection or prevention of these diseases.
Collapse
Affiliation(s)
- Zannatul Ferdous
- Institute of Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Drive, Mail code 0363, Atlanta, GA 30332, USA.
| | | | | |
Collapse
|
19
|
McDonald SJ, Dooley PC, McDonald AC, Schuijers JA, Ward AR, Grills BL. Early fracture callus displays smooth muscle-like viscoelastic properties ex vivo: implications for fracture healing. J Orthop Res 2009; 27:1508-13. [PMID: 19472384 DOI: 10.1002/jor.20923] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cells of early, fibrous callus in bone fractures possess much alpha smooth muscle actin. This callus contracts and relaxes; however, active and passive components of its force production have yet to be defined. We aimed to establish whether passive viscoelastic properties of early soft fracture callus are smooth muscle-like in nature. Under anesthesia one rib was fractured in rats and calluses removed 7 days later for analysis. Urinary bladder detrusor muscle and Achilles tendon were also resected and analyzed. Force production in these tissues was measured using a force transducer when preparations were immersed in calcium-free Krebs-Henseleit solution (pH 7.4, 22 degrees C). Viscoelastic responses were measured in each preparation in response to 50 microN increases and decreases in force after achieving basal tissue tension by preconditioning. Callus, bladder, and tendon all displayed varying, reproducible degrees of stress relaxation (SR) and reverse stress relaxation (RSR) (n = 7 for all groups). Hysteresis was observed in callus, with the first SR response significantly larger than that produced in subsequent stretches (p < 0.05). Callus SR responses were greater than tendon (p < 0.001) but less than bladder (p < 0.001). Callus RSR responses were greater than tendon (p < 0.001), but no significant difference was seen between RSR of callus and bladder. We concluded that early, soft callus displayed significant SR and RSR phenomena similar to smooth muscle tissue, and SR and RSR may be important in maintenance of static tension in early callus by promoting osteogenesis and fracture healing.
Collapse
Affiliation(s)
- Stuart J McDonald
- School of Human Biosciences, Musculoskeletal Research Centre, La Trobe University, Victoria 3086, Australia
| | | | | | | | | | | |
Collapse
|
20
|
Cyclic tensile strain triggers a sequence of autocrine and paracrine signaling to regulate angiogenic sprouting in human vascular cells. Proc Natl Acad Sci U S A 2009; 106:15279-84. [PMID: 19706407 DOI: 10.1073/pnas.0905891106] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Mechanical signals regulate blood vessel development in vivo, and have been demonstrated to regulate signal transduction of endothelial cell (EC) and smooth muscle cell (SMC) phenotype in vitro. However, it is unclear how the complex process of angiogenesis, which involves multiple cell types and growth factors that act in a spatiotemporally regulated manner, is triggered by a mechanical input. Here, we describe a mechanism for modulating vascular cells during sequential stages of an in vitro model of early angiogenesis by applying cyclic tensile strain. Cyclic strain of human umbilical vein (HUV)ECs up-regulated the secretion of angiopoietin (Ang)-2 and PDGF-betabeta, and enhanced endothelial migration and sprout formation, whereas effects were eliminated with shRNA knockdown of endogenous Ang-2. Applying strain to colonies of HUVEC, cocultured on the same micropatterned substrate with nonstrained human aortic (HA)SMCs, led to a directed migration of the HASMC toward migrating HUVECs, with diminished recruitment when PDGF receptors were neutralized. These results demonstrate that a singular mechanical cue (cyclic tensile strain) can trigger a cascade of autocrine and paracrine signaling events between ECs and SMCs critical to the angiogenic process.
Collapse
|
21
|
Cyclic strain inhibits acute pro-inflammatory gene expression in aortic valve interstitial cells. Biomech Model Mechanobiol 2009; 9:117-25. [PMID: 19636599 DOI: 10.1007/s10237-009-0165-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 07/09/2009] [Indexed: 10/20/2022]
Abstract
Mechanical in vitro preconditioning of tissue engineered heart valves is viewed as an essential process for tissue development prior to in vivo implantation. However, a number of pro-inflammatory genes are mechanosensitive and their elaboration could elicit an adverse response in the host. We hypothesized that the application of normal physiological levels of strain to isolated valve interstitial cells would inhibit the expression of pro-inflammatory genes. Cells were subjected to 0, 5, 10, 15 and 20% strain. Expression of VCAM-1, MCP-1, GM-CSF and OPN was then measured using qRT-PCR. With the exception of OPN, all genes were significantly up regulated when no strain was applied. MCP-1 expression was significantly lower in the presence of strain, although strain magnitude did not affect the expression level. VCAM-1 and GM-CSF had the lowest expression levels at 15% strain, which represent normal physiological conditions. These findings were confirmed using confocal microscopy. Additionally, pSMAD 2/3 and IkappaBalpha expression were imaged to elucidate potential mechanisms of gene expression. Data showed that 15% strain increased pSMAD 2/3 expression and prevented phosphorylation of IkappaBalpha. In conclusion, cyclic strain reduces expression of pro-inflammatory genes, which may be beneficial for the in vitro pre-conditioning of tissue engineered heart valves.
