51
|
Breitenbach J, Gruber C, Klausegger A, Trost A, Bogner B, Reitsamer H, Bauer JW. Pseudosyndactyly - an inflammatory and fibrotic wound healing disorder in recessive dystrophic epidermolysis bullosa. J Dtsch Dermatol Ges 2016; 13:1257-66. [PMID: 26612796 DOI: 10.1111/ddg.12839] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND A genetic blistering skin disease, recessive dystrophic epidermolysis bullosa (RDEB), is marked by severe wound healing defects and finger contractures. The purpose of this investigation was to elucidate the mechanisms of impaired wound healing and pseudosyndactyly occurring in RDEB patients by studying the role of known inflammation and fibrosis markers in RDEB pseudosyndactyly tissue. PATIENTS AND METHODS We studied the expression of the fibrosis and/or inflammation markers tenascin-C, α-smooth muscle actin, transforming growth factor-β1, interleukin-1β, and interleukin-6 in scarring and nonscarring tissue from healthy donors and RDEB patients by semiquantitative real time-PCR and, where applicable, by immunoblots. Furthermore, the distribution pattern of α-smooth muscle actin and tenascin-C were assessed by immunofluorescence microscopy. RESULTS Based on mRNA and protein analysis, we found upregulation of tenascin-C, interleukin-1β, and interleukin-6 - but not of transforming growth factor-β1 - in recessive dystrophic epidermolysis bullosa scar samples taken from pseudosyndactyly hands. Unexpectedly, α-smooth muscle actin was not upregulated. CONCLUSIONS Our results confirm inflammation and fibrosis in recessive dystrophic epidermolysis bullosa, especially in scars, suggesting major roles for these processes in pseudosyndactyly. Our data therefore suggests the potential use of antiinflammatory and antifibrotic drugs in the prevention of pseudosyndactyly.
Collapse
Affiliation(s)
- Jenny Breitenbach
- Cluster Rheumatology, Balneology and Rehabilitation, Vienna, Austria
| | - Christina Gruber
- Research Program for Molecular Therapy of Genodermatoses, EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Alfred Klausegger
- Research Program for Molecular Therapy of Genodermatoses, EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Andrea Trost
- Department of Ophthalmology and Optometry, Research program for Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Barbara Bogner
- Department of Ophthalmology and Optometry, Research program for Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Herbert Reitsamer
- Department of Ophthalmology and Optometry, Research program for Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Johann W Bauer
- Research Program for Molecular Therapy of Genodermatoses, EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
52
|
The isolation and characterization of systemic sclerosis vascular smooth muscle cells: enhanced proliferation and apoptosis resistance. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2016. [DOI: 10.5301/jsrd.5000218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background Vascular dysfunction is a major pathologic component of systemic sclerosis (SSc). The role of vascular smooth muscle cells (vSMCs) in the development of vascular dysfunction is still unknown. In this study, we describe a method for isolation of dermal vSMCs from skin biopsies, and we outline their functional phenotype. Methods We obtained 4 mm punch-skin biopsies from three SSc patients and matched controls. After trypsin digestion, cells were cultured for 14 days. vSMCs were isolated by first depleting CD31+ cells (endothelial cells), followed by positive selection of CD146+ cells. The CD31- CD146+ cells were then cultured in media optimized for SMCs proliferation. We evaluated cell proliferation, viability and apoptosis in normal and in low serum culture conditions. Cytoplasmic and nuclear expression levels of β-catenin were also investigated. Results The CD31- CD146+ cell population expressed smooth muscle MYH11, Desmin and Vimentin but did not express NG2. Flow cytometry confirmed the high purity of CD31- CD146+ MYHC11+ cell population that was maintained for up to the eleventh passage. SSc-vSMCs exhibited increased cell proliferation and viability compared to control cells. Under serum starvation conditions, SSc-vSMCs exhibited more proliferative capacity, and resistance to apoptosis compared to control-vSMCs. Furthermore, a cytoplasmic to nuclear translocation of β-catenin was seen in SSc-vSMCs but not in control-vSMCs. Conclusions This is the first report of successful isolation and initial characterization of SSc-vSMCs. It is likely that increased proliferation of SSc-vSMCs in association with resistance to apoptosis can adversely impact the vascular lesion in SSc.
Collapse
|
53
|
Yamada M, Kurihara H, Kinoshita K, Sakai T. Temporal Expression of Alpha–Smooth Muscle Actin and Drebrin in Septal Interstitial Cells during Alveolar Maturation. J Histochem Cytochem 2016; 53:735-44. [PMID: 15928322 DOI: 10.1369/jhc.4a6483.2005] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In rat lung, the definitive alveoli are established during development by the outgrowth of secondary septa from the primary septa present in newborn; however, the mechanism of alveolar formation has not yet been fully clarified. In this study, we characterize the septal interstitial cells in developing alveoli. During the perinatal period, alpha-SMA–containing slender cells were found in the primitive alveolar septa. Alpha-SMA–containing cells were detected at the tips of the septa until postnatal day 21, when the alveolar formation was almost completed, but disappeared in adult. Immunoelectron microscopy demonstrated that alpha-SMA is localized mainly in the cellular protrusions, which are connected with the elastic fibers around the interstitial cells. Developmentally regulated brain protein (drebrin) is also located in the cell extensions containing alpha-SMA in immature alveolar interstitial cells. In adult lung, alpha-SMA–positive cells are located only at the alveolar ducts but are not found in the secondary septa. Desmin is expressed only in alpha-SMA–containing cells at the alveolar ducts but not in those at the tip of alveolar septa. These results suggest that a part of the septal interstitial cells are temporarily alpha-SMA– and drebrin-positive during maturation. Alpha-SMA– and drebrin-containing septal interstitial cells (termed septal myofibroblast-like cells) may play an important role in alveolar formation.
Collapse
Affiliation(s)
- Mie Yamada
- Department of Anatomy, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | | | | | | |
Collapse
|
54
|
Hall AP, Westwood FR, Wadsworth PF. Review of the Effects of Anti-Angiogenic Compounds on the Epiphyseal Growth Plate. Toxicol Pathol 2016; 34:131-47. [PMID: 16537292 DOI: 10.1080/01926230600611836] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The formation of new blood vessels from a pre-existing vascular bed, termed “angiogenesis,” is of critical importance for the growth and development of the animal since it is required for the growth of the skeleton during endochondral ossification, development and cycling of the corpus luteum and uterus, and for the repair of tissues during wound healing. “Vasculogenesis,” the de novo formation of blood vessels is also important for the proper function and development of the vascular system in the embryo. New blood vessel formation is a prominent feature and permissive factor in the relentless progression of many human diseases, one of the most important examples of which is neoplasia. It is for this reason that angiogenesis is considered to be one of the hallmarks of cancer. The development of new classes of drugs that inhibit the growth and proper functioning of new blood vessels in vivo is likely to provide significant therapeutic benefit in the treatment of cancer, as well as other conditions where angiogenesis is a strong driver to the disease process. During the preclinical safety testing of these drugs, it is becoming increasingly clear that their in vivo efficacy is reflected in the profile of “expected toxicity” (resulting from pharmacology) observed in laboratory animals, so much so, that this profile of “desired” toxicity may act as a signature for their anti-angiogenic effect. In this article we review the major mechanisms controlling angiogenesis and its role during endochondral ossification. We also review the effects of perturbation of endochondral ossification through four mechanisms—inhibition of vascular endothelial growth factor (VEGF), pp60 c-Src kinase and matrix metalloproteinases as well as disruption of the blood supply with vascular targeting agents. Inhibition through each of these mechanisms appears to have broadly similar effects on the epiphyseal growth plate characterised by thickening due to the retention of hypertrophic chondrocytes resulting from the inhibition of angiogenesis. In contrast, in the metaphysis there are differing effects reflecting the specific role of these targets at this site.
Collapse
Affiliation(s)
- Anthony P Hall
- AstraZeneca, Mereside, Alderley Park, Macclesfield, Cheshire SK10 4TG, England.
| | | | | |
Collapse
|
55
|
Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle Disorders. Pharmacol Rev 2016; 68:476-532. [PMID: 27037223 PMCID: PMC4819215 DOI: 10.1124/pr.115.010652] [Citation(s) in RCA: 346] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The smooth muscle cell directly drives the contraction of the vascular wall and hence regulates the size of the blood vessel lumen. We review here the current understanding of the molecular mechanisms by which agonists, therapeutics, and diseases regulate contractility of the vascular smooth muscle cell and we place this within the context of whole body function. We also discuss the implications for personalized medicine and highlight specific potential target molecules that may provide opportunities for the future development of new therapeutics to regulate vascular function.
Collapse
Affiliation(s)
- F V Brozovich
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C J Nicholson
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C V Degen
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - Yuan Z Gao
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - M Aggarwal
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - K G Morgan
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| |
Collapse
|
56
|
Surendran S, S Ramegowda K, Suresh A, Binil Raj SS, Lakkappa RKB, Kamalapurkar G, Radhakrishnan N, C Kartha C. Arterialization and anomalous vein wall remodeling in varicose veins is associated with upregulated FoxC2-Dll4 pathway. J Transl Med 2016; 96:399-408. [PMID: 26808710 DOI: 10.1038/labinvest.2015.167] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/20/2015] [Accepted: 12/23/2015] [Indexed: 11/09/2022] Open
Abstract
Varicose veins of lower extremities are a heritable common disorder. Mechanisms underlying its pathogenesis are still vague. Structural failures such as valve weakness and wall dilatation in saphenous vein result in venous retrograde flow in lower extremities of body. Reflux of blood leads to distal high venous pressure resulting in distended veins. In an earlier study, we observed a positive association between c.-512C>T FoxC2 gene polymorphism and upregulated FoxC2 expression in varicose vein specimens. FoxC2 overexpression in vitro in venous endothelial cells resulted in the elevated mRNA expression of arterial endothelial markers such as Delta-like ligand 4 (Dll4) and Hairy/enhancer-of-split related with YRPW motif protein 2 (Hey2). We hypothesized that an altered FoxC2-Dll4 signaling underlies saphenous vein wall remodeling in patients with varicose veins. Saphenous veins specimens were collected from 22 patients with varicose veins and 20 control subjects who underwent coronary artery bypass grafting. Tissues were processed for paraffin embedding and sections were immunostained for Dll4, Hey2, EphrinB2, α-SMA, Vimentin, and CD31 antigens and examined under microscope. These observations were confirmed by quantitative real-time PCR and western blot analysis. An examination of varicose vein tissue specimens by immunohistochemistry indicated an elevated expression of Notch pathway components, such as Dll4, Hey2, and EphrinB2, and smooth muscle markers, which was further confirmed by gene and protein expression analyses. We conclude that the molecular alterations in Dll4-Hey2 signaling are associated with smooth muscle cell hypertrophy and hyperplasia in varicose veins. Our observations substantiate a significant role for altered FoxC2-Dll4 signaling in structural alterations of saphenous veins in patients with varicose veins.
