51
|
Uygur A, Lee RT. Mechanisms of Cardiac Regeneration. Dev Cell 2016; 36:362-74. [PMID: 26906733 DOI: 10.1016/j.devcel.2016.01.018] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/13/2016] [Accepted: 01/25/2016] [Indexed: 02/07/2023]
Abstract
Adult humans fail to regenerate their hearts following injury, and this failure to regenerate myocardium is a leading cause of heart failure and death worldwide. Although all adult mammals appear to lack significant cardiac regeneration potential, some vertebrates can regenerate myocardium throughout life. In addition, new studies indicate that mammals have cardiac regeneration potential during development and very soon after birth. The mechanisms of heart regeneration among model organisms, including neonatal mice, appear remarkably similar. Orchestrated waves of inflammation, matrix deposition and remodeling, and cardiomyocyte proliferation are commonly seen in heart regeneration models. Understanding why adult mammals develop extensive scarring instead of regeneration is a crucial goal for regenerative biology.
Collapse
Affiliation(s)
- Aysu Uygur
- Department of Stem Cell and Regenerative Biology, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Cambridge, MA 02139, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Cambridge, MA 02139, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
52
|
Park SY, Piao Y, Jeong KJ, Dong J, de Groot JF. Periostin (POSTN) Regulates Tumor Resistance to Antiangiogenic Therapy in Glioma Models. Mol Cancer Ther 2016; 15:2187-97. [PMID: 27307601 DOI: 10.1158/1535-7163.mct-15-0427] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/06/2016] [Indexed: 01/12/2023]
Abstract
Periostin (POSTN) interacts with multiple integrins to coordinate a variety of cellular processes, including epithelial-to-mesenchymal transition (EMT) and cell migration. In our previous study, anti-VEGF-A therapy was associated with resistance and EMT. This study sought to determine the role of POSTN in the resistance of glioma stem cells (GSC) to antiangiogenic therapy. In mouse xenograft models of human glioma, POSTN expression was associated with acquired resistance to anti-VEGF-A therapy and had a synergistic effect with bevacizumab in prolonging survival and decreasing tumor volume. Resistance to anti-VEGF-A therapy regulated by POSTN was associated with increased expression of TGFβ1 and hypoxia-inducible factor-1α (HIF1α) in GSCs. At the molecular level, POSTN regulated invasion and expression of EMT (caveolin-1) and angiogenesis-related genes (HIF1α and VEGF-A) through activation of STAT3. Moreover, recombinant POSTN increased GSC invasion. Collectively, our findings suggest that POSTN plays an important role in glioma invasion and resistance to antiangiogenic therapy. Mol Cancer Ther; 15(9); 2187-97. ©2016 AACR.
Collapse
Affiliation(s)
- Soon Young Park
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuji Piao
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kang Jin Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianwen Dong
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
53
|
Walker JT, McLeod K, Kim S, Conway SJ, Hamilton DW. Periostin as a multifunctional modulator of the wound healing response. Cell Tissue Res 2016; 365:453-65. [PMID: 27234502 DOI: 10.1007/s00441-016-2426-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/04/2016] [Indexed: 12/23/2022]
Abstract
During tissue healing, the dynamic and temporal alterations required for effective repair occur in the structure and composition of the extracellular matrix (ECM). Matricellular proteins (MPs) are a group of diverse non-structural ECM components that bind cell surface receptors mediating interactions between the cell and its microenviroment, effectively regulating adhesion, migration, proliferation, signaling, and cell phenotype. Periostin (Postn), a pro-fibrogenic secreted glycoprotein, is defined as an MP based on its expression pattern and regulatory roles during development and healing and in disease processes. Postn consists of a typical signal sequence, an EMI domain responsible for binding to fibronectin, four tandem fasciclin-like domains that are responsible for integrin binding, and a C-terminal region in which multiple splice variants originate. This review focuses specifically on the role of Postn in wound healing and remodeling, an area of intense research during the last 10 years, particularly as related to skin healing and myocardium post-infarction. Postn interacts with cells through various integrin pairs and is an essential downstream effector of transforming growth factor-β superfamily signaling. Across various tissues, Postn is associated with the pro-fibrogenic process: specifically, the transition of fibroblasts to myofibroblasts, collagen fibrillogenesis, and ECM synthesis. Although the complexity of Postn as a modulator of cell behavior in tissue healing is only beginning to be elucidated, its expression is clearly a defining event in moving wound healing through the proliferative and remodeling phases.
