Engineering of the human vessel wall with hair follicle stem cells in vitro

  • Authors:
    • Zhi‑Cheng Xu
    • Qun Zhang
    • Hong Li
  • View Affiliations

  • Published online on: December 9, 2016     https://doi.org/10.3892/mmr.2016.6013
  • Pages: 417-422
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hair follicle stem cells (HFSCs) are increasingly used as a stem cell paradigm in vascular tissue engineering due to the fact that they are a rich source of easily accessible multipotent adult stem cells. Promising results have been demonstrated with small diameter (less than 6 mm) tissue engineered blood vessels under low blood pressure, however engineering large vessels (>6 mm in diameter) remains a challenge due to the fact it demands a higher number of seed cells and higher quality biomechanical properties. The aim of the current study was to engineer a large vessel (6 mm in diameter) with differentiated smooth muscle cells (SMCs) induced from human (h)HFSCs using transforming growth factor‑β1 and platelet‑derived growth factor BB in combination with low‑serum culture medium. The cells were seeded onto polyglycolic acid and then wrapped around a silicone tube and further cultured in vitro. A round vessel wall was formed subsequent to 8 weeks of culture. Histological examination indicated that layers of smooth muscle‑like cells and collagenous fibres were oriented in the induced group. In contrast, disorganised cells and collagenous fibres were apparent in the undifferentiated group. The approach developed in the current study demonstrated potential for constructing large muscular vessels with differentiated SMCs induced from hHFSCs.

Introduction

Cardiovascular diseases remain a significant health and socioeconomic burden in developed countries (1). Surgical bypass with an autologous vein remains the primary treatment (2), however, a usable vascular graft is often absent due to limited sources and donor site morbidity. In addition, current widely used synthetic materials have several limitations including immunological and thrombotic complications. Furthermore, these vascular grafts are usually non-degradable and lack growth potential (3,4).

The development of tissue engineering technology is promising for potential improvement over currently used synthetic grafts. A blood vessel made of autologous cells and a biocompatible scaffold with the potential to remodel, repair and grow would be a major therapeutic advance (5). Promising results have been demonstrated with small diameter (<6 mm) tissue engineered blood vessels (TEBVs) under low blood pressure (68), however, few studies have focused on cases with larger vessels (>6 mm in diameter), which demand an increased number of seed cells and improved biomechanical properties.

However, tissue engineering approaches are limited by the large number of cells that must be obtained for regenerative medicine. Stem cells are promising cell sources with increased proliferation and broad differentiation capacity, making them suitable for the preparation of TEBVs (911). Although several types of stem and progenitor cells have been investigated for their potential as sources of seed cells in vascular tissue engineering (1216), hair follicles are increasingly used for stem cell research due to the fact that they are a rich source of easily accessible multipotent adult stem cells. Hair follicle stem cells (HFSCs) have been demonstrated to possess osteogenic, adipogenic, chondrogenic, neurogenic and myogenic lineage differentiation potential (7,1719).

In a previous study, human HFSCs (hHFSCs) were successfully induced to differentiate into functional smooth muscle cells (SMCs) by transforming growth factor-β1 (TGF-β1) and platelet-derived growth factor BB (PDGF-BB) in combination with low-serum culture medium (20). The aim of the present study was to engineer a large vessel (6 mm in diameter) using induced hHFSCs and polyglycolic acid (PGA) with 8 weeks of in vitro culture. The rapid degradation of the PGA prevents the accumulation of degraded fragments in vivo. The culture system used indicated potential to construct large muscular vessels with differentiated SMCs induced from hHFSCs.

Materials and methods

Isolation and culture of hHFSCs

hHFSCs were obtained from human scalp tissue from healthy adult patients (average age, 30 years) undergoing cosmetic plastic surgery, as described previously (20). All protocols for human tissue handling were approved by the Research Ethical Committee of Shanghai 9th People's Hospital, and written informed consent was obtained from the patients. hHFSCs at the second passage were used in the subsequent study. The hHFSCs were characterised by determining their CD marker profile (K15, K19 and integrin β1) and their ability to differentiate into osteogenic, adipogenic and chondrogenic lineages (data not shown), as reported previously (21,22).