Collapse
|
22
|
Yazdani SK, Watts B, Machingal M, Jarajapu YP, Van Dyke ME, Christ GJ. Smooth Muscle Cell Seeding of Decellularized Scaffolds: The Importance of Bioreactor Preconditioning to Development of a More Native Architecture for Tissue-Engineered Blood Vessels. Tissue Eng Part A 2009; 15:827-40. [DOI: 10.1089/ten.tea.2008.0092] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Saami K. Yazdani
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Benjamin Watts
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Masood Machingal
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Yagna P.R. Jarajapu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Mark E. Van Dyke
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - George J. Christ
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
23
|
Shyu KG. Cellular and molecular effects of mechanical stretch on vascular cells and cardiac myocytes. Clin Sci (Lond) 2009; 116:377-389. [DOI: 10.1042/cs20080163] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Cells in the cardiovascular system are permanently subjected to mechanical forces due to the pulsatile nature of blood flow and shear stress, created by the beating heart. These haemodynamic forces play an important role in the regulation of vascular development, remodelling, wound healing and atherosclerotic lesion formation. Mechanical stretch can modulate several different cellular functions in VSMCs (vascular smooth muscle cells). These functions include, but are not limited to, cell alignment and differentiation, migration, survival or apoptosis, vascular remodelling, and autocrine and paracrine functions. Laminar shear stress exerts anti-apoptotic, anti-atherosclerotic and antithrombotic effects on ECs (endothelial cells). Mechanical stretch of cardiac myocytes can modulate growth, apoptosis, electric remodelling, alterations in gene expression, and autocrine and paracrine effects. The aim of the present review is primarily to summarize the cellular and molecular effects of mechanical stretch on vascular cells and cardiac myocytes, emphasizing the molecular mechanisms underlying the regulation. Knowledge of the impact of mechanical stretch on the cardiovascular system is vital to the understanding of the pathogenesis of cardiovascular diseases, and is also crucial to provide new insights into the prevention and therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Kou-Gi Shyu
- Division of Cardiology, Shin Kong Wu Ho-Su Memorial Hospital, 95 Wen-Chang Rd, Taipei 111, Taiwan, and Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
24
|
Different magnitudes of tensile strain induce human osteoblasts differentiation associated with the activation of ERK1/2 phosphorylation. Int J Mol Sci 2008; 9:2322-2332. [PMID: 19330078 PMCID: PMC2635645 DOI: 10.3390/ijms9122322] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 11/19/2008] [Accepted: 11/25/2008] [Indexed: 11/30/2022] Open
Abstract
Mechanical factors are related to periprosthetic osseointegration following total hip arthroplasty. However, osteoblast response to strain in implanted femurs is unclear because of the absence of accurate stress-measuring methods. In our study, finite element analysis was performed to calculate strain distribution in implanted femurs. 0.8-3.2% tensile strain was then applied to human osteoblasts. Higher magnitudes of strain enhanced the expression of osteocalcin, type I collagen, and Cbfa1/Runx2. Lower magnitudes significantly increased ALP activity. Among these, type I collagen expression increased with the activation of ERK1/2 phosphorylation in a strain-magnitude-dependent manner. Our study marks the first investigation of osteoblast response at different magnitudes of periprosthetic strain. The results indicate that the functional status of human osteoblasts is determined by strain magnitude. The strain distribution in the proximal region of implanted femur should be improved for osseointegration.
Collapse
|
25
|
Peyton SR, Kim PD, Ghajar CM, Seliktar D, Putnam AJ. The effects of matrix stiffness and RhoA on the phenotypic plasticity of smooth muscle cells in a 3-D biosynthetic hydrogel system. Biomaterials 2008; 29:2597-607. [PMID: 18342366 PMCID: PMC2855209 DOI: 10.1016/j.biomaterials.2008.02.005] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 02/09/2008] [Indexed: 11/30/2022]
Abstract
Studies using 2-D cultures have shown that the mechanical properties of the extracellular matrix (ECM) influence cell migration, spreading, proliferation, and differentiation; however, cellular mechanosensing in 3-D remains under-explored. To investigate this topic, a unique biomaterial system based on poly(ethylene glycol)-conjugated fibrinogen was adapted to study phenotypic plasticity in smooth muscle cells (SMCs) as a function of ECM mechanics in 3-D. Tuning the compressive modulus between 448 and 5804 Pa modestly regulated SMC cytoskeletal assembly in 3-D, with spread cells in stiff matrices having a slightly higher degree of F-actin bundling after prolonged culture. However, vinculin expression in all 3-D conditions was qualitatively low and was not assembled into the classic focal adhesions typically seen in 2-D cultures. Given the evidence that RhoA-mediated cytoskeletal contractility represents a critical node in mechanosensing, we molecularly upregulated contractility by inducing SMCs to express constitutively active RhoA. In these cells, F-actin bundling and total vinculin expression increased, and focal adhesion-like structures began to emerge, consistent with RhoA's mechanism of action in cells cultured on 2-D substrates. Furthermore, SMC proliferation in 3-D did not depend significantly on matrix stiffness, and was reduced by constitutive activation of RhoA irrespective of ECM mechanical properties. Conversely, the expression of contractile markers globally increased with constitutive RhoA activation and depended on 3-D matrix stiffness only in cells with heightened RhoA activity. Combined, these data suggest that the synergistic effects of ECM mechanics and RhoA activity on SMC phenotype in 3-D are distinct from those in 2-D, and highlight the importance of studying the mechanical role of cell-matrix interactions in tunable 3-D environments.
Collapse
MESH Headings
- Actins/metabolism
- Aorta/cytology
- Cell Culture Techniques/methods
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Cross-Linking Reagents/chemistry
- Enzyme Activation/drug effects
- Extracellular Matrix/chemistry
- Fibrinogen/chemistry
- Fibrinogen/pharmacology
- Focal Adhesions/drug effects
- Humans
- Hydrogel, Polyethylene Glycol Dimethacrylate/metabolism
- Microscopy, Electron, Scanning
- Muscle Contraction/drug effects
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Polyethylene Glycols/chemistry
- Polyethylene Glycols/pharmacology
- Stress, Mechanical
- Time Factors
- Vinculin/metabolism
- rhoA GTP-Binding Protein/physiology
Collapse
Affiliation(s)
- Shelly R Peyton
- Department of Chemical Engineering and Materials Science, The Henry Samueli School of Engineering, University of California, Irvine, CA 92697-2715, United States
| | | | | | | | | |
Collapse
|
26
|
Xu ZC, Zhang WJ, Li H, Cui L, Cen L, Zhou GD, Liu W, Cao Y. Engineering of an elastic large muscular vessel wall with pulsatile stimulation in bioreactor. Biomaterials 2008; 29:1464-72. [DOI: 10.1016/j.biomaterials.2007.11.037] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Accepted: 11/28/2007] [Indexed: 10/22/2022]
|
27
|
Weenig RH. Pathogenesis of calciphylaxis: Hans Selye to nuclear factor kappa-B. J Am Acad Dermatol 2008; 58:458-71. [PMID: 18206262 DOI: 10.1016/j.jaad.2007.12.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 11/30/2007] [Accepted: 12/04/2007] [Indexed: 01/06/2023]
Abstract
The clinical syndrome of calciphylaxis is characterized by arteriolar medial calcification, thrombotic cutaneous ischemia, necrotic skin ulceration, and a high mortality rate. This review integrates calciphylaxis risk factors with the molecular processes governing osseous and extraosseous mineralization. As the pathogenesis of calciphylaxis is better understood, targeted therapies aimed at disease prevention and reversal will follow.