Collapse
Affiliation(s)
- John T Walker
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1
| | - Karrington McLeod
- Graduate Program in Biomedical Engineering, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1
| | - Shawna Kim
- Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Douglas W Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1.
- Graduate Program in Biomedical Engineering, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1.
- Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1.
| |
Collapse
|
54
|
Kim JH, Kang MS, Eltohamy M, Kim TH, Kim HW. Dynamic Mechanical and Nanofibrous Topological Combinatory Cues Designed for Periodontal Ligament Engineering. PLoS One 2016; 11:e0149967. [PMID: 26989897 PMCID: PMC4798756 DOI: 10.1371/journal.pone.0149967] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/08/2016] [Indexed: 11/18/2022] Open
Abstract
Complete reconstruction of damaged periodontal pockets, particularly regeneration of periodontal ligament (PDL) has been a significant challenge in dentistry. Tissue engineering approach utilizing PDL stem cells and scaffolding matrices offers great opportunity to this, and applying physical and mechanical cues mimicking native tissue conditions are of special importance. Here we approach to regenerate periodontal tissues by engineering PDL cells supported on a nanofibrous scaffold under a mechanical-stressed condition. PDL stem cells isolated from rats were seeded on an electrospun polycaprolactone/gelatin directionally-oriented nanofiber membrane and dynamic mechanical stress was applied to the cell/nanofiber construct, providing nanotopological and mechanical combined cues. Cells recognized the nanofiber orientation, aligning in parallel, and the mechanical stress increased the cell alignment. Importantly, the cells cultured on the oriented nanofiber combined with the mechanical stress produced significantly stimulated PDL specific markers, including periostin and tenascin with simultaneous down-regulation of osteogenesis, demonstrating the roles of topological and mechanical cues in altering phenotypic change in PDL cells. Tissue compatibility of the tissue-engineered constructs was confirmed in rat subcutaneous sites. Furthermore, in vivo regeneration of PDL and alveolar bone tissues was examined under the rat premaxillary periodontal defect models. The cell/nanofiber constructs engineered under mechanical stress showed sound integration into tissue defects and the regenerated bone volume and area were significantly improved. This study provides an effective tissue engineering approach for periodontal regeneration—culturing PDL stem cells with combinatory cues of oriented nanotopology and dynamic mechanical stretch.
Collapse
Affiliation(s)
- Joong-Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Min Sil Kang
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Mohamed Eltohamy
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Tae-Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Republic of Korea
- * E-mail:
| |
Collapse
|
55
|
Wang Z, Calpe B, Zerdani J, Lee Y, Oh J, Bae H, Khademhosseini A, Kim K. High-throughput investigation of endothelial-to-mesenchymal transformation (EndMT) with combinatorial cellular microarrays. Biotechnol Bioeng 2015; 113:1403-12. [PMID: 26666585 DOI: 10.1002/bit.25905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 01/09/2023]
Abstract
In the developing heart, a specific subset of endocardium undergoes an endothelial-to-mesenchymal transformation (EndMT) thus forming nascent valve leaflets. Extracellular matrix (ECM) proteins and growth factors (GFs) play important roles in regulating EndMT but the combinatorial effect of GFs with ECM proteins is less well understood. Here we use microscale engineering techniques to create single, binary, and tertiary component microenvironments to investigate the combinatorial effects of ECM proteins and GFs on the attachment and transformation of adult ovine mitral valve endothelial cells to a mesenchymal phenotype. With the combinatorial microenvironment microarrays, we utilized 60 different combinations of ECM proteins (Fibronectin, Collagen I, II, IV, Laminin) and GFs (TGF-β1, bFGF, VEGF) and were able to identify new microenvironmental conditions capable of modulating EndMT in MVECs. Experimental results indicated that TGF-β1 significantly upregulated the EndMT while either bFGF or VEGF downregulated EndMT process markedly. Also, ECM proteins could influence both the attachment of MVECs and the response of MVECs to GFs. In terms of attachment, fibronectin is significantly better for the adhesion of MVECs among the five tested proteins. Overall collagen IV and fibronectin appeared to play important roles in promoting EndMT process. Great consistency between macroscale and microarrayed experiments and present studies demonstrates that high-throughput cellular microarrays are a promising approach to study the regulation of EndMT in valvular endothelium. Biotechnol. Bioeng. 2016;113: 1403-1412. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zongjie Wang
- School of Engineering, University of British Columbia, Kelowna, BC, V1V1V7, Canada
| | - Blaise Calpe
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland.,Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts.,Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts
| | - Jalil Zerdani
- Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts.,Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts.,Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Youngsang Lee
- Department of Mathematics and Statistics, University of British Columbia, Kelowna, BC, Canada
| | - Jonghyun Oh
- Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts.,Division of Mechanical Design Engineering, Chonbuk National University, Jeonjoo, Republic of Korea.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| | - Hojae Bae
- Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139.,Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, Republic of Korea
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts. .,Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts. .,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139. .,Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia.