Induction of SM differentiation

As previously reported (20), hHFSCs reaching subconfluence were cultured in low-glucose Dulbecco's modified Eagle's medium (LG-DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% foetal bovine serum (FBS; GE Healthcare Life Sciences, Logan, UT, USA) supplemented with 5 ng/ml recombinant human TGF-β1 (R&D Systems, Inc., Minneapolis, MN, USA) and 10 ng/ml recombinant human PDGF-BB (R&D Systems, Inc.) with 1% FBS. DMEM supplemented with 1% FBS was defined as the basal medium (BM). Human umbilical artery SMCs (hUASMCs) were obtained from ScienCell Research Laboratories (Carlsbad, CA, USA) and used as the positive control. The culture media were changed every 2 days. Cell characterisation and functional evaluation (data not shown) were performed subseqeuent to 8 days of culture as described previously (20).

Culture of hHFSC-PGA sheets in dishes

In the current study, 35 mg unwoven PGA fibres (Albany International Research Co., Albany, NY, USA) were constructed into an approximately 35×80×2 mm mesh. The scaffold was first soaked in 75% ethanol (Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) for 2 h. Subsequently, it was washed three times with phosphate-buffered saline and incubated in DMEM for 10 min. The medium was removed, and the scaffold was incubated in an incubator (Binder GmbH, Tuttlingen, German) at 37°C prior to use. Differentiated and undifferentiated hHFSCs (6×107) were each evenly seeded onto the PGA mesh in 100-mm culture dishes (Falcon; BD Biosciences, San Jose, CA, USA). To accomplish the complete adhesion of the hHFSCs to the fibres, the cell-scaffold constructs were then maintained in the incubator at 37°C with 95% humidity and 5% CO2 for approximately 4 h. Thereafter, sufficient induced culture medium or BM was added to the two dishes to cover the constructs. The cell-PGA sheets were incubated at 37°C for another 5 days prior to use.

Induced culture in dishes

Subsequent to culture in dishes for 5 days, the cell-PGA sheets were wrapped around the silicone tubes and fixed by biodegradable sutures (Ethicon, Inc., Somerville, NJ, USA) for another 8 weeks of culture. The culture media was changed twice a week. The cell-PGA constructs cultured in BM were used as the controls.

Histological analysis

Subsequent to 8 weeks of culture, the engineered vessel walls were harvested, fixed in 10% formalin (Sigma-Aldrich; Merck Millipore) and embedded in paraffin (Sigma-Aldrich; Merck Millipore). Following this, they were sequentially cut into sections of 4 mm thickness. The sections were then tested with haematoxylin and eosin or Masson's trichrome and Gömöri staining (all stains were from Sigma-Aldrich; Merck Millipore).

Hydroxyproline assay

For hydroxyproline assessment, the vessel wall was dried and weighed. The total hydroxyproline content of each vessel was determined by a colorimetric assay described by Reddy and Ewemeka (23). In the current study, a Sigma-MAK008, Hydroxyproline Assay Kit (Sigma-Aldrich; Merck Millipore) and a Genesys 20 Spectrophotometer (Z376027; Sigma-Aldrich; Merck Millipore) were used. Normal human saphenous vein with a diameter of 4 mm served as the control. The veins were obtained from human adult patients undergoing cardiovascular surgery with autologous vein graft. The residual saphenous veins were contributed for future experimental studies, written informed consent was obtained from the patients. All protocols for human tissue handling were approved by the Research Ethical Committee of Shanghai 9th People's Hospital.

Statistical analysis

Each experiment was repeated a minimum of three times. The results were expressed as the mean ± standard deviation. Significant differences were measured using Student's t-test. P<0.05 was considered to indicate a statistically significant differences. All of the statistical analyses were performed using SPSS software, version 16 (SPSS, Inc., Chicago, IL, USA).

Results

Culture of hHFSC-PGA constructs in dishes

Subsequent to 24 h of culture, the cells began to spread and extended along the length of the fibres. Following an additional 5 days of culture in the dishes, a cell-PGA sheet had formed (Fig. 1A). Micrographs indicated that abundant hHFSCs had adhered to the PGA fibres, with secreted extracellular matrix (ECM) filling the spaces between the fibres (Fig. 1B).