Collapse
Affiliation(s)
- Roger H Weenig
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota 55905, USA.
| |
Collapse
|
28
|
Abstract
Vascular grafts are in large demand for coronary and peripheral bypass surgeries. Although synthetic grafts have been developed, replacement of vessels with purely synthetic polymeric conduits often leads to the failure of such graft, especially in the grafts less than 6 mm in diameter or in the areas of low blood flow, mainly due to the early formation of thrombosis. Moreover, the commonly used materials lack growth potential, and long-term results have revealed several material-related failures, such as stenosis, thromboembolization, calcium deposition and infection. Tissue engineering has become a promising approach for generating a bio-compatible vessel graft with growth potential. Since the first success of constructing blood vessels with collagen and cultured vascular cells by Weinberg and Bell, there has been considerable progress in the area of vessel engineering. To date, tissue- engineered blood vessels (TEBVs) could be successfully constructed in vitro, and be used to repair the vascular defects in animal models. This review describes the major progress in the field, including the seeding cell sources, the biodegradable scaffolds, the construction technologies, as well as the encouraging achievements in clinical applications. The remaining challenges are also discussed.
Collapse
Affiliation(s)
- Wen Jie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, National Tissue Engineering Center of China, Shanghai, China
| | | | | | | |
Collapse
|
29
|
Yao L, Liu J, Andreadis ST. Composite fibrin scaffolds increase mechanical strength and preserve contractility of tissue engineered blood vessels. Pharm Res 2007; 25:1212-21. [PMID: 18092140 DOI: 10.1007/s11095-007-9499-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 11/08/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVES We recently demonstrated that fibrin-based tissue engineered blood vessels (TEV) exhibited vascular reactivity, matrix remodeling and sufficient strength for implantation into the veins of an ovine animal model, where they remained patent for 15 weeks. Here we present an approach to improve the mechanical properties of fibrin-based TEV and examine the relationship between mechanical strength and smooth muscle cell (SMC) function. MATERIALS AND METHODS To this end, we prepared TEV that were composed of two layers: a cellular layer containing SMC embedded in fibrin hydrogel to provide contractility and matrix remodeling; and a second cell-free fibrin layer composed of high concentration fibrinogen to provide mechanical strength. RESULTS The ultimate tensile force of double-layered TEV increased with FBG concentration in the cell-free layer in a dose-dependent manner. Double-layered TEV exhibited burst pressure that was ten-fold higher than single-layered tissues but vascular reactivity remained high even though the cells were constricting an additional tissue layer. CONCLUSION These results showed that mechanical strength results largely from the biomaterial but contractility requires active cellular machinery. Consequently, they may suggest novel approaches for engineering biomaterials that satisfy the requirement for high mechanical strength while preserving SMC function.
Collapse
Affiliation(s)
- Lan Yao
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, State University of New York at Buffalo, Amherst, NY 14260, USA
| | | | | |
Collapse
|
30
|
Peyton SR, Ghajar CM, Khatiwala CB, Putnam AJ. The emergence of ECM mechanics and cytoskeletal tension as important regulators of cell function. Cell Biochem Biophys 2007; 47:300-20. [PMID: 17652777 DOI: 10.1007/s12013-007-0004-y] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/17/2022]
Abstract
The ability to harvest and maintain viable cells from mammalian tissues represented a critical advance in biomedical research, enabling individual cells to be cultured and studied in molecular detail. However, in these traditional cultures, cells are grown on rigid glass or polystyrene substrates, the mechanical properties of which often do not match those of the in vivo tissue from which the cells were originally derived. This mechanical mismatch likely contributes to abrupt changes in cellular phenotype. In fact, it has been proposed that mechanical changes in the cellular microenvironment may alone be responsible for driving specific cellular behaviors. Recent multidisciplinary efforts from basic scientists and engineers have begun to address this hypothesis more explicitly by probing the effects of ECM mechanics on cell and tissue function. Understanding the consequences of such mechanical changes is physiologically relevant in the context of a number of tissues in which altered mechanics may either correlate with or play an important role in the onset of pathology. Examples include changes in the compliance of blood vessels associated with atherosclerosis and intimal hyperplasia, as well as changes in the mechanical properties of developing tumors. Compelling evidence from 2-D in vitro model systems has shown that substrate mechanical properties induce changes in cell shape, migration, proliferation, and differentiation, but it remains to be seen whether or not these same effects translate to 3-D systems or in vivo. Furthermore, the molecular "mechanotransduction" mechanisms by which cells respond to changes in ECM mechanics remain unclear. Here, we provide some historical context for this emerging area of research, and discuss recent evidence that regulation of cytoskeletal tension by changes in ECM mechanics (either directly or indirectly) may provide a critical switch that controls cell function.