Collapse
Affiliation(s)
- Sumi Surendran
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Kalpana S Ramegowda
- Departments of Pathology & Cardiovascular Surgery, Sri Jayadeva Institute for Cardiovascular Sciences and Research, Bangalore, Bangalore, India
| | - Aarcha Suresh
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - S S Binil Raj
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Ravi Kumar B Lakkappa
- Department of Vascular Surgery, Kempegowda Institute of Medical Sciences, Bangalore, Bangalore, India
| | - Giridhar Kamalapurkar
- Departments of Pathology & Cardiovascular Surgery, Sri Jayadeva Institute for Cardiovascular Sciences and Research, Bangalore, Bangalore, India
| | - N Radhakrishnan
- St Thomas Institute of Research on Venous Diseases, Changanassery, Kerala, India
| | - Chandrasekharan C Kartha
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
57
|
Breitenbach J, Gruber C, Klausegger A, Trost A, Bogner B, Reitsamer H, Bauer JW. Pseudosyndaktylie - eine entzündliche und fibrotische Wundheilungsstörung bei rezessiver Epidermolysis bullosa dystrophica. J Dtsch Dermatol Ges 2015. [DOI: 10.1111/ddg.140_12839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Jenny Breitenbach
- Ludwig Boltzmann-Institut für Rheumatologie und Balneologie Cluster für Rheumatologie, Balneologie und Rehabilitation Wien, Österreich
| | - Christina Gruber
- Forschungsprogramm für Molekulare Therapie bei Genodermatosen; EB-Haus Austria; Universitätsklinik für Dermatologie; Universitätsklinikum der Paracelsus Medizinischen Privatuniversität; Salzburg Österreich
| | - Alfred Klausegger
- Forschungsprogramm für Molekulare Therapie bei Genodermatosen; EB-Haus Austria; Universitätsklinik für Dermatologie; Universitätsklinikum der Paracelsus Medizinischen Privatuniversität; Salzburg Österreich
| | - Andrea Trost
- Universitätsklinik für Augenheilkunde und Optometrie; Forschungsprogramm für Experimentelle Ophtalmologie und Glaukomforschung; Paracelsus Medizinische Privatuniversität; Salzburg Österreich
| | - Barbara Bogner
- Universitätsklinik für Augenheilkunde und Optometrie; Forschungsprogramm für Experimentelle Ophtalmologie und Glaukomforschung; Paracelsus Medizinische Privatuniversität; Salzburg Österreich
| | - Herbert Reitsamer
- Universitätsklinik für Augenheilkunde und Optometrie; Forschungsprogramm für Experimentelle Ophtalmologie und Glaukomforschung; Paracelsus Medizinische Privatuniversität; Salzburg Österreich
| | - Johann W. Bauer
- Forschungsprogramm für Molekulare Therapie bei Genodermatosen; EB-Haus Austria; Universitätsklinik für Dermatologie; Universitätsklinikum der Paracelsus Medizinischen Privatuniversität; Salzburg Österreich
| |
Collapse
|
58
|
Tonar Z, Kubíková T, Prior C, Demjén E, Liška V, Králíčková M, Witter K. Segmental and age differences in the elastin network, collagen, and smooth muscle phenotype in the tunica media of the porcine aorta. Ann Anat 2015; 201:79-90. [DOI: 10.1016/j.aanat.2015.05.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 12/18/2022]
|
59
|
Akbareian SE, Pitsillides AA, Macharia RG, McGonnell IM. Occipital foramina development involves localised regulation of mesenchyme proliferation and is independent of apoptosis. J Anat 2015; 226:560-74. [PMID: 25994127 DOI: 10.1111/joa.12304] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2015] [Indexed: 01/14/2023] Open
Abstract
Cranial foramina are holes within the skull, formed during development, allowing entry and exit of blood vessels and nerves. Once formed they must remain open, due to the vital structures they contain, i.e. optic nerves, jugular vein, carotid artery, and other cranial nerves and blood vessels. Understanding cranial foramina development is essential as cranial malformations lead to the stenosis or complete closure of these structures, resulting in blindness, deafness, facial paralysis, raised intracranial pressure and lethality. Here we focus on describing early events in the formation of the jugular, carotid and hypoglossal cranial foramina that form in the mesoderm-derived, endochondral occipital bones at the base of the embryonic chick skull. Whole-mount skeletal staining of skulls indicates the appearance of these foramina from HH32/D7.5 onwards. Haematoxylin & eosin staining of sections shows that the intimately associated mesenchyme, neighbouring the contents of these cranial foramina, is initially very dense and gradually becomes sparser as development proceeds. Histological examination also revealed that these foramina initially contain relatively large-diameter nerves, which later become refined, and are closely associated with the blood vessel, which they also innervate within the confines of the foramina. Interestingly cranial foramina in the base of the skull contain blood vessels lacking smooth muscle actin, which suggests these blood vessels belong to glomus body structures within the foramina. The blood vessel shape also appears to dictate the overall shape of the resulting foramina. We initially hypothesised that cranial foramina development could involve targeted proliferation and local apoptosis to cause 'mesenchymal clearing' and the creation of cavities in a mechanism similar to joint cavitation. We find that this is not the case, and propose that a mechanism reliant upon local nerve/blood vessel-derived restriction of ossification may contribute to foramina formation during cranial development.
Collapse
Affiliation(s)
- Sophia E Akbareian
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Andrew A Pitsillides
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Raymond G Macharia
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Imelda M McGonnell
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| |
Collapse
|
60
|
Ye GJC, Nesmith AP, Parker KK. The role of mechanotransduction on vascular smooth muscle myocytes' [corrected] cytoskeleton and contractile function. Anat Rec (Hoboken) 2015; 297:1758-69. [PMID: 25125187 DOI: 10.1002/ar.22983] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 06/06/2014] [Indexed: 12/29/2022]
Abstract
Smooth muscle (SM) exhibits a highly organized structural hierarchy that extends over multiple spatial scales to perform a wide range of functions at the cellular, tissue, and organ levels. Early efforts primarily focused on understanding vascular SM (VSM) function through biochemical signaling. However, accumulating evidence suggests that mechanotransduction, the process through which cells convert mechanical stimuli into biochemical cues, is requisite for regulating contractility. Cytoskeletal proteins that comprise the extracellular, intercellular, and intracellular domains are mechanosensitive and can remodel their structure and function in response to external mechanical cues. Pathological stimuli such as malignant hypertension can act through the same mechanotransductive pathways to induce maladaptive remodeling, leading to changes in cellular shape and loss of contractile function. In both health and disease, the cytoskeletal architecture integrates the mechanical stimuli and mediates structural and functional remodeling in the VSM.
Collapse
Affiliation(s)
- George J C Ye
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering and the School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | | | | |
Collapse
|
61
|
Wilson JL, Yu J, Taylor L, Polgar P. Hyperplastic Growth of Pulmonary Artery Smooth Muscle Cells from Subjects with Pulmonary Arterial Hypertension Is Activated through JNK and p38 MAPK. PLoS One 2015; 10:e0123662. [PMID: 25905460 PMCID: PMC4408087 DOI: 10.1371/journal.pone.0123662] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/06/2015] [Indexed: 12/20/2022] Open
Abstract
Smooth muscle in the pulmonary artery of PAH subjects, both idiopathic and hereditary, is characterized by hyperplasia. Smooth muscle cells (HPASMC) isolated from subjects with or without PAH retain their in vivo phenotype as illustrated by their expression of alpha-smooth muscle actin and expression of H-caldesmon. Both non PAH and PAH HPASMC display a lengthy, approximately 94h, cell cycle. The HPASMC from both idiopathic and hereditary PAH display an abnormal proliferation characterized by continued growth under non-proliferative, non-growth stimulated conditions. This effector independent proliferation is JNK and p38 MAP kinase dependent. Blocking the activation of either abrogates the HPASMC growth. HPASMC from non PAH donors under quiescent conditions display negligible proliferation but divide upon exposure to growth factors such as PDGF-BB or FGF2 but not EGF. This growth does not involve the MAP kinases. Instead it routes via the tyrosine kinase receptor through mTOR and then 6SK. In the PAH cells PDGF-BB and FGF2 augment the dysregulated cell proliferation, also through mTOR/6SK. Additionally, blocking the activation of mTOR also modulates the MAP kinase promoted dysregulated growth. These results highlight key alterations in the growth of HPASMC from subjects with PAH which contribute to the etiology of the disease and can clearly be targeted at various regulatory points for future therapies.
Collapse
Affiliation(s)
- Jamie L. Wilson
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jun Yu
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Linda Taylor
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Peter Polgar
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
62
|
Tardif K, Hertig V, Duquette N, Villeneuve L, El-Hamamsy I, Tanguay JF, Calderone A. Nestin upregulation characterizes vascular remodeling secondary to hypertension in the rat. Am J Physiol Heart Circ Physiol 2015; 308:H1265-74. [PMID: 25770244 DOI: 10.1152/ajpheart.00804.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/03/2015] [Indexed: 11/22/2022]
Abstract
Proliferation and hypertrophy of vascular smooth muscle cells represent hallmark features of vessel remodeling secondary to hypertension. The intermediate filament protein nestin was recently identified in vascular smooth muscle cells and in other cell types directly participated in proliferation. The present study tested the hypothesis that vessel remodeling secondary to hypertension was characterized by nestin upregulation in vascular smooth muscle cells. Two weeks after suprarenal abdominal aorta constriction of adult male Sprague-Dawley rats, elevated mean arterial pressure increased the media area and thickness of the carotid artery and aorta and concomitantly upregulated nestin protein levels. In the normal adult rat carotid artery, nestin immunoreactivity was observed in a subpopulation of vascular smooth muscle cells, and the density significantly increased following suprarenal abdominal aorta constriction. Filamentous nestin was detected in cultured rat carotid artery- and aorta-derived vascular smooth muscle cells and an analogous paradigm observed in human aorta-derived vascular smooth muscle cells. ANG II and EGF treatment of vascular smooth muscle cells stimulated DNA and protein synthesis and increased nestin protein levels. Lentiviral short-hairpin RNA-mediated nestin depletion of carotid artery-derived vascular smooth muscle cells inhibited peptide growth factor-stimulated DNA synthesis, whereas protein synthesis remained intact. These data have demonstrated that vessel remodeling secondary to hypertension was characterized in part by nestin upregulation in vascular smooth muscle cells. The selective role of nestin in peptide growth factor-stimulated DNA synthesis has revealed that the proliferative and hypertrophic responses of vascular smooth muscle cells were mediated by divergent signaling events.