Induced culture in dishes

The hHFSC-PGA constructs were incubated in culture dishes for 8 weeks subsequent to being wrapped around silicone tubes (Fig. 2). In the induced group, the constructs demonstrated a glossy and tubular structure with a round lumen 6 mm in diameter (Fig. 3A). In contrast, the vessel walls in the static culture group exhibited a collapsed lumen and rough surface (Fig. 3B).

Histological observation

Subsequent to 8 weeks of further induced culture in vitro, several smooth muscle-like cells and few collagenous fibres were observed by histological examination (Fig. 4A and B). The PGA fibres had degraded completely, and few elastic fibres were observed at this time (Fig. 4C). In contrast, disorganised cells, randomly collagenous fibres and small elastic fibres were observed in the undifferentiated group (Fig. 4D-F). The above results were further confirmed by immunohistochemical staining for smooth muscle α-actin and calponin (data not shown), using the hUASMCs as a positive control (Fig. 4G-I).

Hydroxyproline assay

The hydroxyproline content was significantly higher (P<0.05) in the induced group than in the control group at the same time points (Fig. 5). In addition, the hydroxyproline concentration in the induced group reached approximately 65% of that in the hUASMCs.

Discussion

hHFSCs have been previously successfully isolated from patients, expanded, differentiated and used to construct autologous tissue (22,2426). This method eliminates the need for immunosuppressants and mitigates the risk of teratoma formation associated with embryonic stem cells and induced pluripotent stem cells (27). In a previous study, hHFSCs were induced to differentiate into functional SMCs by TGF-β1 and PDGF-BB in combination with low-serum culture medium (20). In the current study, a large diameter vessel wall was engineered using the aforementioned differentiated hHFSCs and PGA unwoven fibre mesh in vitro. Subsequent to culture, the newly formed tissues exhibited SM-like characteristics, including morphological performance, the expression of SM cell-specific markers (SM α-actin and calponin) and appropriate hydroxyproline content. These results demonstrated that hHFSCs may be utilised as a potential cell source for the tissue engineering of SMs, particularly that of the large diameter aorta.

Tissue engineering predominantly focuses on the incorporation of isolated cells with supporting scaffolds (28). An optimal scaffold degrades proportionally with tissue regeneration to be gradually replaced by newly formed functional tissue, and it supports cellular adhesion and collagenous matrix deposition (29). In vascular tissue engineering, the scaffold should reflect the biomechanical properties of blood vessels and serve as a platform for cell attachment and proliferation (30). It should be non-thrombogenic, non-immunogenic, biocompatible, haemocompatible, biodegradable and elastic (31,32). Furthermore, it should also control the extent and the strength of cell adhesion, proliferation, differentiation and maturation to achieve the desired phenotype and proper function (33). PGA is one of the most extensively used polymer scaffold materials in the engineering of numerous types of tissues, including blood vessels. It is a polyester that undergoes rapid degradation via hydrolysis of ester bonds, leaving behind glycolic acid and is further catabolised into water and carbon dioxide (34). The mechanical properties and degradation profile of PGA make it an attractive candidate for vascular tissue engineering (35). Numerous studies have demonstrated the successful use of PGA scaffolds for constructing vascular grafts (7,31,32,34,36). In the current study, cells in the experimental group were demonstrated to possess good proliferative ability and ECM secretion on PGA.

In the histological examination, it was identified that the content and distribution of SMCs and elastin in the experimental group were not as dense as those in normal blood vessels. Normal blood vessels have been demonstrated to develop under the influence of the mechanical force of blood, which is an important physiological component of the environment experienced by cells: It promotes the circumferential orientation of the cells in addition to the deposition of the extracellular matrix, and it likely contributes to the survival of implanted substitutes (36). It is suggested that dynamic culturing is important in vascular tissue engineering. In vitro investigations have demonstrated that low shear stress induces SMC proliferation (3740) and promotes collagen alignment (41) and that cyclic stretching induces clear alterations in the SMC phenotype, function and gene expression (42,43). SMCs use multiple sensing mechanisms to perceive the mechanical stimulus generated from pulsatile stretching and transduce it into intracellular signals. This results in the modulation of gene expression and cellular functions including proliferation, apoptosis, migration and remodelling (4449). Therefore, a bioreactor, which is currently is under development, may be used in future studies to mimic the physiological environment of the arterial vessel wall.