| | - Keekyoung Kim
- School of Engineering, University of British Columbia, Kelowna, BC, V1V1V7, Canada. .,Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts. .,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139.
| |
Collapse
|
56
|
Gould RA, Yalcin HC, MacKay JL, Sauls K, Norris R, Kumar S, Butcher JT. Cyclic Mechanical Loading Is Essential for Rac1-Mediated Elongation and Remodeling of the Embryonic Mitral Valve. Curr Biol 2015; 26:27-37. [PMID: 26725196 DOI: 10.1016/j.cub.2015.11.033] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 10/02/2015] [Accepted: 11/16/2015] [Indexed: 10/22/2022]
Abstract
During valvulogenesis, globular endocardial cushions elongate and remodel into highly organized thin fibrous leaflets. Proper regulation of this dynamic process is essential to maintain unidirectional blood flow as the embryonic heart matures. In this study, we tested how mechanosensitive small GTPases, RhoA and Rac1, coordinate atrioventricular valve (AV) differentiation and morphogenesis. RhoA activity and its regulated GTPase-activating protein FilGAP are elevated during early cushion formation but decreased considerably during valve remodeling. In contrast, Rac1 activity was nearly absent in the early cushions but increased substantially as the valve matured. Using gain- and loss-of-function assays, we determined that the RhoA pathway was essential for the contractile myofibroblastic phenotype present in early cushion formation but was surprisingly insufficient to drive matrix compaction during valve maturation. The Rac1 pathway was necessary to induce matrix compaction in vitro through increased cell adhesion, elongation, and stress fiber alignment. Facilitating this process, we found that acute cyclic stretch was a potent activator of RhoA and subsequently downregulated Rac1 activity via FilGAP. On the other hand, chronic cyclic stretch reduced active RhoA and downstream FilGAP, which enabled Rac1 activation. Finally, we used partial atrial ligation experiments to confirm in vivo that altered cyclic mechanical loading augmented or restricted cushion elongation and thinning, directly through potentiation of active Rac1 and active RhoA, respectively. Together, these results demonstrate that cyclic mechanical signaling coordinates the RhoA to Rac1 signaling transition essential for proper embryonic mitral valve remodeling.