Collapse
Affiliation(s)
- Kim Tardif
- Program in Biomedical Sciences, Université de Montréal, Montréal, Québec, Canada; Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Vanessa Hertig
- Departement of Physiology, Université de Montréal, Montréal, Québec, Canada; and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Natacha Duquette
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Ismail El-Hamamsy
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Jean-François Tanguay
- Program in Biomedical Sciences, Université de Montréal, Montréal, Québec, Canada; Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Angelino Calderone
- Departement of Physiology, Université de Montréal, Montréal, Québec, Canada; and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
63
|
Intranodal palisaded myofibroblastoma: another mesenchymal neoplasm with CTNNB1 (β-catenin gene) mutations: clinicopathologic, immunohistochemical, and molecular genetic study of 18 cases. Am J Surg Pathol 2015; 39:197-205. [PMID: 25025452 DOI: 10.1097/pas.0000000000000299] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Intranodal palisaded myofibroblastoma is a benign, lymph node-based myofibroblastic tumor of unknown pathogenesis. We report the clinicopathologic, immunohistochemical, and molecular genetic features of this rare entity. The study cohort consisted of 14 men and 4 women ranging in age from 31 to 65 (mean, 47; median 49) years with tumors arising in inguinal lymph nodes (n=15), a neck lymph node (n=1), and undesignated lymph nodes (n=2). Most individuals presented with a painless mass or lump. Possible trauma/injury to the inguinal region was documented in 4 cases. Tumors ranged in size from 1.0 to 4.2 (mean, 3.1; median; 3.0) cm. Microscopically, the process presented as a well-circumscribed, oftentimes pseudoencapsulated nodule (n=17) or nodules (n=1). Tumors consisted of a cellular proliferation of cytologically bland, spindled cells arranged in short fascicles and whorls within a finely collagenous (n=11) or myxocollagenous (n=7) matrix. In 12 tumors, scattered fibromatosis-like fascicles of spindled cells were noted. Histologic features characteristic of the process included nuclear palisades (n=16 cases), collagenous bodies (n=15), and perinuclear intracytoplasmic hyaline globules (n=10). Mitotic activity ranged from 0 to 8 (mean, 2; median, 1) mitotic figures/50 high-powered fields with no atypical division figures identified. Immunohistochemically, all tumors tested expressed smooth muscle actin and/or muscle-specific actin (n=5, each), and nuclear β-catenin and cyclin D1 (n=8, each). The latter 2 results prompted a screening for mutations in the β-catenin gene glycogen synthase kinase-3 β phosphorylation mutational "hotspot" region in exon 3 using polymerase chain reaction amplification and Sanger sequencing. Single nucleotide substitutions leading to missense mutations at the protein level were identified in 7 of 8 (88%) analyzed tumors and are responsible for the abnormal expression of β-catenin and cyclin D1. These results demonstrate that mutational activation of the β-catenin gene is likely a pivotal event in the pathogenesis of intranodal palisaded myofibroblastoma.
Collapse
|
64
|
Differential expression and regulation of Klotho by paricalcitol in the kidney, parathyroid, and aorta of uremic rats. Kidney Int 2015; 87:1141-52. [PMID: 25692955 PMCID: PMC4449811 DOI: 10.1038/ki.2015.22] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 11/21/2014] [Accepted: 12/11/2014] [Indexed: 12/29/2022]
Abstract
Klotho plays an important role in the pathogenesis of cardiovascular disease in chronic kidney disease (CKD). Klotho is highly expressed in the kidney and parathyroid glands, but its presence in the vasculature is debated. Renal Klotho is decreased in CKD, but the effect of uremia on Klotho in other tissues is not defined. The effect of vitamin D receptor activator therapy in CKD on expression of Klotho in various tissues is also in debate. In uremic rats (surgical 5/6th nephrectomy model), we compared 3-months of treatment with and without paricalcitol on Klotho immunostaining in the kidney, parathyroid glands and aorta. With uremia, Klotho was unchanged in the parathyroid, significantly decreased in the kidney (66%) and the intimal-medial area of the aorta (69%), and significantly increased in the adventitial area of the aorta (67%) compared with controls. Paricalcitol prevented the decrease in Klotho in the kidney, increased expression in the parathyroid (31%), had no effect in the aortic media, but blunted the increase of Klotho in aortic adventitia. We propose that fibroblasts are responsible for expression of Klotho in the adventitia. In hyperplastic human parathyroid tissue from uremic patients, Klotho was higher in oxyphil compared with chief cells. Thus, under our conditions of moderate CKD and mild-to-moderate hyperphosphatemia in rats, the differential expression of Klotho and its regulation by paricalcitol in uremia is tissue-dependent.
Collapse
|
65
|
Fat Graft Viability in the Subcutaneous Plane versus the Local Fat Pad. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2015; 2:e260. [PMID: 25587494 PMCID: PMC4292242 DOI: 10.1097/gox.0000000000000228] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/08/2014] [Indexed: 01/22/2023]
Abstract
Background: Fat grafting has been increasingly utilized in both aesthetic and reconstructive surgical procedures, yet the basic scientific understanding of fat grafting has lagged behind the pace of clinical innovation and utilization. This lack of basic scientific understanding has perhaps manifested itself in the wide range of graft viability reported across the literature. This study attempts to further the underlying mechanisms of fat graft take and viability through the comparison of the subcutaneous plane and the local fat pad in athymic rats. Methods: Lipoaspirate from a consenting patient was grafted into 2 locations in the subcutaneous plane and into the 2 inguinal fat pads in each of 4 athymic rats. Specimens were then collected after 47 days, and immunohistochemistry was utilized to determine angiogenesis in the fat grafts as a measure of fat graft take. Data were analyzed using the Student’s t test and analysis of variance followed by multiple comparisons. Results: There was no statistically significant difference (P = 0.2913) between the inguinal fat pad and the subcutaneous plane when measuring neovascularization. Analysis of variance comparing the graft locations also indicated no statistically significant difference when comparing each of the rats. Conclusions: Investigation into fat graft injection location indicates that there is no statistically significant difference in angiogenesis signals between the subcutaneous plane and the local fat pad in the athymic rat model. Further research should aim to continue to close the gap between clinical practice and basic scientific understanding of fat grafting.
Collapse
|
66
|
Chen R, Zhang F, Song L, Shu Y, Lin Y, Dong L, Nie X, Zhang D, Chen P, Han M. Transcriptome profiling reveals that the SM22α-regulated molecular pathways contribute to vascular pathology. J Mol Cell Cardiol 2014; 72:263-72. [DOI: 10.1016/j.yjmcc.2014.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/20/2014] [Accepted: 04/04/2014] [Indexed: 01/11/2023]
|
67
|
Tang Y, Gan X, Cheheltani R, Curran E, Lamberti G, Krynska B, Kiani MF, Wang B. Targeted delivery of vascular endothelial growth factor improves stem cell therapy in a rat myocardial infarction model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1711-8. [PMID: 24941463 DOI: 10.1016/j.nano.2014.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 05/16/2014] [Accepted: 06/01/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED Rebuilding of infarcted myocardium by mesenchymal stem cells (MSCs) has not been successful because of poor cell survival due in part to insufficient blood supply after myocardial infarction (MI). We hypothesize that targeted delivery of vascular endothelial growth factor (VEGF) to MI can help regenerate vasculature in support of MSC therapy in a rat model of MI. VEGF-encapsulated immunoliposomes targeting overexpressed P-selectin in MI tissue were infused by tail vein immediately after MI. One week later, MSCs were injected intramyocardially. The cardiac function loss was moderated slightly by targeted delivery of VEGF or MSC treatment. Targeted VEGF+MSC combination treatment showed highest attenuation in cardiac function loss. The combination treatment also increased blood vessel density (80%) and decreased collagen content in post-MI tissue (33%). Engraftment of MSCs in the combination treatment group was significantly increased and the engrafted cells contributed to the restoration of blood vessels. FROM THE CLINICAL EDITOR VEGF immunoliposomes targeting myocardial infarction tissue resulted in significantly higher attenuation of cardiac function loss when used in combination with mesenchymal stem cells. MSCs were previously found to have poor ability to restore cardiac tissue, likely as a result of poor blood supply in the affected areas. This new method counterbalances that weakness by the known effects of VEGF, as demonstrated in a rat model.