Collapse
Affiliation(s)
- Shelly R Peyton
- Department of Chemical Engineering and Materials Science, The Henry Samueli School of Engineering, University of California, Irvine, CA 92697-2715, USA
| | | | | | | |
Collapse
|
31
|
Matsumoto T, Yung YC, Fischbach C, Kong HJ, Nakaoka R, Mooney DJ. Mechanical strain regulates endothelial cell patterning in vitro. ACTA ACUST UNITED AC 2007; 13:207-17. [PMID: 17518594 DOI: 10.1089/ten.2006.0058] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Blood vessels of the vertebrate circulatory system typically exhibit tissue-specific patterning. However, the cues that guide the development of these patterns remain unclear. We investigated the effect of cyclic uniaxial strain on vascular endothelial cell dynamics and sprout formation in vitro in two-dimensional (2D) and three-dimensional (3D) culture systems under the influence of growth factors. Cells preferentially aligned and moved in the direction perpendicular to the major strain axis in monolayer culture, and mechanical strain also regulated the spatial location of cell proliferation in 2D cell culture. Cells in 3D cell culture could be induced to form sprouts by exposure to appropriate growth factor combinations (vascular endothelial growth factor and hepatocyte growth factor), and the strain direction regulated the directionality of this process. Moreover, cyclic uniaxial strain inhibited branching of the structures formed by endothelial cells and increased their thickness. Taken together, these data support the importance of external mechanical stimulation in the regulation of endothelial cell migration, proliferation, and differentiation into primitive vessels.
Collapse
Affiliation(s)
- Takuya Matsumoto
- Division of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Yin X, Mayr M, Xiao Q, Wang W, Xu Q. Proteomic analysis reveals higher demand for antioxidant protection in embryonic stem cell-derived smooth muscle cells. Proteomics 2007; 6:6437-46. [PMID: 17163435 DOI: 10.1002/pmic.200600351] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Embryonic stem (ES) cells can differentiate into vascular smooth muscle cells (SMCs), but differences in protein composition, function and behaviour between stem cell-derived and mature SMCs remain to be characterized. Using differential in gel electrophoresis (DIGE) and MS, we identified 146 proteins that differed between ES cell-derived SMCs (esSMCs) and aortic SMCs, including proteins involved in DNA maintenance (higher in esSMCs), cytoskeletal proteins and calcium-binding proteins (higher in aortic SMCs). Notably, esSMCs showed decreased expression of mitochondrial, but a compensatory increase of cytosolic antioxidants. Subsequent experiments revealed that mitochondrial-derived reactive oxygen species (ROS) were markedly increased in esSMCs. Despite a three-fold rise in glutathione (GSH) reductase activity, esSMCs had lower levels of reduced GSH, and depletion of GSH by diethyl maleate or inhibition of GSH reductase by carmustine (BCNU) resulted in more pronounced cell death compared to aortic SMCs, while addition of antioxidants improved the viability of esSMCs. We present the first proteomic analysis of esSMCs demonstrating that stem cell-derived SMCs are more sensitive to oxidative stress due to increased generation of mitochondrial-derived ROS and require additional antioxidant protection for survival.
Collapse
Affiliation(s)
- Xiaoke Yin
- Cardiovascular Division, School of Medicine, King's College, University of London, UK
| | | | | | | | | |
Collapse
|
33
|
Jeong SI, Kim SY, Cho SK, Chong MS, Kim KS, Kim H, Lee SB, Lee YM. Tissue-engineered vascular grafts composed of marine collagen and PLGA fibers using pulsatile perfusion bioreactors. Biomaterials 2007; 28:1115-22. [PMID: 17112581 DOI: 10.1016/j.biomaterials.2006.10.025] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Accepted: 10/21/2006] [Indexed: 10/23/2022]
Abstract
Novel tubular scaffolds of marine source collagen and PLGA fibers were fabricated by freeze drying and electrospinning processes for vascular grafts. The hybrid scaffolds, composed of a porous collagen matrix and a fibrous PLGA layer, had an average pore size of 150+/-50 microm. The electrospun fibrous PLGA layer on the surface of a porous tubular collagen scaffold improved the mechanical strength of the collagen scaffolds in both the dry and wet states. Smooth muscle cells (SMCs)- and endothelial cells (ECs)-cultured collagen/PLGA scaffolds exhibited mechanical properties similar to collagen/PLGA scaffolds unseeded with cells, even after culturing for 23 days. The effect of a mechanical stimulation on the proliferation and phenotype of SMCs and ECs, cultured on collagen/PLGA scaffolds, was evaluated. The pulsatile perfusion system enhanced the SMCs and ECs proliferation. In addition, a significant cell alignment in a direction radial to the distending direction was observed in tissues exposed to radial distention, which is similar to the phenomenon of native vessel tissues in vivo. On the other hand, cells in tissues engineered in the static condition were randomly aligned. Immunochemical analyses showed that the expressions of SM alpha-actin, SM myosin heavy chain, EC von Willebrand factor, and EC nitric oxide were upregulated in tissues engineered under a mechano-active condition, compared to vessel tissues engineered in the static condition. These results indicated that the co-culturing of SMCs and ECs, using collagen/PLGA hybrid scaffolds under a pulsatile perfusion system, leads to the enhancement of vascular EC development, as well as the retention of the differentiated cell phenotype.
Collapse
Affiliation(s)
- Sung In Jeong
- School of Chemical Engineering, College of Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Park SA, Kim IA, Lee YJ, Shin JW, Kim CR, Kim JK, Yang YI, Shin JW. Biological responses of ligament fibroblasts and gene expression profiling on micropatterned silicone substrates subjected to mechanical stimuli. J Biosci Bioeng 2007; 102:402-12. [PMID: 17189167 DOI: 10.1263/jbb.102.402] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Accepted: 07/31/2006] [Indexed: 12/20/2022]
Abstract
In this study, ligament fibroblasts were cultivated on micropatterned silicone substrates and subjected to cyclic stretching to simulate the in vivo biomechanical environment during ligament healing. Without stretching, ligament fibroblasts were aligned parallel to the microgrooves on the silicone substrate surface. However, we previously reported that uniaxial cyclic stretching induces alignment perpendicular to the stretching axis. With stretching on a microgrooved surface, cell proliferation and collagen production were greatly enhanced. The exact functions of the micropatterned surface and mechanical stimuli are unknown. Therefore, in gene expression microarray experiments, genes whose expression is inhibited by subculture from passage 0 (P0) to passage 8 (P8) and enhanced by micropatterning and stretching were sought out. The following six genes were selected: MGP, GADD45A, UNC5B, TGFB1, COL4A1, and COL4A2. The selected genes play fundamental roles in cell proliferation, differentiation, apoptosis, and structural maintenance. On the basis of the obtained gene expression profiles, we identified candidate genes that might be involved in responses to a micropatterned surface and mechanical stretching.