Collapse
Affiliation(s)
- Russell A Gould
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Huseyin C Yalcin
- Qatar Cardiovascular Research Center (QCRC), Sidra Medical and Research Center, Doha, Qatar; Department of Mechanical Engineering, Dogus University, Istanbul 34722, Turkey
| | - Joanna L MacKay
- Department of Bioengineering, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kimberly Sauls
- Department of Regenerative Medicine and Cell Biology, School of Medicine, Cardiovascular Developmental Biology Center, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Russell Norris
- Department of Regenerative Medicine and Cell Biology, School of Medicine, Cardiovascular Developmental Biology Center, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California Berkeley, Berkeley, CA 94720, USA
| | - Jonathan T Butcher
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
57
|
Bowen CJ, Zhou J, Sung DC, Butcher JT. Cadherin-11 coordinates cellular migration and extracellular matrix remodeling during aortic valve maturation. Dev Biol 2015; 407:145-57. [PMID: 26188246 DOI: 10.1016/j.ydbio.2015.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 06/15/2015] [Accepted: 07/13/2015] [Indexed: 12/30/2022]
Abstract
Proper remodeling of the endocardial cushions into thin fibrous valves is essential for gestational progression and long-term function. This process involves dynamic interactions between resident cells and their local environment, much of which is not understood. In this study, we show that deficiency of the cell-cell adhesion protein cadherin-11 (Cad-11) results in significant embryonic and perinatal lethality primarily due to valve related cardiac dysfunction. While endocardial to mesenchymal transformation is not abrogated, mesenchymal cells do not homogeneously cellularize the cushions. These cushions remain thickened with disorganized ECM, resulting in pronounced aortic valve insufficiency. Mice that survive to adulthood maintain thickened and stenotic semilunar valves, but interestingly do not develop calcification. Cad-11 (-/-) aortic valve leaflets contained reduced Sox9 activity, β1 integrin expression, and RhoA-GTP activity, suggesting that remodeling defects are due to improper migration and/or cellular contraction. Cad-11 deletion or siRNA knockdown reduced migration, eliminated collective migration, and impaired 3D matrix compaction by aortic valve interstitial cells (VIC). Cad-11 depleted cells in culture contained few filopodia, stress fibers, or contact inhibited locomotion. Transfection of Cad-11 depleted cells with constitutively active RhoA restored cell phenotypes. Together, these results identify cadherin-11 mediated adhesive signaling for proper remodeling of the embryonic semilunar valves.
Collapse
Affiliation(s)
- Caitlin J Bowen
- Department of Biomedical Engineering, Cornell University, United States
| | - Jingjing Zhou
- Department of Biomedical Engineering, Cornell University, United States
| | - Derek C Sung
- Department of Biomedical Engineering, Cornell University, United States
| | - Jonathan T Butcher
- Department of Biomedical Engineering, Cornell University, United States.
| |
Collapse
|
58
|
Heidari P, Esfahani SA, Turker NS, Wong G, Wang TC, Rustgi AK, Mahmood U. Imaging of Secreted Extracellular Periostin, an Important Marker of Invasion in the Tumor Microenvironment in Esophageal Cancer. J Nucl Med 2015; 56:1246-51. [PMID: 26069303 DOI: 10.2967/jnumed.115.156216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/28/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Periostin, an extracellular matrix protein, plays key role in cell adhesion and motility within the tumor microenvironment and is correlated with tumor invasion. We developed and characterized a PET tracer that specifically targets periostin and evaluated the probe in preclinical models of esophageal squamous cell carcinoma (ESCC). METHODS The Institutional Animal Care and Use Committee approved all animal studies. Antiperiostin-F(ab')2 was generated from a monoclonal antibody by enzymatic digestion, conjugated to DOTA, and labeled with (64)Cu. Human ESCC cell lines, TE-11 with high and TT with minimal periostin expression, were implanted in nu/nu mice to generate the positive and control tumor models, respectively. PET/CT imaging was performed at 6, 12, and 24 h and organ-specific biodistribution at 24 h after probe injection. Additionally the probe was tested in a genetically engineered mouse model of periostin-expressing distal esophageal/forestomach ESCC. Tissue microarrays of esophageal neoplasms and ESCC as well as extracted tumor samples were stained for periostin. RESULTS We generated a (64)Cu-DOTA-anti-periostin-F(ab')2 with a dissociation constant of 29.2 ± 3.0 nM. PET/CT images and biodistribution studies showed significantly higher tracer uptake in TE-11 than TT tumors (maximum standardized uptake value, 24 h: 0.67 ± 0.09 vs. 0.36 ± 0.03, P < 0.0005; percentage injected dose per gram, 24 h: 3.24 ± 0.65 vs. 1.63 ± 0.49, P < 0.0001). In genetically engineered mouse models, ESCC high periostin tracer uptake anatomically correlated with the (18)F-FDG uptake at the gastroesophageal junction. All of the ESCC cores and 96.2% of adenocarcinoma stained positive for periostin, with most stained strongly (67.3% and 69.3%, respectively). CONCLUSION We demonstrated that specific imaging of extracellular matrix periostin in ESCC is feasible using a targeted PET tracer. Detection of periostin in the tumor microenvironment may help with early detection, postsurgical follow-up, and in situ characterization of primary and metastatic lesions.