In conclusion, a large vessel (6 mm in diameter) was constructed using PGA seeded with hHFSCs in vitro. Induced culture constructs exhibited improved performance histologically and in hydroxyproline content when compared with constructs of the undifferentiated group. Further research focused on dynamic culturing of the constructs and further seeding of endothelial cells on the surface of the lumen to engineer composite vascular conduits is required, in order to progress in this area of bio-engineering.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81000842). The authors would additionally like to thank Mr Demin Ying, Mrs Lijuan Zong and Mr Bing Zhong for their technical assistance.

References

1 

Kohn JC, Lampi MC and Reinhart-King CA: Age-related vascular stiffening: Causes and consequences. Front Genet. 6:1122015.PubMed/NCBI

2 

Lee AY, Mahler N, Best C, Lee YU and Breuer CK: Regenerative implants for cardiovascular tissue engineering. Transl Res. 163:321–341. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Kurobe H, Maxfield MW, Breuer CK and Shinoka T: Concise review: Tissue-engineered vascular grafts for cardiac surgery: Past, present, and future. Stem Cells Transl Med. 1:566–571. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Nemeno-Guanzon JG, Lee S, Berg JR, Jo YH, Yeo JE, Nam BM, Koh YG and Lee JI: Trends in tissue engineering for blood vessels. J Biomed Biotechnol. 2012:9563452012.PubMed/NCBI

5 

Teebken OE and Haverich A: Tissue engineering of small diameter vascular grafts. Eur J Vasc Endovasc Surg. 23:475–485. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Wang Y, Hu J, Jiao J, Liu Z, Zhou Z, Zhao C, Chang LJ, Chen YE, Ma PX and Yang B: Engineering vascular tissue with functional smooth muscle cells derived from human iPS cells and nanofibrous scaffolds. Biomaterials. 35:8960–8969. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Wang C, Cen L, Yin S, Liu Q, Liu W, Cao Y and Cui L: A small diameter elastic blood vessel wall prepared under pulsatile conditions from polyglycolic acid mesh and smooth muscle cells differentiated from adipose-derived stem cells. Biomaterials. 31:621–630. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Wilhelmi M, Jockenhoevel S and Mela P: Bioartificial fabrication of regenerating blood vessel substitutes: Requirements and current strategies. Biomed Tech (Berl). 59:185–195. 2014.PubMed/NCBI

9 

Nerem RM and Seliktar D: Vascular tissue engineering. Annu Rev Biomed Eng. 3:225–243. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Naito Y, Shinoka T, Duncan D, Hibino N, Solomon D, Cleary M, Rathore A, Fein C, Church S and Breuer C: Vascular tissue engineering: Towards the next generation vascular grafts. Adv Drug Deliv Rev. 63:312–323. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Khait L and Birla RK: Bypassing the patient: Comparison of biocompatible models for the future of vascular tissue engineering. Cell Transplant. 21:269–283. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Zhu GC, Gu YQ, Geng X, Feng ZG, Zhang SW, Ye L and Wang ZG: Experimental study on the construction of small three-dimensional tissue engineered grafts of electrospun poly-ε-caprolactone. J Mater Sci Mater Med. 26:1122015. View Article : Google Scholar : PubMed/NCBI

13 

Koobatian MT, Liang MS, Swartz DD and Andreadis ST: Differential effects of culture senescence and mechanical stimulation on the proliferation and leiomyogenic differentiation of MSC from different sources: Implications for engineering vascular grafts. Tissue Eng Part A. 21:1364–1375. 2015. View Article : Google Scholar : PubMed/NCBI