Collapse
Affiliation(s)
- Su A Park
- Department of Future Technology, Korea Institute of Machinery & Materials, Jang-dong, Yuseong-gu, Daejeon 305-343, Korea
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Kie Shim J, Ryong Oh S, Bong Lee S, Cho KM. Preparation of hydrogels composed of poly(vinyl alcohol) and polyethyleneimine and their electrical response. J Appl Polym Sci 2007. [DOI: 10.1002/app.26676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
36
|
Matsumoto T, Yung YC, Fischbach C, Kong HJ, Nakaoka R, Mooney DJ. Mechanical Strain Regulates Endothelial Cell Patterningin Vitro. ACTA ACUST UNITED AC 2006. [DOI: 10.1089/ten.2006.12.ft-294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
37
|
Peyton SR, Raub CB, Keschrumrus VP, Putnam AJ. The use of poly(ethylene glycol) hydrogels to investigate the impact of ECM chemistry and mechanics on smooth muscle cells. Biomaterials 2006; 27:4881-93. [PMID: 16762407 DOI: 10.1016/j.biomaterials.2006.05.012] [Citation(s) in RCA: 274] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2006] [Accepted: 05/10/2006] [Indexed: 01/16/2023]
Abstract
Hydrogels based on poly(ethylene glycol) (PEG) are of increasing interest for regenerative medicine applications and are ideal materials to direct cell function due to the ability to confer key functionalities of native extracellular matrix (ECM) on PEG's otherwise inert backbone. Given extensive recent evidence that ECM compliance influences a variety of cell functions, PEG-based hydrogels are also attractive due to the ease with which their mechanical properties can be controlled. In these studies, we exploited the chemical and mechanical tunability of PEG-based gels to study the impact of ECM chemistry and mechanics on smooth muscle cells (SMCs) in both 2-D and 3-D model systems. First, by controlling the extent of crosslinking and therefore the mechanical properties of PEG-based hydrogels (tensile moduli from 13.7 to 423.9kPa), we report here that the assembly of F-actin stress fibers and focal adhesions, indicative of the state of actin contractility, were influenced by the compliance of 2-D PEG gels functionalized with either short adhesive peptides or full-length ECM proteins. Varying ECM ligand density and identity independent of gel compliance affected the physical properties of the focal adhesions, and also influenced SMC spreading in 2-D. Furthermore, SMCs proliferated to a greater extent as gel stiffness was increased. In contrast, the degree of SMC differentiation, which was qualitatively assessed by the extent of smooth muscle alpha-actin bundling and the association of calponin and caldesmon with the alpha-actin fibrils, was found to decrease with substrate stiffness in 2-D cultures. In 3-D, despite the fact that their viability and degree of spreading were greatly reduced, SMCs did express some contractile markers indicative of their differentiated phenotype when cultured within PEG-RGDS constructs. Combined, these data suggest that the mechanical and chemical properties of PEG hydrogels can be tuned to influence SMC phenotype in both 2-D and 3-D.
Collapse
Affiliation(s)
- Shelly R Peyton
- Department of Chemical Engineering and Materials Science, The Henry Samueli School of Engineering, University of California, Irvine, CA 92697-2575, USA
| | | | | | | |
Collapse
|
38
|
London GM, Marchais SJ, Guérin AP, Métivier F. Arteriosclerosis, vascular calcifications and cardiovascular disease in uremia. Curr Opin Nephrol Hypertens 2005; 14:525-31. [PMID: 16205470 DOI: 10.1097/01.mnh.0000168336.67499.c0] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Arterial calcification in chronic kidney disease (CKD) is associated with increased cardiovascular risk. The mechanisms responsible for arterial calcification include alterations of mineral metabolism and expression of mineral-regulating proteins. RECENT FINDINGS Arterial calcification is similar to bone formation, involving differentiation of vascular smooth muscle cells (VSMCs) into phenotypically distinct osteoblast-like cells. Elevated phosphate and/or calcium trigger a concentration-dependent increase of calcium precipitates in VSMC in vitro. The calcification is initiated by VSMC release of membrane-bound matrix vesicles and formation of apoptotic bodies. The presence of serum prevents these changes, indicating the presence of calcification inhibitors. Arterial calcification occurs in two sites: the tunica intima and tunica media. Intimal calcification is a marker of atherosclerotic disease and is associated with arterial stenotic lesions. Medial calcification influences outcome by promoting arterial stiffening whose principal consequences are left-ventricular hypertrophy and altered coronary perfusion. Aortic stiffness is an independent predictor of all-cause and cardiovascular mortality in CKD patients. Age, duration of dialysis, smoking and diabetes are risk factors for the development of arterial calcification in end-stage renal disease. Oversuppression of parathyroid hormone and low bone turnover potentiate the development of arterial calcification. SUMMARY Arterial disease in CKD patients is characterized by extensive calcification. Evidence has accumulated pointing to the active and regulated nature of the calcification process. Elevated phosphate and calcium may stimulate sodium-dependent phosphate cotransport involving osteoblast-like changes in cellular gene expression. Arterial calcification is responsible for stiffening of the arteries with increased left-ventricular afterload and abnormal coronary perfusion as the principal clinical consequences.