Collapse
Affiliation(s)
- Yuan Tang
- Department of Biomedical Engineering, Widener University, Chester, PA USA; Department of Mechanical Engineering, Temple University, Philadelphia, PA USA
| | - Xiaoliang Gan
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China
| | - Rabe'e Cheheltani
- Department of Mechanical Engineering, Temple University, Philadelphia, PA USA
| | - Elizabeth Curran
- Department of Mechanical Engineering, Temple University, Philadelphia, PA USA
| | - Giuseppina Lamberti
- Department of Mechanical Engineering, Temple University, Philadelphia, PA USA
| | - Barbara Krynska
- Shriners Hospitals Pediatric Research Center and Department of Neurology, Temple University School of Medicine, Philadelphia, PA USA
| | - Mohammad F Kiani
- Department of Mechanical Engineering, Temple University, Philadelphia, PA USA
| | - Bin Wang
- Department of Biomedical Engineering, Widener University, Chester, PA USA; Department of Mechanical Engineering, Temple University, Philadelphia, PA USA.
| |
Collapse
|
68
|
Abstract
BACKGROUND Understanding the physiology of pregnancy enables effective management of pregnancy complications that could otherwise be life threatening for both mother and fetus. A functional uterus (i) retains the fetus in utero during pregnancy without initiating stretch-induced contractions and (ii) is able to dilate the cervix and contract the myometrium at term to deliver the fetus. The onset of labour is associated with successful cervical remodelling and contraction of myometrium, arising from concomitant activation of uterine immune and endocrine systems. A large body of evidence suggests that actions of local steroid hormones may drive changes occurring in the uterine microenvironment at term. Although there have been a number of studies considering the potential role(s) played by progesterone and estrogen at the time of parturition, the bio-availability and effects of androgens during pregnancy have received less scrutiny. The aim of this review is to highlight potential roles of androgens in the biology of pregnancy and parturition. METHODS A review of published literature was performed to address (i) androgen concentrations, including biosynthesis and clearance, in maternal and fetal compartments throughout gestation, (ii) associations of androgen concentrations with adverse pregnancy outcomes, (iii) the role of androgens in the physiology of cervical remodelling and finally (iv) the role of androgens in the physiology of myometrial function including any impact on contractility. RESULTS Some, but not all, androgens increase throughout gestation in maternal circulation. The effects of this increase are not fully understood; however, evidence suggests that increased androgens might regulate key processes during pregnancy and parturition. For example, androgens are believed to be critical for cervical remodelling at term, in particular cervical ripening, via regulation of cervical collagen fibril organization. Additionally, a number of studies highlight potential roles for androgens in myometrial relaxation via non-genomic, AR-independent pathways critical for the pregnancy reaching term. Understanding of the molecular events leading to myometrial relaxation is an important step towards development of novel targeted tocolytic drugs. CONCLUSIONS The increase in androgen levels throughout gestation is likely to be important for establishment and maintenance of pregnancy and initiation of parturition. Further investigation of the underlying mechanisms of androgen action on cervical remodelling and myometrial contractility is needed. The insights gained may facilitate the development of new therapeutic approaches to manage pregnancy complications such as preterm birth.
Collapse
Affiliation(s)
- Sofia Makieva
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom and Tommy's Centre for Maternal and Fetal Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Philippa T K Saunders
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom and
| | - Jane E Norman
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom and Tommy's Centre for Maternal and Fetal Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
69
|
Zhao Y, Zha D, Wang L, Qiao L, Lu L, Mei L, Chen C, Qiu J. Phenotypic characterization of GPR120-expressing cells in the interstitial tissue of pancreas. Tissue Cell 2013; 45:421-7. [DOI: 10.1016/j.tice.2013.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 07/18/2013] [Accepted: 07/18/2013] [Indexed: 01/27/2023]
|
70
|
Tajsic T, Morrell NW. Smooth muscle cell hypertrophy, proliferation, migration and apoptosis in pulmonary hypertension. Compr Physiol 2013; 1:295-317. [PMID: 23737174 DOI: 10.1002/cphy.c100026] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pulmonary hypertension is a multifactorial disease characterized by sustained elevation of pulmonary vascular resistance (PVR) and pulmonary arterial pressure (PAP). Central to the pathobiology of this disease is the process of vascular remodelling. This process involves structural and functional changes to the normal architecture of the walls of pulmonary arteries (PAs) that lead to increased muscularization of the muscular PAs, muscularization of the peripheral, previously nonmuscular, arteries of the respiratory acinus, formation of neointima, and formation of plexiform lesions. Underlying or contributing to the development of these lesions is hypertrophy, proliferation, migration, and resistance to apoptosis of medial cells and this article is concerned with the cellular and molecular mechanisms of these processes. In the first part of the article we focus on the concept of smooth muscle cell phenotype and the difficulties surrounding the identification and characterization of the cell/cells involved in the remodelling of the vessel media and we review the general mechanisms of cell hypertrophy, proliferation, migration and apoptosis. Then, in the larger part of the article, we review the factors identified thus far to be involved in PH intiation and/or progression and review and discuss their effects on pulmonary artery smooth muscle cells (PASMCs) the predominant cells in the tunica media of PAs.
Collapse
Affiliation(s)
- Tamara Tajsic
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | |
Collapse
|
71
|
Vascular smooth muscle cells in cerebral aneurysm pathogenesis. Transl Stroke Res 2013; 5:338-46. [PMID: 24323713 DOI: 10.1007/s12975-013-0290-1] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 09/25/2013] [Indexed: 10/26/2022]
Abstract
Vascular smooth muscle cells (SMC) maintain significant plasticity. Following environmental stimulation, SMC can alter their phenotype from one primarily concerned with contraction to a pro-inflammatory and matrix remodeling phenotype. This is a critical process behind peripheral vascular disease and atherosclerosis, a key element of cerebral aneurysm pathology. Evolving evidence demonstrates that SMCs and phenotypic modulation play a significant role in cerebral aneurysm formation and rupture. Pharmacological alteration of smooth muscle cell function and phenotypic modulation could provide a promising medical therapy to inhibit cerebral aneurysm progression. This study reviews vascular SMC function and its contribution to cerebral aneurysm pathophysiology.
Collapse
|
72
|
Turner EC, Huang CL, Govindarajan K, Caplice NM. Identification of a Klf4-dependent upstream repressor region mediating transcriptional regulation of the myocardin gene in human smooth muscle cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1191-201. [PMID: 24060351 DOI: 10.1016/j.bbagrm.2013.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/28/2013] [Accepted: 09/13/2013] [Indexed: 01/25/2023]
Abstract
Phenotypic switching of smooth muscle cells (SMCs) plays a central role in the development of vascular diseases such as atherosclerosis and restenosis. However, the factors regulating expression of the human myocardin (Myocd) gene, the master gene regulator of SMC differentiation, have yet to be identified. In this study, we sought to identify the critical factors regulating Myocd expression in human SMCs. Using deletion/genetic reporter analyses, an upstream repressor region (URR) was localised within the Myocd promoter, herein termed PrmM. Bioinformatic analysis revealed three evolutionary conserved Klf4 sites within the URR and disruption of those elements led to substantial increases in PrmM-directed gene expression. Furthermore, ectopic expression established that Klf4 significantly decreased Myocd mRNA levels and PrmM-directed gene expression while electrophoretic mobility shift assays and chromatin immunoprecipitation (ChIP) assays confirmed specific binding of endogenous Klf4, and not Klf5 or Klf2, to the URR of PrmM. Platelet-derived growth factor BB (PDGF-BB), a potent inhibitor of SMC differentiation, reduced Myocd mRNA levels and PrmM-directed gene expression in SMCs. A PDGF-BB-responsive region (PRR) was also identified within PrmM, overlapping with the previously identified URR, where either siRNA knockdown of Klf4 or the combined disruption of the Klf4 elements completely abolished PDGF-BB-mediated repression of PrmM-directed gene expression in SMCs. Moreover, ChIP analysis established that PDGF-BB-induced repression of Myocd gene expression is most likely regulated by enhanced binding of Klf4 and Klf5 to a lesser extent, to the PRR of PrmM. Taken together, these data provide critical insights into the transcriptional regulation of the Myocd gene in vascular SMCs, including during SMC differentiation.
Collapse
Affiliation(s)
- Elizebeth C Turner
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland.
| | | | | | | |
Collapse
|
73
|
Arnoldi R, Hiltbrunner A, Dugina V, Tille JC, Chaponnier C. Smooth muscle actin isoforms: A tug of war between contraction and compliance. Eur J Cell Biol 2013; 92:187-200. [DOI: 10.1016/j.ejcb.2013.06.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 06/26/2013] [Accepted: 06/27/2013] [Indexed: 11/28/2022] Open
|
74
|
Zhang G, Zhang W, Lou Y, Xi W, Cui J, Geng M, Zhu F, Chen YH, Liu S. TIPE2 deficiency accelerates neointima formation by downregulating smooth muscle cell differentiation. Cell Cycle 2013; 12:501-10. [PMID: 23324338 PMCID: PMC3587451 DOI: 10.4161/cc.23325] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Phenotypic switching of vascular smooth muscle cells (VSMCs) is known to play a key role in the development of atherosclerosis. However, the mechanisms that mediate VSMC phenotypic switching are unclear. We report here that TIPE2, the tumor necrosis factor (TNF) α-induced protein 8-like 2 (TNFAIP8L2), plays an atheroprotective role by regulating phenotypic switching of VSMCs in response to oxidized low-density lipoprotein (ox-LDL) stimuli. TIPE2-deficient VSMCs treated with ox-LDL expressed lower levels of contractile proteins such as SMαA, SM-MHC and calponin, whereas the proliferation, migration and the synthetic capacity for growth factors and cytokines were increased remarkably. Furthermore, TIPE2 inhibited VSMCs proliferation by preventing G 1/S phase transition. Interestingly, these effects of TIPE2 on VSMCs were dependent on P38 and ERK1/2 kinase signals. As a result, neointima formation was accelerated in the carotid arteries of TIPE2-deficient mice. These results indicate that TIPE2 is a potential inhibitor of atherosclerosis.