Collapse
Affiliation(s)
- Pedram Heidari
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Shadi A Esfahani
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Nazife S Turker
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Gabrielle Wong
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York; and
| | - Timothy C Wang
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Anil K Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York; and
| | - Umar Mahmood
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
59
|
Wu H, Xie J, Li GN, Chen QH, Li R, Zhang XL, Kang LN, Xu B. Possible involvement of TGF-β/periostin in fibrosis of right atrial appendages in patients with atrial fibrillation. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:6859-6869. [PMID: 26261573 PMCID: PMC4525907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/20/2015] [Indexed: 06/04/2023]
Abstract
Atrial fibrosis contributes to development and recurrence of atrial fibrillation (AF). TGF-β and periostin have been reported to be involved in fibrogenesis. Here we investigated the role of TGF-β and periostin in atrial fibrosis of AF and in the recurrence of AF after surgery ablation. Western blot, Masson staining, immunohistochemistry and colorimetry were performed to detect the degree of atrial fibrosis and the expression of TGF-β, periostin and collagens in 70 biopsies of right atrial appendage (RAA) obtained in this study. Then the patients who received surgical ablation were followed up for about one year. The results showed an increasing gradient of atrial expression of TGF-β, periostin and collagens paralleled by a higher level of atrial fibrosis in control, SR and AF groups. The expression of TGF-β and periostin was significantly correlated with fibrotic markers. In addition, LAD and the expression of TGF-β were larger or higher in recurrence group than that in nonrecurrence group after surgery ablation. The results suggest that upregulated expression of TGF-β and periostin in RAAs is correlated with the degree of atrial fibrosis in patients with AF.
Collapse
Affiliation(s)
- Han Wu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| | - Jun Xie
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| | - Guan-Nan Li
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| | - Qin-Hua Chen
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| | - Ran Li
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| | - Xin-Lin Zhang
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| | - Li-Na Kang
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| | - Biao Xu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School Nanjing 210008, China
| |
Collapse
|
60
|
Duan B, Hockaday LA, Das S, Xu C, Butcher JT. Comparison of Mesenchymal Stem Cell Source Differentiation Toward Human Pediatric Aortic Valve Interstitial Cells within 3D Engineered Matrices. Tissue Eng Part C Methods 2015; 21:795-807. [PMID: 25594437 DOI: 10.1089/ten.tec.2014.0589] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Living tissue-engineered heart valves (TEHV) would be a major benefit for children who require a replacement with the capacity for growth and biological integration. A persistent challenge for TEHV is accessible human cell source(s) that can mimic native valve cell phenotypes and matrix remodeling characteristics that are essential for long-term function. Mesenchymal stem cells derived from bone marrow (BMMSC) or adipose tissue (ADMSC) are intriguing cell sources for TEHV, but they have not been compared with pediatric human aortic valve interstitial cells (pHAVIC) in relevant 3D environments. In this study, we compared the spontaneous and induced multipotency of ADMSC and BMMSC with that of pHAVIC using different induction media within three-dimensional (3D) bioactive hybrid hydrogels with material modulus comparable to that of aortic heart valve leaflets. pHAVIC possessed some multi-lineage differentiation capacity in response to induction media, but limited to the earliest stages and much less potent than either ADMSC or BMMSC. ADMSC expressed cell phenotype markers more similar to pHAVIC when conditioned in basic fibroblast growth factor (bFGF) containing HAVIC growth medium, while BMMSC generally expressed similar extracellular matrix remodeling characteristics to pHAVIC. Finally, we covalently attached bFGF to PEG monoacrylate linkers and further covalently immobilized in the 3D hybrid hydrogels. Immobilized bFGF upregulated vimentin expression and promoted the fibroblastic differentiation of pHAVIC, ADMSC, and BMMSC. These findings suggest that stem cells retain a heightened capacity for osteogenic differentiation in 3D culture, but can be shifted toward fibroblast differentiation through matrix tethering of bFGF. Such a strategy is likely important for utilizing stem cell sources in heart valve tissue engineering applications.