14 

GN, Tan A, Gundogan B, Farhatnia Y, Nayyer L, Mahdibeiraghdar S, Rajadas J, De Coppi P, Davies AH and Seifalian AM: Tissue engineering vascular grafts a fortiori: Looking back and going forward. Expert Opin Biol Ther. 15:231–244. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Sundaram S, One J, Siewert J, Teodosescu S, Zhao L, Dimitrievska S, Qian H, Huang AH and Niklason L: Tissue-engineered vascular grafts created from human induced pluripotent stem cells. Stem Cells Transl Med. 3:1535–1543. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Rammal H, Harmouch C, Lataillade JJ, Laurent-Maquin D, Labrude P, Menu P and Kerdjoudj H: Stem cells: A promising source for vascular regenerative medicine. Stem Cells Dev. 23:2931–2949. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Heydarkhan-Hagvall S, Schenke-Layland K, Yang JQ, Heydarkhan S, Xu Y, Zuk PA, MacLellan WR and Beygui RE: Human adipose stem cells: A potential cell source for cardiovascular tissue engineering. Cells Tissues Organs. 187:263–274. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Harris LJ, Abdollahi H, Zhang P, McIlhenny S, Tulenko TN and DiMuzio PJ: Differentiation of adult stem cells into smooth muscle for vascular tissue engineering. J Surg Res. 168:306–314. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Hsu YC, Pasolli HA and Fuchs E: Dynamics between stem cells, niche, and progeny in the hair follicle. Cell. 144:92–105. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Xu ZC, Zhang Q and Li H: Human hair follicle stem cell differentiation into contractile smooth muscle cells is induced by transforming growth factor-β1 and platelet-derived growth factor BB. Mol Med Rep. 8:1715–1721. 2013.PubMed/NCBI

21 

Jahoda CA, Whitehouse J, Reynolds AJ and Hole N: Hair follicle dermal cells differentiate into adipogenic and osteogenic lineages. Exp Dermatol. 12:849–859. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Yu H, Fang D, Kumar SM, Li L, Nguyen TK, Acs G, Herlyn M and Xu X: Isolation of a novel population of multipotent adult stem cells from human hair follicles. Am J Pathol. 168:1879–1888. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Reddy GK and Ewemeka CS: A simplified method for the analysis of hydroxyproline in biological tissues. Clin Biochem. 29:225–229. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Naito Y, Rocco K, Kurobe H, Maxfield M, Breuer C and Shinoka T: Tissue engineering in the vasculature. Anat Rec (Hoboken). 297:83–97. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Drewa T, Joachimiak R, Kaznica A, Sarafian V and Pokrywczynska M: Hair stem cells for bladder regeneration in rats: Preliminary results. Transplant Proc. 41:4345–4351. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Lin H, Liu F, Zhang C, Zhang Z, Kong Z, Zhang X and Hoffman RM: Characterization of nerve conduits seeded with neurons and Schwann cells derived from hair follicle neural crest stem cells. Tissue Eng Part A. 17:1691–1698. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, et al: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 418:41–49. 2002. View Article : Google Scholar : PubMed/NCBI

28 

DiMuzio P and Tulenko T: Tissue engineering applications to vascular bypass graft development: The use of adipose-derived stem cells. J Vasc Surg. 45:(Suppl A). A99–A103. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Collins MN and Birkinshaw C: Hyaluronic acid based scaffolds for tissue engineering-a review. Carbohydr Polym. 92:1262–1279. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Lee SJ, Liu J, Oh SH, Soker S, Atala A and Yoo JJ: Development of a composite vascular scaffolding system that withstands physiological vascular conditions. Biomaterials. 29:2891–2898. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Pankajakshan D and Agrawal DK: Scaffolds in tissue engineering of blood vessels. Can J Physiol Pharmacol. 88:855–873. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Gui L, Zhao L, Spencer RW, Burghouwt A, Taylor MS, Shalaby SW and Niklason LE: Development of novel biodegradable polymer scaffolds for vascular tissue engineering. Tissue Eng Part A. 17:1191–1200. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Bačáková L, Novotná K and Pařízek M: Polysaccharides as cell carriers for tissue engineering: The use of cellulose in vascular wall reconstruction. Physiol Res. 63:(Suppl 1). S29–S47. 2014.PubMed/NCBI

34 

Thottappillil N and Nair PD: Scaffolds in vascular regeneration: Current status. Vasc Health Risk Manag. 11:79–91. 2015.PubMed/NCBI

35 

Dong Y, Yong T, Liao S, Chan CK, Stevens MM and Ramakrishna S: Distinctive degradation behaviors of electrospun polyglycolide, poly (dl-lactide-co-glycolide), and poly (l-Lactide-co-epsilon-caprolactone) nanofibers cultured with/without porcine smooth muscle cells. Tissue Eng Part A. 16:283–298. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Xu ZC, Zhang Q and Li H: An elastic large muscular vessel wall engineered with bone marrow-derived cells under pulsatile stimulation in a bioreactor. Mol Med Rep. 12:6005–6012. 2015.PubMed/NCBI