Collapse
Affiliation(s)
- Guizhong Zhang
- Institute of Immunology; Shandong University School of Medicine; Ji’nan, China
| | - Wenqian Zhang
- Institute of Immunology; Shandong University School of Medicine; Ji’nan, China
| | - Yunwei Lou
- Institute of Immunology; Shandong University School of Medicine; Ji’nan, China
| | - Wenjin Xi
- Department of Immunology; Fourth Military Medical University; Xi’an, China
| | - Jian Cui
- Institute of Immunology; Shandong University School of Medicine; Ji’nan, China
| | - Minghong Geng
- Institute of Immunology; Shandong University School of Medicine; Ji’nan, China
| | - Faliang Zhu
- Institute of Immunology; Shandong University School of Medicine; Ji’nan, China
| | - Youhai H. Chen
- Department of Pathology and Laboratory Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Suxia Liu
- Institute of Immunology; Shandong University School of Medicine; Ji’nan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research; Chinese Ministry of Education and Chinese Ministry of Health; Shandong University Qilu Hospital; Jinan, China
| |
Collapse
|
75
|
Boyd NL, Nunes SS, Krishnan L, Jokinen JD, Ramakrishnan VM, Bugg AR, Hoying JB. Dissecting the role of human embryonic stem cell-derived mesenchymal cells in human umbilical vein endothelial cell network stabilization in three-dimensional environments. Tissue Eng Part A 2013; 19:211-223. [PMID: 22971005 PMCID: PMC3530951 DOI: 10.1089/ten.tea.2011.0408] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 07/30/2012] [Indexed: 10/27/2022] Open
Abstract
The microvasculature is principally composed of two cell types: endothelium and mural support cells. Multiple sources are available for human endothelial cells (ECs) but sources for human microvascular mural cells (MCs) are limited. We derived multipotent mesenchymal progenitor cells from human embryonic stem cells (hES-MC) that can function as an MC and stabilize human EC networks in three-dimensional (3D) collagen-fibronectin culture by paracrine mechanisms. Here, we have investigated the basis for hES-MC-mediated stabilization and identified the pleiotropic growth factor hepatocyte growth factor/scatter factor (HGF/SF) as a putative hES-MC-derived regulator of EC network stabilization in 3D in vitro culture. Pharmacological inhibition of the HGF receptor (Met) (1 μm SU11274) inhibits EC network formation in the presence of hES-MC. hES-MC produce and release HGF while human umbilical vein endothelial cells (HUVEC) do not. When HUVEC are cultured alone the networks collapse, but in the presence of recombinant human HGF or conditioned media from human HGF-transduced cells significantly more networks persist. In addition, HUVEC transduced to constitutively express human HGF also form stable networks by autocrine mechanisms. By enzyme-linked immunosorbent assay, the coculture media were enriched in both angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2), but at significantly different levels (Ang1=159±15 pg/mL vs. Ang2=30,867±2685 pg/mL) contributed by hES-MC and HUVEC, respectively. Although the coculture cells formed stabile network architectures, their morphology suggests the assembly of an immature plexus. When HUVEC and hES-MC were implanted subcutaneously in immune compromised Rag1 mice, hES-MC increased their contact with HUVEC along the axis of the vessel. This data suggests that HUVEC and hES-MC form an immature plexus mediated in part by HGF and angiopoietins that is capable of maturation under the correct environmental conditions (e.g., in vivo). Therefore, hES-MC can function as microvascular MCs and may be a useful cell source for testing EC-MC interactions.
Collapse
Affiliation(s)
- Nolan L Boyd
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA.
| | | | | | | | | | | | | |
Collapse
|
76
|
Yamin R, Morgan KG. Deciphering actin cytoskeletal function in the contractile vascular smooth muscle cell. J Physiol 2012; 590:4145-54. [PMID: 22687615 DOI: 10.1113/jphysiol.2012.232306] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review focuses on the vascular smooth muscle cells present in the medial layer of the blood vessels wall in the fully differentiated state (dVSMCs). The dVSMC contractile phenotype enables these cells to respond in a highly regulated manner to changes in extracellular stimuli. Through modulation of vascular contractile force and vascular compliance dVSMCs regulate blood pressure and blood flow. The cellular and molecular mechanisms by which vascular smooth muscle contractile functions are regulated are not completely elucidated. Recent studies have documented a critical role for actin polymerization and cytoskeletal dynamics in the regulation of contractile function. Here we will review the current understanding of actin cytoskeletal dynamics and focal adhesion function in dVSMCs in order to better understand actin cytoskeleton connections to the extracellular matrix and the effects of cytoskeletal remodelling on vascular contractility and vascular stiffness in health and disease.
Collapse
Affiliation(s)
- Rina Yamin
- Health Sciences Department, Boston University, 635 Commonwealth Ave, Boston, MA 02215, USA
| | | |
Collapse
|
77
|
Abstract
Smooth muscle cells (SMCs) possess remarkable phenotypic plasticity that allows rapid adaptation to fluctuating environmental cues, including during development and progression of vascular diseases such as atherosclerosis. Although much is known regarding factors and mechanisms that control SMC phenotypic plasticity in cultured cells, our knowledge of the mechanisms controlling SMC phenotypic switching in vivo is far from complete. Indeed, the lack of definitive SMC lineage-tracing studies in the context of atherosclerosis, and difficulties in identifying phenotypically modulated SMCs within lesions that have down-regulated typical SMC marker genes, and/or activated expression of markers of alternative cell types including macrophages, raise major questions regarding the contributions of SMCs at all stages of atherogenesis. The goal of this review is to rigorously evaluate the current state of our knowledge regarding possible phenotypes exhibited by SMCs within atherosclerotic lesions and the factors and mechanisms that may control these phenotypic transitions.
Collapse
Affiliation(s)
- Delphine Gomez
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, 415 Lane Road, PO Box 801394, Room 1322 Medical Research Building 5, Charlottesville, VA 22908, USA
| | | |
Collapse
|
78
|
Mitochondrial involvement in carbachol-induced intracellular Ca2+ mobilization and contraction in rat gastric smooth muscle. Life Sci 2011; 89:757-64. [DOI: 10.1016/j.lfs.2011.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 06/27/2011] [Accepted: 07/27/2011] [Indexed: 11/22/2022]
|
79
|
Alexander MR, Owens GK. Epigenetic control of smooth muscle cell differentiation and phenotypic switching in vascular development and disease. Annu Rev Physiol 2011; 74:13-40. [PMID: 22017177 DOI: 10.1146/annurev-physiol-012110-142315] [Citation(s) in RCA: 575] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The vascular smooth muscle cell (SMC) in adult animals is a highly specialized cell whose principal function is contraction. However, this cell displays remarkable plasticity and can undergo profound changes in phenotype during repair of vascular injury, during remodeling in response to altered blood flow, or in various disease states. There has been extensive progress in recent years in our understanding of the complex mechanisms that control SMC differentiation and phenotypic plasticity, including the demonstration that epigenetic mechanisms play a critical role. In addition, recent evidence indicates that SMC phenotypic switching in adult animals involves the reactivation of embryonic stem cell pluripotency genes and that mesenchymal stem cells may be derived from SMC and/or pericytes. This review summarizes the current state of our knowledge in this field and identifies some of the key unresolved challenges and questions that we feel require further study.
Collapse
Affiliation(s)
- Matthew R Alexander
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA.
| | | |
Collapse
|
80
|
Correlation between antizyme 1 and differentiation of vascular smooth muscle cells cultured in honeycomb-like type-I collagen matrix. Amino Acids 2011; 42:565-75. [DOI: 10.1007/s00726-011-1034-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 03/26/2011] [Indexed: 10/24/2022]
|
81
|
Gantus MAV, Alves LM, Stipursky J, Souza ECL, Teodoro AJ, Alves TR, Carvalho DP, Martinez AMB, Gomes FCA, Nasciutti LE. Estradiol modulates TGF-β1 expression and its signaling pathway in thyroid stromal cells. Mol Cell Endocrinol 2011; 337:71-9. [PMID: 21315800 DOI: 10.1016/j.mce.2011.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Revised: 01/31/2011] [Accepted: 02/01/2011] [Indexed: 01/14/2023]
Abstract
The higher prevalence of thyroid disease in women suggests that estrogen (E2) might be involved in the pathophysiology of thyroid dysfunction. To approach the question of the effect of stromal cells in the modulation of thyroid epithelial cells activity, we established and characterized a homogeneous stromal cell population (TS7 cells) of rat thyroid gland. These fibroblastic cells synthesize the cytoskeleton proteins α-smooth muscle actin and vimentin, produce basement membrane components and express the cytokine transforming growth factor beta 1 (TGF-β1). Here, we hypothesized that the effects of E2 on follicular thyroid cells are mediated by TGF-β1 synthesis and secretion by stromal cells (paracrine action). Thus we investigated the effect of E2 on TGF-β1 synthesis and its signaling pathway in TS7 cells. In addition, we analyzed the role of TGF-β1 signaling pathway as mediator of TS7-PC CL3 thyroid epithelial cells interactions. We report that TS7 stromal cells expressed α and β estrogen receptors (ERα and ERβ). Further, both isoforms of TGF-β1 receptors, TGFRI and TGFRII, were also identified in TS7 cells, suggesting that these cells might be a target for this cytokine in vitro. Treatment of TS7 cells with E2 induced both synthesis and secretion of TGF-β1. This event was followed by phosphorylation of the transcription factor Smad2, a hallmark of TGF-β1 pathway activation. Co-culture of PC CL3 cells onto TS7 cells monolayers yielded round aggregates of PC CL3 cells surrounded by TS7 cells. TS7 cells induced a decrease in iodide uptake by PC CL3 cells, probably by a mechanism involving TGF-β1. Moreover, E2 affected synthesis and organization of the extracellular matrix (ECM) components, tenascin C and chondroitin sulfate, in these co-culture cells. Our results point to the TGF-β1/Smad-2 signaling pathway as a putative target of estrogen actions on thyroid stromal cells and contribute to understanding the interplay between stromal and follicular cells in thyroid physiology.
Collapse
Affiliation(s)
- M A V Gantus
- Laboratory of Cellular Interactions, Program of Cellular Biology and Development, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
[Histochemical and immunohistochemical analysis of ruptured atherosclerotic abdominal aortic aneurysm wall]. VOJNOSANIT PREGL 2011; 67:959-64. [PMID: 21425554 DOI: 10.2298/vsp1012959t] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIM The main complication of the atherosclerotic abdominal aortic aneurysm (AAA) is her rupture that begins with lesion in intima and rupture. The purpose of this work was to determine immunocytochemical and morphofunctional characteristics of the cells in aortic wall in ruptured atherosclerotic abdominal aortic aneurysm. METHOD During the course of this study, 20 samples of atherosclerotic AAA were analyzed, all of them obtained during authopsy. The samples were fixed in 4% formalin and embedded in paraffin. Sections of 5 microm thickness were stained histochemically (of Heidenhain azan stain and Periodic acid Schiff--PAS stain) and immunocytochemically using a DAKO LSAB+/HRP technique to identify alpha-smooth muscle actin (alpha-SMA), vimentin, myosin heavy chains (MHC), desmin, S-100 protein, CD45 and CD68 (DAKO specification). RESULTS The results of our study showed that ruptured atherosclerotic AAA is characterized by a complete absence of endothelial cells, the disruption of basal membrane and internal elastic lamina, as well as a presence of the remains of hypocellular complicated atherosclerotic lesion in intima. On the plaque margins, as well as in the media, smooth muscle cells (SMCs) are present, which express a alpha-SMA and vimentin (but without MHC or desmin expression), as well as leukocyte infiltration, and a large number of foam cells. Some of the foam cells show a CD68- immunoreactivity, while the others show vimentin- and S-100 protein-immunoreactivity. Media is thinned out with a disorganized elastic lamellas, while adventitia is characterized by inflammatory inflitrate (infection). CONCLUSION Rupture of aneurysm occurs from the primary intimal disruption, which spreads into thinned out media and adventitia. Rupture is caused by unstable atherom, hypocellularity, loss of contractile characteristics of smooth muscle cells in intima and media, neovascularization of the media, as well as by the activity of the macrophages in the lesion.