Collapse
Affiliation(s)
- Bin Duan
- 1 Department of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Laura A Hockaday
- 1 Department of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Shoshana Das
- 2 Department of Biological and Environmental Engineering, Cornell University , Ithaca, New York
| | - Charlie Xu
- 2 Department of Biological and Environmental Engineering, Cornell University , Ithaca, New York
| | - Jonathan T Butcher
- 1 Department of Biomedical Engineering, Cornell University , Ithaca, New York
| |
Collapse
|
61
|
Zhang Z, Elsayed AK, Shi Q, Zhang Y, Zuo Q, Li D, Lian C, Tang B, Xiao T, Xu Q, Chang G, Chen G, Zhang L, Wang K, Wang Y, Jin K, Wang Y, Song J, Cui H, Li B. Crucial genes and pathways in chicken germ stem cell differentiation. J Biol Chem 2015; 290:13605-21. [PMID: 25847247 DOI: 10.1074/jbc.m114.601401] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Indexed: 12/16/2022] Open
Abstract
Male germ cell differentiation is a subtle and complex regulatory process. Currently, its regulatory mechanism is still not fully understood. In our experiment, we performed the first comprehensive genome and transcriptome-wide analyses of the crucial genes and signaling pathways in three kinds of crucial cells (embryonic stem cells, primordial germ cell, and spermatogonial stem cells) that are associated with the male germ cell differentiation. We identified thousands of differentially expressed genes in this process, and from these we chose 173 candidate genes, of which 98 genes were involved in cell differentiation, 19 were involved in the metabolic process, and 56 were involved in the differentiation and metabolic processes, like GAL9, AMH, PLK1, and PSMD7 and so on. In addition, we found that 18 key signaling pathways were involved mainly in cell proliferation, differentiation, and signal transduction processes like TGF-β, Notch, and Jak-STAT. Further exploration found that the candidate gene expression patterns were the same between in vitro induction experiments and transcriptome results. Our results yield clues to the mechanistic basis of male germ cell differentiation and provide an important reference for further studies.
Collapse
Affiliation(s)
- Zhentao Zhang
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Ahmed Kamel Elsayed
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China, the Anatomy and Embryology Department, College of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Qingqing Shi
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Yani Zhang
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China,
| | - Qisheng Zuo
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Dong Li
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Chao Lian
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Beibei Tang
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Tianrong Xiao
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Qi Xu
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Guobin Chang
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Guohong Chen
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Lei Zhang
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Kehua Wang
- the Poultry Institute, Chinese Academy of Agricultural Sciences, 225009 Yangzhou, China
| | - Yingjie Wang
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Kai Jin
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Yilin Wang
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Jiuzhou Song
- the Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland 20740, and
| | - Hengmi Cui
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China
| | - Bichun Li
- From the College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, China,
| |
Collapse
|
62
|
Crawford J, Nygard K, Gan BS, O'Gorman DB. Periostin induces fibroblast proliferation and myofibroblast persistence in hypertrophic scarring. Exp Dermatol 2015; 24:120-6. [DOI: 10.1111/exd.12601] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Justin Crawford
- Cell and Molecular Biology Laboratory; Roth
- McFarlane Hand and Upper Limb Centre; Western University; London ON Canada
- Lawson Health Research Institute; Western University; London ON Canada
- Department of Biochemistry; Western University; London ON Canada
| | - Karen Nygard
- Biotron Experimental Climate Change Research Facility; Western University; London ON Canada
| | - Bing Siang Gan
- Cell and Molecular Biology Laboratory; Roth
- McFarlane Hand and Upper Limb Centre; Western University; London ON Canada
- Lawson Health Research Institute; Western University; London ON Canada