Collapse
Affiliation(s)
- Gérard M London
- Service d'Hémodialyse, Hôpital F.H. Manhès, 8, rue Roger Clavier, 91712 Fleury-Mérogis, Cedex, France.
| | | | | | | |
Collapse
|
39
|
Stegemann JP, Hong H, Nerem RM. Mechanical, biochemical, and extracellular matrix effects on vascular smooth muscle cell phenotype. J Appl Physiol (1985) 2005; 98:2321-7. [PMID: 15894540 DOI: 10.1152/japplphysiol.01114.2004] [Citation(s) in RCA: 208] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The vascular smooth muscle cell (VSMC) is surrounded by a complex extracellular matrix that provides and modulates a variety of biochemical and mechanical cues that guide cell function. Conventional two-dimensional monolayer culture systems recreate only a portion of the cellular environment, and therefore there is increasing interest in developing more physiologically relevant three-dimensional culture systems. This review brings together recent studies on how mechanical, biochemical, and extracellular matrix stimulation can be applied to study VSMC function and how the combination of these factors leads to changes in phenotype. Particular emphasis is placed on in vitro experimental studies in which multiple stimuli are combined, especially in three-dimensional culture systems and in vascular tissue engineering applications. These studies have provided new insight into how VSMC phenotype is controlled, and they have underscored the interdependence of biochemical and mechanical signaling. Future improvements in creating more complex in vitro culture environments will lead to a better understanding of VSMC biology, new treatments for vascular disease, as well as improved blood vessel substitutes.
Collapse
Affiliation(s)
- Jan P Stegemann
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | |
Collapse
|
40
|
Riha GM, Lin PH, Lumsden AB, Yao Q, Chen C. Roles of Hemodynamic Forces in Vascular Cell Differentiation. Ann Biomed Eng 2005; 33:772-9. [PMID: 16078617 DOI: 10.1007/s10439-005-3310-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The pulsatile nature of blood flow is a key stimulus for the modulation of vascular cell differentiation. Within the vascular media, physiologic stress is manifested as cyclic strain, while in the lumen, cells are subjected to shear stress. These two respective biomechanical forces influence the phenotype and degree of differentiation or proliferation of smooth muscle cells and endothelial cells within the human vasculature. Elucidation of the effect of these mechanical forces on cellular differentiation has led to a surge of research into this area because of the implications for both the treatment of atherosclerotic disease and the future of vascular tissue engineering. The use of mechanical force to directly control vascular cell differentiation may be utilized as an invaluable engineering tool in the future. However, an understanding of the role of hemodynamics in vascular cell differentiation and proliferation is critical before application can be realized. Thus, this review will provide a current perspective on the latest research and controversy behind the role of hemodynamic forces for vascular cell differentiation and phenotype modulation. Furthermore, this review will illustrate the application of hemodynamic force for vascular tissue engineering and explicate future directions for research.
Collapse
Affiliation(s)
- Gordon M Riha
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
41
|
Jeong SI, Kwon JH, Lim JI, Cho SW, Jung Y, Sung WJ, Kim SH, Kim YH, Lee YM, Kim BS, Choi CY, Kim SJ. Mechano-active tissue engineering of vascular smooth muscle using pulsatile perfusion bioreactors and elastic PLCL scaffolds. Biomaterials 2005; 26:1405-11. [PMID: 15482828 DOI: 10.1016/j.biomaterials.2004.04.036] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2004] [Accepted: 04/23/2004] [Indexed: 10/26/2022]
Abstract
Blood vessels are subjected in vivo to mechanical forces in a form of radial distention, encompassing cyclic mechanical strain due to the pulsatile nature of blood flow. Vascular smooth muscle (VSM) tissues engineered in vitro with a conventional tissue engineering technique may not be functional, because vascular smooth muscle cells (VSMCs) cultured in vitro typically revert from a contractile phenotype to a synthetic phenotype. In this study, we hypothesized that pulsatile strain and shear stress stimulate VSM tissue development and induce VSMCs to retain the differentiated phenotype in VSM engineering in vitro. To test the hypothesis, rabbit aortic smooth muscle cells (SMCs) were seeded onto rubber-like elastic, three-dimensional PLCL [poly(lactide-co-caprolactone), 50:50] scaffolds and subjected to pulsatile strain and shear stress by culturing them in pulsatile perfusion bioreactors for up to 8 weeks. As control experiments, VSMCs were cultured on PLCL scaffolds statically. The pulsatile strain and shear stress enhanced the VSMCs proliferation and collagen production. In addition, a significant cell alignment in a direction radial to the distending direction was observed in VSM tissues exposed to radial distention, which is similar to that of native VSM tissues in vivo, whereas VSMs in VSM tissues engineered in the static condition randomly aligned. Importantly, the expression of SM alpha-actin, a differentiated phenotype of SMCs, was upregulated by 2.5-fold in VSM tissues engineered under the mechano-active condition, compared to VSM tissues engineered in the static condition. This study demonstrates that tissue engineering of VSM tissues in vitro by using pulsatile perfusion bioreactors and elastic PLCL scaffolds leads to the enhancement of tissue development and the retention of differentiated cell phenotype.