Collapse
|
83
|
Abstract
BACKGROUND/AIM Myomas of the uterus, the most common benign tumors, have been studied for decades from the aspects of different basic and clinical disciplines. Despite this fact, their pathogenesis is still poorly understood. The aim of this study was to determine immunocytochemical characteristics of smooth muscle cells and connective tissue components of submucosal myomas of the uterus. METHOD During the course of this study, 25 samples of submucosal myomas of the uterus were analyzed, all of them obtained during the surgery, after abdominal histerctomy by Aldridge. The samples were fixed in 4% formalin and embedded in paraffin. Sections of 5 microm thickness were stained immunocytochemically using the DAKO LSAB+/HRP technique to identify alpha-smooth muscle actin (alpha-SMA), vimentin, desmin, CD34, CD45, CD68 and PCNA (DAKO specification). RESULTS Our results suggest that submucosal myomas of the uterus are build-up of smooth muscle cells which are immunoreactive to alpha-SMA and desmin, but also to a certain number of smooth muscle cells which are immunoreactive to alpha-SMA and vimentin. Some of vimentin-immunoreactive cells also show an immunoreactivity of PCNA. In the build-up of connective stroma CD34-immunoreactive fibroblasts and neovascular formations are also present. By examining the distribution of CD45 antigen, at all the analyzed samples we observed a weak reaction. CONCLUSION Submucosal myomas of the uterus are made-up of smooth muscle cells of the highly differentiated contractile phenotype (alpha-SMA- and desmin-immunoreactivity), as well as smooth muscle cell of the synthetic phenotype which proliferate (alpha-SMA-, vimentin- and PCNA-immunoreactivity). In submucosal myoma of the uterus there is a significant presence of connective tissue as a result of synthetic activity of fibroblasts, which clearly differ in their immunocytochemical characteristics from smooth muscle cells of the synthetic phenotype.
Collapse
|
84
|
DeFalco T, Takahashi S, Capel B. Two distinct origins for Leydig cell progenitors in the fetal testis. Dev Biol 2011; 352:14-26. [PMID: 21255566 PMCID: PMC3055913 DOI: 10.1016/j.ydbio.2011.01.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 12/17/2010] [Accepted: 01/07/2011] [Indexed: 12/27/2022]
Abstract
During the differentiation of the mammalian embryonic testis, two compartments are defined: the testis cords and the interstitium. The testis cords give rise to the adult seminiferous tubules, whereas steroidogenic Leydig cells and other less well characterized cell types differentiate in the interstitium (the space between testis cords). Although the process of testis cord formation is essential for male development, it is not entirely understood. It has been viewed as a Sertoli-cell driven process, but growing evidence suggests that interstitial cells play an essential role during testis formation. However, little is known about the origin of the interstitium or the molecular and cellular diversity within this early stromal compartment. To better understand the process of mammalian gonad differentiation, we have undertaken an analysis of developing interstitial/stromal cells in the early mouse testis and ovary. We have discovered molecular heterogeneity in the interstitium and have characterized new markers of distinct cell types in the gonad: MAFB, C-MAF, and VCAM1. Our results show that at least two distinct progenitor lineages give rise to the interstitial/stromal compartment of the gonad: the coelomic epithelium and specialized cells along the gonad-mesonephros border. We demonstrate that both these populations give rise to interstitial precursors that can differentiate into fetal Leydig cells. Our analysis also reveals that perivascular cells migrate into the gonad from the mesonephric border along with endothelial cells and that these vessel-associated cells likely represent an interstitial precursor lineage. This study highlights the cellular diversity of the interstitial cell population and suggests that complex cell-cell interactions among cells in the interstitium are involved in testis morphogenesis.
Collapse
Affiliation(s)
- Tony DeFalco
- The Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Doctoral Program in Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575 Japan
| | - Blanche Capel
- The Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
85
|
Kriz W, Kaissling B, Le Hir M. Epithelial-mesenchymal transition (EMT) in kidney fibrosis: fact or fantasy? J Clin Invest 2011; 121:468-74. [PMID: 21370523 DOI: 10.1172/jci44595] [Citation(s) in RCA: 370] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) has become widely accepted as a mechanism by which injured renal tubular cells transform into mesenchymal cells that contribute to the development of fibrosis in chronic renal failure. However, an increasing number of studies raise doubts about the existence of this process in vivo. Herein, we review and summarize both sides of this debate, but it is our view that unequivocal evidence supporting EMT as an in vivo process in kidney fibrosis is lacking.
Collapse
Affiliation(s)
- Wilhelm Kriz
- Department of Anatomy and Developmental Biology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| | | | | |
Collapse
|
86
|
Sugihara M, Morita H, Matsuda M, Umebayashi H, Kajioka S, Ito S, Nishida M, Inoue R, Futatsuki T, Yamazaki J, Mori Y, Inoue R, Ito Y, Abe K, Hirata M. Dual signaling pathways of arterial constriction by extracellular uridine 5'-triphosphate in the rat. J Pharmacol Sci 2011; 115:293-308. [PMID: 21350312 DOI: 10.1254/jphs.10281fp] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
We investigated actions of uridine 5'-triphosphate (UTP) in rat aorta, cerebral and mesenteric arteries, and their single myocytes. UTP (≥10 µM) elicited an inward-rectifying current strongly reminiscent of activation of P2X(1) receptor, and a similar current was also induced by α,β-methylene adenosine 5'-triphosphate (ATP) (≥100 nM). UTP desensitized α,β-methylene ATP-evoked current, and vice versa. The UTP-activated current was insensitive to G-protein modulators, TRPC3 inhibitors, or TRPC3 antibody, but was sensitive to P2-receptor inhibitors or P2X(1)-receptor antibody. Both UTP (1 mM) and α,β-methylene ATP (10 µM) elicited similar conductance single channel activities. UTP (≥10 µM) provoked a dose-dependent contraction of de-endothelialized aortic ring preparation consisting of phasic and tonic components. Removal of extracellular Ca(2+) or bath-applied 2',3'-O-(2,4,6-trinitrophenyl)-ATP (TNP-ATP) (30 µM) or nifedipine (10 µM) completely inhibited the phasic contraction while only partially reducing the tonic one. The tonic contraction was almost completely abolished by additional application of thapsigargin (2 µM). Similar biphasic rises in [Ca(2+)](i) were also evoked by UTP in rat aortic myocytes. In contrast to the low expression of TRPC3, significant expression of P2X(1) receptor was detected in all arteries by RT-PCR and immunoblotting, and its localization was limited to plasma membrane of myocytes as indicated by immunohistochemistry. These results suggest that UTP dually activates P2X(1)-like and P2Y receptors, but not TRPC3.
Collapse
Affiliation(s)
- Megumi Sugihara
- Special Patient Oral Care Unit, Kyushu University Hospital, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Majesky MW, Dong XR, Regan JN, Hoglund VJ. Vascular smooth muscle progenitor cells: building and repairing blood vessels. Circ Res 2011; 108:365-77. [PMID: 21293008 PMCID: PMC3382110 DOI: 10.1161/circresaha.110.223800] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 12/27/2010] [Indexed: 01/17/2023]
Abstract
Molecular pathways that control the specification, migration, and number of available smooth muscle progenitor cells play key roles in determining blood vessel size and structure, capacity for tissue repair, and progression of age-related disorders. Defects in these pathways produce malformations of developing blood vessels, depletion of smooth muscle progenitor cell pools for vessel wall maintenance and repair, and aberrant activation of alternative differentiation pathways in vascular disease. A better understanding of the molecular mechanisms that uniquely specify and maintain vascular smooth muscle cell precursors is essential if we are to use advances in stem and progenitor cell biology and somatic cell reprogramming for applications directed to the vessel wall.
Collapse
Affiliation(s)
- Mark W Majesky
- Seattle Children's Research Institute, University of Washington, 1900 Ninth Ave, M/S C9S-5, Seattle, WA 98101, USA.
| | | | | | | |
Collapse
|
88
|
Milewicz DM, Østergaard JR, Ala-Kokko LM, Khan N, Grange DK, Mendoza-Londono R, Bradley TJ, Olney AH, Adès L, Maher JF, Guo D, Buja LM, Kim D, Hyland JC, Regalado ES. De novo ACTA2 mutation causes a novel syndrome of multisystemic smooth muscle dysfunction. Am J Med Genet A 2010; 152A:2437-43. [PMID: 20734336 PMCID: PMC3573757 DOI: 10.1002/ajmg.a.33657] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Smooth muscle cells (SMCs) contract to perform many physiological functions, including regulation of blood flow and pressure in arteries, contraction of the pupils, peristalsis of the gut, and voiding of the bladder. SMC lineage in these organs is characterized by cellular expression of the SMC isoform of α-actin, encoded by the ACTA2 gene. We report here on a unique and de novo mutation in ACTA2, R179H, that causes a syndrome characterized by dysfunction of SMCs throughout the body, leading to aortic and cerebrovascular disease, fixed dilated pupils, hypotonic bladder, malrotation, and hypoperistalsis of the gut and pulmonary hypertension.
Collapse
Affiliation(s)
- Dianna M Milewicz
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Abstract
IMPORTANCE OF THE FIELD Mesenchymal stem cells are multipotent adult stem cell populations that have broad differentiation plasticity and immunosuppressive potential that render them of great importance in cell-based therapies. They are identified by in vitro characteristics based on their differentiation potential for clinical approaches while their biological properties and in vivo identities are often less understood. AREAS COVERED IN THIS REVIEW Recent research carried out in the last decade on mesenchymal stem cell biology suggests that mesenchymal stem cells from various tissues reside in a perivascular location and these can be identified as pericytes that function as mural cells in microvessels. WHAT THE READER WILL GAIN This review covers recent progress on understanding the link between pericytes and mesenchymal stem cells discussing specific points such as response to injury and tissue-specific functions. TAKE HOME MESSAGE Despite a long and controversial history, there is a growing acceptance that perivascular cells are connected with mesenchymal stem cells, all that is really lacking is genetic evidence to show differentiation of pericytes into different cells types.