37 

Sterpetti AV, Cucina A, Santoro L, Cardillo B and Cavallaro A: Modulation of arterial smooth muscle cell growth by haemodynamic forces. Eur J Vasc Surg. 6:16–20. 1992. View Article : Google Scholar : PubMed/NCBI

38 

Sho M, Sho E, Singh TM, Komatsu M, Sugita A, Xu C, Nanjo H, Zarins CK and Masuda H: Subnormalshear stress-induced intimal thickening requires medial smooth muscle cell proliferation and migration. Exp Mol Pathol. 72:150–160. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Palumbo R, Gaetano C, Antonini A, Pompilio G, Bracco E, Rönnstrand L, Heldin CH and Capogrossi MC: Different effects of high and low shear stress on platelet-derived growth factor isoform release by endothelial cells: Consequences for smooth muscle cell migration. Arterioscler Thromb Vasc Biol. 22:405–411. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Qi YX, Qu MJ, Long DK, Liu B, Yao QP, Chien S and Jiang ZL: Rho-GDP dissociation inhibitor alpha downregulated by low shear stress promotes vascular smooth muscle cell migration and apoptosis: A proteomic analysis. Cardiovasc Res. 80:114–122. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Ng CP, Hinz B and Swartz MA: Interstitial fluid flow induces myofibroblast differentiation and collagen alignment in vitro. J Cell Sci. 118:4731–4739. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Halka AT, Turner NJ, Carter A, Ghosh J, Murphy MO, Kirton JP, Kielty CM and Walker MG: The effects of stretch on vascular smooth muscle cell phenotype in vitro. Cardiovasc Pathol. 17:98–102. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Jakkaraju S, Zhe X and Schuger L: Role of stretch in activation of smooth muscle cell lineage. Trends Cardiovasc Med. 13:330–335. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Haga JH, Li YS and Chien S: Molecular basis of the effects of mechanical stretch on vascular smooth muscle cells. J Biomech. 40:947–960. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Standley PR, Cammarata A, Nolan BP, Purgason CT and Stanley MA: Cyclic stretch induces vascular smooth muscle cell alignment via NO signaling. Am J Physiol Heart Circ Physiol. 283:H1907–H1914. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Zhu JH, Chen CL, Flavahan S, Harr J, Su B and Flavahan NA: Cyclic stretch stimulates vascular smooth muscle cell alignment by redox-dependent activation of Notch3. Am J Physiol Heart Circ Physiol. 300:H1770–H1780. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Chapman GB, Durante W, Hellums JD and Schafer AI: Physiological cyclic stretch causes cell cycle arrest in cultured vascular smooth muscle cells. Am J Physiol Heart Circ Physiol. 278:H748–H754. 2000.PubMed/NCBI

48 

Iwasaki H, Yoshimoto T, Sugiyama T and Hirata Y: Activation of cell adhesion kinase beta by mechanical stretch in vascular smooth muscle cells. Endocrinology. 144:2304–2310. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Qiu J, Zheng Y, Hu J, Liao D, Gregersen H, Deng X, Fan Y and Wang G: Biomechanical regulation of vascular smooth muscle cell functions: From in vitro to in vivo understanding. J R Soc Interface. 11:201308522013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2017
Volume 15 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu ZC, Zhang Q and Li H: Engineering of the human vessel wall with hair follicle stem cells in vitro. Mol Med Rep 15: 417-422, 2017
APA
Xu, Z., Zhang, Q., & Li, H. (2017). Engineering of the human vessel wall with hair follicle stem cells in vitro. Molecular Medicine Reports, 15, 417-422. https://doi.org/10.3892/mmr.2016.6013
MLA
Xu, Z., Zhang, Q., Li, H."Engineering of the human vessel wall with hair follicle stem cells in vitro". Molecular Medicine Reports 15.1 (2017): 417-422.
Chicago
Xu, Z., Zhang, Q., Li, H."Engineering of the human vessel wall with hair follicle stem cells in vitro". Molecular Medicine Reports 15, no. 1 (2017): 417-422. https://doi.org/10.3892/mmr.2016.6013