Collapse
Affiliation(s)
- Sung In Jeong
- School of Chemical Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bratz IN, Dick GM, Partridge LD, Kanagy NL. Reduced molecular expression of K(+) channel proteins in vascular smooth muscle from rats made hypertensive with N{omega}-nitro-L-arginine. Am J Physiol Heart Circ Physiol 2005; 289:H1277-83. [PMID: 15792990 DOI: 10.1152/ajpheart.01052.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Smooth muscle membrane potential (E(m)) depends on K(+) channels, and arteries from rats made hypertensive with N(omega)-nitro-l-arginine (LHR) are depolarized compared with control. We hypothesized that decreased K(+) channel function, due to decreased K(+) channel protein expression, underlies E(m) depolarization. Furthermore, K(+) channel blockers should move control E(m) (-46 +/- 1 mV) toward that in LHR (-37 +/- 2 mV) and normalize contraction. The E(m) vs. K(+) relationship was less steep in LHR (23 +/- 2 vs. 28 +/- 1 mV/log K(+) concentration), and contractile sensitivity to K(+) was increased (EC(50) = 37 +/- 1 vs. 23 +/- 1 mM). Iberiotoxin (10 nM), an inhibitor of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels, depolarized control and LHR E(m) to -35 +/- 1 and -30 +/- 2 mV, respectively; however, effects on K(+) sensitivity were more profound in LHR (EC(50) = 25 +/- 2 vs. 15 +/- 3 mM). The voltage-dependent K(+) (K(V)) channel blocker 4-aminopyridine (3 mM) depolarized control E(m) to the level of LHR (-28 +/- 1 vs. -28 +/- 1 mV); however, effects on K(+) sensitivity were greater in LHR (EC(50) = 17 +/- 4 vs. 4 +/- 4 mM). Western blots revealed reduced BK(Ca) and K(V)1.5 channel expression in LHR arteries. The findings suggest that diminished expression of K(+) channels contributes to depolarization and enhanced contractile sensitivity. These conclusions are supported by direct electrophysiological assessment of BK(Ca) and K(V) channel function in control and LHR smooth muscle cells.
Collapse
Affiliation(s)
- Ian N Bratz
- Department of Physiology, LSU Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
43
|
Kumar A, Murphy R, Robinson P, Wei L, Boriek AM. Cyclic mechanical strain inhibits skeletal myogenesis through activation of focal adhesion kinase, Rac-1 GTPase, and NF-kappaB transcription factor. FASEB J 2005; 18:1524-35. [PMID: 15466361 DOI: 10.1096/fj.04-2414com] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Myogenesis is a multistep developmental program that generates and regenerates skeletal muscles. Several extracellular factors have been identified that participate in the regulation of myogenesis. Although skeletal muscles are always subjected to mechanical stress in vivo, the role of mechanical forces in the regulation of myogenesis remains unknown. We have investigated the molecular mechanisms by which cyclic mechanical strain modulates myogenesis. Application of cyclic mechanical strain using the computer-controlled Flexcell Strain Unit increased the proliferation of C2C12 cells and inhibited their differentiation into myotubes. Cyclic strain increased the activity of cyclin-dependent kinase 2 (cdk2) and the cellular level of cyclin A, and inhibited the expression of myosin heavy chain and formation of myotubes in C2C12 cultures. The activity of nuclear factor-kappa B (NF-kappaB) transcription factor and the expression of NF-kappaB-regulated genes, cyclin D1 and IL-6, were augmented in response to mechanical strain. Cyclic strain also increased the activity of Rho GTPases, especially Rac-1. The inhibition of Rho GTPases activity, by overexpression of Rho GDP dissociation inhibitor (Rho-GDI), inhibited the strain-induced activation of NF-kappaB in C2C12 cells. Overexpression of either NF-kappaB inhibitory protein IkappaBalphaDeltaN (a degradation resistant mutant IkappaBalpha) or Rho-GDI blocked the strain-induced proliferation of C2C12 cells. Furthermore, overexpression of FRNK, a dominant negative mutant of focal adhesion kinase (FAK), inhibited the strain-induced proliferation of C2C12 cells. Our study demonstrates that cyclic mechanical strain inhibits myogenesis through the activation of FAK, Rac-1, and NF-kappaB.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Medicine, Pulmonary and Critical Care Section, Suite 520B, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
44
|
Silva EA, Mooney DJ. Synthetic extracellular matrices for tissue engineering and regeneration. Curr Top Dev Biol 2005; 64:181-205. [PMID: 15563948 DOI: 10.1016/s0070-2153(04)64008-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The need for replacement tissues or organs requires a tissue supply that cannot be satisfied by the donor supply. The tissue engineering and regeneration field is focused on the development of biological tissue and organ substitutes and may provide functional tissues to restore, maintain, or improve tissue formation. This field is already providing new therapeutic options to bypass the limitations of organ?tissue transplantation and will likely increase in medical importance in the future. This interdisciplinary field accommodates principles of life sciences and engineering and encompasses three major strategies. The first, guided tissue regeneration, relies on synthetic matrices that are conductive to host cells populating a tissue defect site and reforming the lost tissue. The second approach, inductive strategy, involves the delivery of growth factors, typically using drug delivery strategies, which are targeted to specific cell populations in the tissues surrounding the tissue defect. In the third approach, specific cell populations, typically multiplied in culture, are directly delivered to the site at which one desires to create a new tissue or organ. In all of these approaches, the knowledge acquired from developmental studies often serves as a template for the tissue engineering approach for a specific tissue or organ. This article overviews the development of synthetic extracellular matrices (ECMs) for use in tissue engineering that aim to mimic functions of the native ECM of developing and regenerating tissues. In addition to the potential therapeutic uses of these materials, they also provide model systems for basic studies that may shed light on developmental processes.
Collapse
Affiliation(s)
- Eduardo A Silva
- Division of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| | | |
Collapse
|
45
|
Williams C, Wick TM. Perfusion bioreactor for small diameter tissue-engineered arteries. ACTA ACUST UNITED AC 2005; 10:930-41. [PMID: 15265311 DOI: 10.1089/1076327041348536] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A scaleable perfusion bioreactor has been developed for tissue engineering of small diameter arterial constructs. This modular bioreactor allows for dynamic sequential seeding of smooth muscle and endothelial cells, biomechanical stimulation of cells during culture, and monitoring of tissue growth and maturation. Bovine aortic smooth muscle and endothelial cells were seeded onto porous tubular poly(glycolic acid) nonwoven scaffolds and cultured in the bioreactor under pulsatile flow conditions for up to 25 days. Cell proliferation was more than 3-fold after 4 days, smooth muscle cells expressed differentiated phenotype after 16 days, and collagen and elastin were distributed throughout the construct after 25 days of culture. In bioreactor experiments in which the construct lumen was seeded with endothelial cells by perfusion after 13 days of smooth muscle cell culture, endothelial cell seeding efficiency was 100%, and a confluent monolayer was observed in the lumen within 48 h. These data demonstrate that this perfusion bioreactor supports sequential seeding of constructs with smooth muscle and endothelial cells. Dynamic culture under pulsatile flow leads to cellular expression of differentiated function and extracellular matrix deposition toward the development of tissue-engineered arterial constructs.