Collapse
Affiliation(s)
- Jifan Feng
- Department of Craniofacial Development and MRC Centre for Transplantation, NIHR comprehensive Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and Kings College, London, UK
| | | | | |
Collapse
|
90
|
Qiu H, Zhu Y, Sun Z, Trzeciakowski JP, Gansner M, Depre C, Resuello RR, Natividad FF, Hunter WC, Genin GM, Elson EL, Vatner DE, Meininger GA, Vatner SF. Short communication: vascular smooth muscle cell stiffness as a mechanism for increased aortic stiffness with aging. Circ Res 2010; 107:615-9. [PMID: 20634486 PMCID: PMC2936100 DOI: 10.1161/circresaha.110.221846] [Citation(s) in RCA: 256] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 07/01/2010] [Accepted: 07/02/2010] [Indexed: 11/16/2022]
Abstract
RATIONALE Increased aortic stiffness, an important feature of many vascular diseases, eg, aging, hypertension, atherosclerosis, and aortic aneurysms, is assumed because of changes in extracellular matrix (ECM). OBJECTIVE We tested the hypothesis that the mechanisms also involve intrinsic stiffening of vascular smooth muscle cells (VSMCs). METHODS AND RESULTS Stiffness was measured in vitro both by atomic force microscopy (AFM) and in a reconstituted tissue model, using VSMCs from aorta of young versus old male monkeys (Macaca fascicularis) (n=7/group), where aortic stiffness increases by 200% in vivo. The apparent elastic modulus was increased (P<0.05) in old (41.7+/-0.5 kPa) versus young (12.8+/-0.3 kPa) VSMCs but not after disassembly of the actin cytoskeleton with cytochalasin D. Stiffness of the VSMCs in the reconstituted tissue model was also higher (P<0.05) in old (23.3+/-3.0 kPa) than in young (13.7+/-2.4 kPa). CONCLUSIONS These data support the novel concept, not appreciated previously, that increased vascular stiffness with aging is attributable not only to changes in ECM but also to intrinsic changes in VSMCs.
Collapse
Affiliation(s)
- Hongyu Qiu
- Dept Cell Biology, UMDNJ-New Jersey Medical School, Newark, NJ
| | | | - Zhe Sun
- Dalton Cardiovascular Res Ctr and Dept Pharmacology &.Physiology, University of Missouri, Columbia, MO
| | | | | | | | | | | | - William C. Hunter
- Dept Cell Biology, UMDNJ-New Jersey Medical School, Newark, NJ
- Dept Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ
| | - Guy M. Genin
- Depts Mechanical and Aerospace Engineering and Biochemistry, Washington University St. Louis, MO
| | - Elliot L. Elson
- Depts Mechanical and Aerospace Engineering and Biochemistry, Washington University St. Louis, MO
| | | | - Gerald A. Meininger
- Dalton Cardiovascular Res Ctr and Dept Pharmacology &.Physiology, University of Missouri, Columbia, MO
| | | |
Collapse
|
91
|
Carrillo-Sepúlveda MA, Barreto-Chaves MLM. Phenotypic modulation of cultured vascular smooth muscle cells: a functional analysis focusing on MLC and ERK1/2 phosphorylation. Mol Cell Biochem 2010; 341:279-89. [DOI: 10.1007/s11010-010-0459-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 03/25/2010] [Indexed: 12/31/2022]
|
92
|
Ritchie AC, Wijaya S, Ong WF, Zhong SP, Chian KS. Dependence of alignment direction on magnitude of strain in esophageal smooth muscle cells. Biotechnol Bioeng 2009; 102:1703-11. [PMID: 19170241 DOI: 10.1002/bit.22190] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The response of cells in vitro to mechanical forces has been the subject of much research using devices to exert controlled mechanical stimulation on cultured cells or isolated tissue. In this study, esophageal smooth muscle cells were seeded on flexible polyurethane membranes to form a confluent cell layer. The cells were then subjected to uniform cyclic stretch of varying magnitudes at a frequency of approximately five cycles per minute in a custom made mechatronic bioreactor, providing similar strains experienced in the in vivo mechanical environment of the esophagus. The results show that the orientation response is dependent on the magnitude of cyclic stretch applied. Smooth muscle cells showed parallel alignment to the force direction at low cyclic strains (2%) compared to the hill-valley morphology of static controls. At higher strains (5% and 10% magnitude), the cells exhibited a consistent alignment perpendicular to the strain. To our knowledge, this is the first time that the alignment direction's dependence on strain magnitude has been demonstrated. MTS analysis indicated that cell metabolism was reduced when mechanical strain was applied, and proliferation was inhibited by mechanical strain. Protein expression indicates a decrease in smooth muscle alpha-actin, indicative of changes in cell phenotype, an increase in vimentin, which is associated with increased cell motility, and an increase in desmin, indicating differentiation in stimulated cells.
Collapse
Affiliation(s)
- A C Ritchie
- Nanyang Technological University, Singapore, Singapore.
| | | | | | | | | |
Collapse
|
93
|
Santoro MM, Pesce G, Stainier DY. Characterization of vascular mural cells during zebrafish development. Mech Dev 2009; 126:638-49. [PMID: 19539756 PMCID: PMC2732398 DOI: 10.1016/j.mod.2009.06.1080] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 05/28/2009] [Accepted: 06/12/2009] [Indexed: 12/30/2022]
Abstract
Development and maturation of the nascent cardiovascular system requires the recruitment of mural cells (MCs) around the vascular tree in a process called vascular myogenesis. Understanding the origin and development of vascular MCs has been hampered by difficulties in observing these cells in vivo and performing defined genetic and experimental manipulations in available model organisms. Here, we investigate the origin of vascular MCs using molecular and genetic tools in zebrafish. We show that vascular MCs are present around the lateral dorsal aortae (LDA) and anterior mesenteric arteries (AMA) of developing animals, and that they also contribute to the outflow tract of the developing heart and ventral aorta (VA). Genetic data indicate that the vascular MCs of the LDA and AMA do not arise from blood or endothelial progenitors but from other derivatives of the lateral plate mesoderm. We further show that zebrafish vascular MCs share many of the morphological, molecular and functional characteristics of vascular smooth muscle cells and pericytes found in higher vertebrates. These data establish the zebrafish as a useful cellular and genetic model to study vascular myogenesis as well as tumor angiogenesis and other MC-associated diseases.
Collapse
Affiliation(s)
- Massimo M Santoro
- Molecular Biotechnology Center, University of Torino, Via Nizza, 52, 10126 Torino, Italy.
| | | | | |
Collapse
|
94
|
van Engelen E, Breeveld-Dwarkasing VNA, Everts ME, van der Weyden GC, Taverne MAM, Rutten VPMG. Smooth muscle cells of the bovine cervical stroma may have a secretory, rather than a contractile function during parturition. Reprod Domest Anim 2009; 44:303-11. [PMID: 19323797 DOI: 10.1111/j.1439-0531.2008.01070.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The bovine cervix contains a large amount of smooth muscle cells distributed over an outer muscular layer and within a stromal layer. The stromal layer exhibits no electromyographic (EMG) activity at parturition. This leads to the question whether the stromal smooth muscle cells of the bovine cervix are prepared to contract with parturition, or whether they have another function. To this end, cervical biopsies were repeatedly taken from 10 pregnant cows at day-185 and -275 of gestation, at spontaneous, uncomplicated calving and at 30 days after calving. The smooth muscle bundles of the stroma were immunohistochemically analysed (n = 5) with regard to their integrity and cellular density, and the degree of staining for connexin-43, smooth muscle actin alpha (SMA), desmin and vimentin. Additionally, the mRNA expression for connexin-43, SMA, desmin and vimentin was determined with RT-PCR (n = 5). The smooth muscle tissue was arranged in bundles, also at parturition. However, the cellular density of these bundles and the SMA mRNA expression were decreased at parturition. Additionally, the SMA staining and connexin-43 expression and staining remained constant during pregnancy and at parturition. This might indicate that stromal smooth muscle cells are not prepared to contract with parturition, in contrast to the myometrial smooth muscle cells. The smooth muscle cells, stained for SMA, also expressed vimentin, and the proportion of co-expression was increased at day-275 of pregnancy. This suggests that the stromal smooth muscle cells predominantly have a secretory function in cows.
Collapse
Affiliation(s)
- E van Engelen
- Department of Pathobiology, Division of Anatomy and Physiology, Utrecht University, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
95
|
Expression of nestin, desmin and vimentin in intact and regenerating muscle spindles of rat hind limb skeletal muscles. Histochem Cell Biol 2008; 131:197-206. [PMID: 18941770 DOI: 10.1007/s00418-008-0523-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2008] [Indexed: 10/21/2022]
Abstract
We describe the expression and distribution patterns of nestin, desmin and vimentin in intact and regenerating muscle spindles of the rat hind limb skeletal muscles. Regeneration was induced by intramuscular isotransplantation of extensor digitorum longus (EDL) or soleus muscles from 15-day-old rats into the EDL muscle of adult female inbred Lewis rats. The host muscles with grafts were excised after 7-, 16-, 21- and 29-day survival and immunohistochemically stained. Nestin expression in intact spindles in host muscles was restricted to Schwann cells of sensory and motor nerves. In transplanted muscles, however, nestin expression was also found in regenerating "spindle fibers", 7 and 16 days after grafting. From the 21st day onwards, the regenerated spindle fibers were devoid of nestin immunoreactivity. Desmin was detected in spindle fibers at all developmental stages in regenerating as well as in intact spindles. Vimentin was expressed in cells of the outer and inner capsules of all muscle spindles and in newly formed myoblasts and myotubes of regenerating spindles 7 days after grafting. Our results show that the expression pattern of these intermediate filaments in regenerating spindle fibers corresponds to that found in regenerating extrafusal fibers, which supports our earlier suggestion that they resemble small-diameter extrafusal fibers.