Collapse
Affiliation(s)
- Chrysanthi Williams
- School of Chemical and Biomolecular Engineering, Wallace H. Coulter Department of Biomedical Engineering, and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | |
Collapse
|
46
|
Magne D, Julien M, Vinatier C, Merhi-Soussi F, Weiss P, Guicheux J. Cartilage formation in growth plate and arteries: from physiology to pathology. Bioessays 2005; 27:708-16. [PMID: 15954094 DOI: 10.1002/bies.20254] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vascular calcifications are the consequence of several pathological conditions such as atherosclerosis, diabetes, hypercholesterolemia and chronic renal insufficiency. They are associated with risks of amputation, ischemic heart disease, stroke and increased mortality. A growing body of evidence indicates that vascular smooth muscle cells (VSMCs) undergo chondrogenic commitment eventually leading to vascular calcification, by mechanisms similar to those governing ossification in the cartilage growth plate. Our knowledge of the formation of cartilage growth plate can therefore help us to understand why and how arteries calcify and, consequently, develop new therapeutic strategies. Reciprocally, thorough consideration of the events leading to ectopic chondrocyte differentiation appears crucial to further increase our understanding of growth plate formation. In this context, we will review the effects of known or suspected factors that promote chondrogenic differentiation in growth plate and arteries.
Collapse
Affiliation(s)
- D Magne
- INSERM EM 99-03, Research Center on Osteoarticular and Dental Tissue Engineering, Nantes, France
| | | | | | | | | | | |
Collapse
|
47
|
Simmons CA, Nikolovski J, Thornton AJ, Matlis S, Mooney DJ. Mechanical stimulation and mitogen-activated protein kinase signaling independently regulate osteogenic differentiation and mineralization by calcifying vascular cells. J Biomech 2004; 37:1531-41. [PMID: 15336928 DOI: 10.1016/j.jbiomech.2004.01.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2004] [Indexed: 11/16/2022]
Abstract
Ectopic calcification of vascular tissue is associated with several cardiovascular pathologies and likely involves active regulation by vascular smooth muscle cells and osteoblast-like vascular cells. This process often occurs in sites with altered mechanical environments, suggesting a role for mechanical stimuli in calcification. In this study, we investigated the effect of mechanical stimulation on the proliferation, osteogenic differentiation, calcification, and mitogen-activated protein kinase (MAPK) signaling in calcifying vascular cells (CVCs), a subpopulation of aortic smooth muscle cells putatively involved in vascular calcification. Application of equibiaxial cyclic strain (7%, 0.25 Hz) to CVCs had no effect on cell proliferation, but accelerated alkaline phosphatase expression and significantly increased mineralization by 3.1-fold over unstrained cells. Fluid motion in the absence of strain also enhanced mineralization, but to a lesser degree. Because MAPK pathways mediate mechanically regulated osteoblast differentiation, we tested whether similar signaling was involved in mineralization by CVCs. In static cultures, pharmacological inhibition of the extracellular signal-regulated kinase (ERK1/2), p38 MAPK, and c-Jun N-terminal kinase pathways significantly attenuated mineral production by as much as -94%, compared with uninhibited CVCs. Strikingly, although mechanical stimulation activated each of the MAPK pathways, inhibition of these pathways had no effect on the mechanically induced enhancement of alkaline phosphatase activity or mineralization. These novel data indicate that mechanical signals regulate calcification by CVCs, and although MAPK signaling is critical to CVC osteogenic differentiation and mineralization, it is not involved directly in transduction of mechanical signals to regulate these processes under the conditions utilized in this study.
Collapse
Affiliation(s)
- Craig A Simmons
- Department of Biomedical Engineering, University of Michigan, 1011 N University Avenue, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | |
Collapse
|
48
|
Cho SW, Kim IK, Lim SH, Kim DI, Kang SW, Kim SH, Kim YH, Lee EY, Choi CY, Kim BS. Smooth muscle-like tissues engineered with bone marrow stromal cells. Biomaterials 2004; 25:2979-86. [PMID: 14967530 DOI: 10.1016/j.biomaterials.2003.09.068] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2003] [Accepted: 09/18/2003] [Indexed: 11/28/2022]
Abstract
Bone marrow-derived cells have demonstrated the ability to differentiate into multiple mesenchymal cell lineages. Here we tested whether smooth muscle (SM)-like tissues can be created in vivo with bone marrow stromal cells (BMSCs). Cultured canine BMSCs, which expressed SM cell-specific markers including SM alpha-actin and SM myosin heavy chain, were seeded on three-dimensional, biodegradable polymer scaffolds and implanted into peritoneal cavity of athymic mice. The cell-scaffold constructs retrieved 4 weeks after implantation formed three-dimensional tissues. Immunohistochemical analyses showed that the tissue reconstructs expressed SM alpha-actin and SM myosin heavy chain. Masson's trichrome staining showed the presence of significant amounts of collagen in the tissue reconstructs. Cells labeled with a fluorescent tracer prior to implantation were still present in the tissue reconstructs 4 weeks after implantation. Non-seeded scaffolds (control groups) retrieved 4 weeks after implantation did not exhibit extensive tissue formation. This study demonstrates the potential of BMSCs as an alternative cell source for tissue engineering of SM.
Collapse
Affiliation(s)
- Seung-Woo Cho
- Department of Chemical Engineering, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|