Collapse
|
96
|
Guo H, Makarova N, Cheng Y, Shuyu E, Ji RR, Zhang C, Farrar P, Tigyi G. The early- and late stages in phenotypic modulation of vascular smooth muscle cells: differential roles for lysophosphatidic acid. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1781:571-81. [PMID: 18602022 PMCID: PMC3446793 DOI: 10.1016/j.bbalip.2008.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/15/2008] [Revised: 06/02/2008] [Accepted: 06/04/2008] [Indexed: 12/16/2022]
Abstract
Lysophosphatidic acid (LPA) has been implicated as causative in phenotypic modulation (PM) of cultured vascular smooth muscle cells (VSMC) in their transition to the dedifferentiated phenotype. We evaluated the contribution of the three major LPA receptors, LPA1 and LPA2 GPCR and PPARgamma, on PM of VSMC. Expression of differentiated VSMC-specific marker genes, including smooth muscle alpha-actin, smooth muscle myosin heavy chain, calponin, SM-22alpha, and h-caldesmon, was measured by quantitative real-time PCR in VSMC cultures and aortic rings kept in serum-free chemically defined medium or serum- or LPA-containing medium using wild-type C57BL/6, LPA1, LPA2, and LPA1&2 receptor knockout mice. Within hours after cells were deprived of physiological cues, the expression of VSMC marker genes, regardless of genotype, rapidly decreased. This early PM was neither prevented by IGF-I, inhibitors of p38, ERK1/2, or PPARgamma nor significantly accelerated by LPA or serum. To elucidate the mechanism of PM in vivo, carotid artery ligation with/without replacement of blood with Krebs solution was used to evaluate contributions of blood flow and pressure. Early PM in the common carotid was induced by depressurization regardless of the presence/absence of blood, but eliminating blood flow while maintaining blood pressure or after sham surgery elicited no early PM. The present results indicate that LPA, serum, dissociation of VSMC, IGF-I, p38, ERK1/2, LPA1, and LPA2 are not causative factors of early PM of VSMC. Tensile stress generated by blood pressure may be the fundamental signal maintaining the fully differentiated phenotype of VSMC.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Apoptosis
- Blood Pressure/physiology
- Cell Differentiation/drug effects
- Cells, Cultured
- Insulin-Like Growth Factor I/antagonists & inhibitors
- Lysophospholipids/metabolism
- Male
- Mice
- Mitogen-Activated Protein Kinases/antagonists & inhibitors
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phenotype
- Time Factors
Collapse
Affiliation(s)
- Huazhang Guo
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Suite 426, Memphis, TN 38163, USA
| | - Natalia Makarova
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Suite 426, Memphis, TN 38163, USA
| | - Yunhui Cheng
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Suite 426, Memphis, TN 38163, USA
| | - E Shuyu
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Suite 426, Memphis, TN 38163, USA
| | - Rui-Rui Ji
- Department of Medicine, University of Tennessee Health Science Center, Memphis, USA
| | - Chunxiang Zhang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, USA
| | - Patricia Farrar
- Department of Comparative Medicine, University of Tennessee Health Science Center, Memphis, USA
| | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Suite 426, Memphis, TN 38163, USA
| |
Collapse
|
97
|
Abstract
The intermediate filament (IF) network is one of the three cytoskeletal systems in smooth muscle. The type III IF proteins vimentin and desmin are major constituents of the network in smooth muscle cells and tissues. Lack of vimentin or desmin impairs contractile ability of various smooth muscle preparations, implying their important role for smooth muscle force development. The IF framework has long been viewed as a fixed cytostructure that solely provides mechanical integrity for the cell. However, recent studies suggest that the IF cytoskeleton is dynamic in mammalian cells in response to various external stimulation. In this review, the structure and biological properties of IF proteins in smooth muscle are summarized. The role of IF proteins in the modulation of smooth muscle force development and redistribution/translocation of signaling partners (such as p130 Crk-associated substrate, CAS) is depicted. This review also summarizes our latest understanding on how the IF network may be regulated in smooth muscle.
Collapse
Affiliation(s)
- Dale D Tang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
98
|
Abstract
The hepatic stellate cell has surprised and engaged physiologists, pathologists, and hepatologists for over 130 years, yet clear evidence of its role in hepatic injury and fibrosis only emerged following the refinement of methods for its isolation and characterization. The paradigm in liver injury of activation of quiescent vitamin A-rich stellate cells into proliferative, contractile, and fibrogenic myofibroblasts has launched an era of astonishing progress in understanding the mechanistic basis of hepatic fibrosis progression and regression. But this simple paradigm has now yielded to a remarkably broad appreciation of the cell's functions not only in liver injury, but also in hepatic development, regeneration, xenobiotic responses, intermediary metabolism, and immunoregulation. Among the most exciting prospects is that stellate cells are essential for hepatic progenitor cell amplification and differentiation. Equally intriguing is the remarkable plasticity of stellate cells, not only in their variable intermediate filament phenotype, but also in their functions. Stellate cells can be viewed as the nexus in a complex sinusoidal milieu that requires tightly regulated autocrine and paracrine cross-talk, rapid responses to evolving extracellular matrix content, and exquisite responsiveness to the metabolic needs imposed by liver growth and repair. Moreover, roles vital to systemic homeostasis include their storage and mobilization of retinoids, their emerging capacity for antigen presentation and induction of tolerance, as well as their emerging relationship to bone marrow-derived cells. As interest in this cell type intensifies, more surprises and mysteries are sure to unfold that will ultimately benefit our understanding of liver physiology and the diagnosis and treatment of liver disease.
Collapse
Affiliation(s)
- Scott L Friedman
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York 10029-6574, USA.
| |
Collapse
|
99
|
Goldberg SR, McKinstry RP, Sykes V, Lanning DA. Rapid closure of midgestational excisional wounds in a fetal mouse model is associated with altered transforming growth factor-beta isoform and receptor expression. J Pediatr Surg 2007; 42:966-71; discussion 971-3. [PMID: 17560204 DOI: 10.1016/j.jpedsurg.2007.01.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND Many pediatric diseases are characterized by excessive tissue contraction. Because of a poor understanding of contraction, few therapies exist. We developed a murine fetal excisional wound model of contraction and theorize that wound closure is associated with changes in transforming growth factor-beta (TGF-beta) expression. METHODS Pregnant FVB mice underwent hysterotomy at midgestational (E15) or late-gestational (E18) ages. Three-millimeter excisional wounds were made in fetuses and harvested at 32 hours. Real-time polymerase chain reaction was performed for TGF-beta1, TGF-beta2, TGF-beta3, TbetaR-1, and TbetaR-2 in wounds and normal skin and normalized to glyceraldehyde-3-phosphate dehydrogenase. Data were analyzed by paired t test (P < .05). H&E staining of wounds was performed. RESULTS E15 wounds (80.5% +/- 4.4%) were smaller than E18 wounds (10.4% +/- 10.5%; P < .001) at 32 hours. E15 wounds expressed higher levels of TGF-beta1 compared with normal skin (P = .001). TbetaR-2 levels were elevated in E15 and E18 wounds compared with their respective normal skin (P = .02, P = .01) and in E18 normal skin compared with E15 normal skin (P = .002). CONCLUSION This study demonstrates that rapid midgestational wound closure in a murine model is associated with increased TGF-beta1 and TbetaR-2 expression. Elucidating the role of the TGF-beta pathways may lead to an improved understanding of wound contraction.
Collapse
MESH Headings
- Activin Receptors, Type I/biosynthesis
- Activin Receptors, Type I/genetics
- Animals
- Female
- Gene Expression Regulation
- Gestational Age
- Hysterotomy
- Mice
- Models, Animal
- Polymerase Chain Reaction
- Pregnancy
- Protein Isoforms/physiology
- Protein Serine-Threonine Kinases
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptor, Transforming Growth Factor-beta Type I
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/biosynthesis
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/physiology
- Skin/embryology
- Skin/injuries
- Skin/metabolism
- Transforming Growth Factor beta/physiology
- Transforming Growth Factor beta1/biosynthesis
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta2/biosynthesis
- Transforming Growth Factor beta2/genetics
- Transforming Growth Factor beta3/biosynthesis
- Transforming Growth Factor beta3/genetics
- Wound Healing/physiology
Collapse
Affiliation(s)
- Stephanie R Goldberg
- Division of Pediatric Surgery, Department of Surgery, Medical College of Virginia Hospitals, Virginia Commonwealth University Health System, P.O. Box 980015, Richmond, VA 23298-0015, USA
| | | | | | | |
Collapse
|
100
|
Zhou W, Dasgupta C, Negash S, Raj JU. Modulation of pulmonary vascular smooth muscle cell phenotype in hypoxia: role of cGMP-dependent protein kinase. Am J Physiol Lung Cell Mol Physiol 2007; 292:L1459-66. [PMID: 17322285 DOI: 10.1152/ajplung.00143.2006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic hypoxia triggers pulmonary vascular remodeling, which is associated with a modulation of the vascular smooth muscle cell (SMC) phenotype from a contractile, differentiated to a synthetic, dedifferentiated state. We previously reported that acute hypoxia represses cGMP-dependent protein kinase (PKG) expression in ovine fetal pulmonary venous SMCs (FPVSMCs). Therefore, we tested if altered expression of PKG could explain SMC phenotype modulation after exposure to hypoxia. Hypoxia-induced reduction in PKG protein expression strongly correlated with the repressed expression of SMC phenotype markers, myosin heavy chain (MHC), calponin, vimentin, alpha-smooth muscle actin (alphaSMA), and thrombospondin (TSP), indicating that hypoxic exposure of SMC induced phenotype modulation to dedifferentiated state, and PKG may be involved in SMC phenotype modulation. PKG-specific small interfering RNA (siRNA) transfection in FPVSMCs significantly attenuated calponin, vimentin, and MHC expression, with no effect on alphaSMA and TSP. Treatment with 30 microM Drosophila Antennapedia (DT-3), a membrane-permeable peptide inhibitor of PKG, attenuated the expression of TSP, MHC, alphaSMA, vimentin, and calponin. The results from PKG siRNA and DT-3 studies indicate that hypoxia-induced reduction in protein expression was also similarly impacted by PKG inhibition. Overexpression of PKG in FPVSMCs by transfection with a full-length PKG construct tagged with green fluorescent fusion protein (PKG-GFP) reversed the effect of hypoxia on the expression of SMC phenotype marker proteins. These results suggest that PKG could be one of the determinants for the expression of SMC phenotype marker proteins and may be involved in the maintenance of the differentiated phenotype in pulmonary vascular SMCs in hypoxia.
Collapse
Affiliation(s)
- Weilin Zhou
- Division of Neonatology, Harbor-UCLA Medical Center, Los Angeles Biomedical Research Institute at Harbor-UCLA, 1124 West Carson St., Torrance, CA 90502, USA.
| | | | | | | |
Collapse
|