- Department of Medical Biophysics; Western University; London ON Canada
- Department of Surgery; Western University; London ON Canada
| | - David Brian O'Gorman
- Cell and Molecular Biology Laboratory; Roth
- McFarlane Hand and Upper Limb Centre; Western University; London ON Canada
- Lawson Health Research Institute; Western University; London ON Canada
- Department of Biochemistry; Western University; London ON Canada
- Department of Surgery; Western University; London ON Canada
| |
Collapse
|
63
|
Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth. Nat Cell Biol 2015; 17:170-82. [PMID: 25580734 PMCID: PMC4312504 DOI: 10.1038/ncb3090] [Citation(s) in RCA: 692] [Impact Index Per Article: 69.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 11/26/2014] [Indexed: 02/06/2023]
Abstract
Tumor-associated macrophages (TAMs) are enriched in glioblastoma (GBM) that contains glioma stem cells (GSCs) at the apex of its cellular hierarchy. The correlation between TAM density and glioma grade suggests a supportive role of TAMs in tumor progression. Here we interrogated the molecular link between GSCs and TAM recruitment in GBMs and demonstrated that GSCs secrete Periostin (POSTN) to recruit TAMs. TAM density correlates with POSTN levels in human GBMs. Silencing POSTN in GSCs markedly reduced TAM density, inhibited tumor growth, and increased survival of mice bearing GSC-derived xenografts. We found that TAMs in GBMs are not brain-resident microglia, but mainly monocyte-derived macrophages from peripheral blood. Disrupting POSTN specifically attenuated the tumor supportive M2 type of TAMs in xenografts. POSTN recruits TAMs through integrin αvβ3 as blocking this signaling by an RGD peptide inhibited TAM recruitment. Our findings highlight the possibility of improving GBM treatment by targeting POSTN-mediated TAM recruitment.
Collapse
|
64
|
Abstract
Over the last decade, identification and characterization of novel markers of progression and targets for therapy of chronic kidney disease (CKD) have been challenging for the research community. Several promising candidates have emerged, mainly from experimental models of CKD that are yet to be investigated in clinical studies. The authors identified two candidate genes: periostin, an extracellular matrix protein involved in bone and dental development, and the discoidin domain receptor 1 (DDR1), a collagen-binding membrane receptor with tyrosine kinase activity. Both genes are inactive in adulthood under normal conditions but have been shown to be highly inducible following injury to glomerular or tubular epithelial cells. The objective of this review is to summarize recent evidence supporting the role of periostin and DDR1 as potential novel biomarkers and therapeutic targets in CKD.
Collapse
|
65
|
Hwang EY, Jeong MS, Park EK, Kim JH, Jang SB. Structural characterization and interaction of periostin and bone morphogenetic protein for regulation of collagen cross-linking. Biochem Biophys Res Commun 2014; 449:425-31. [PMID: 24858685 DOI: 10.1016/j.bbrc.2014.05.055] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/14/2014] [Indexed: 11/19/2022]
Abstract
Periostin appears to be a unique extracellular protein secreted by fibroblasts that is upregulated following injury to the heart or changes in the environment. Periostin has the ability to associate with other critical extracellular matrix (ECM) regulators such as TGF-β, tenascin, and fibronectin, and is a critical regulator of fibrosis that functions by altering the deposition and attachment of collagen. Periostin is known to be highly expressed in carcinoma cells, but not in normal breast tissues. The protein has a structural similarity to insect fasciclin-1 (Fas 1) and can be induced by transforming growth factor-β (TGF-β) and bone morphogenetic protein (BMP)-2. To investigate the molecular interaction of periostin and bone morphogenetic protein, we modeled these three-dimensional structures and their binding sites. We demonstrated direct interaction between periostin and BMP1/2 in vitro using several biochemical and biophysical assays. We found that the structures of the first, second, and fourth Fas1 domains in periostin are similar to that of the fourth Fas 1 domain of TGFBIp. However, the structure of the third Fas 1 domain in periostin is different from those of the first, second, and fourth Fas1 domains, while it is similar to the NMR structure of Fasciclin-like protein from Rhodobacter sphaeroides. These results will useful in further functional analysis of the interaction of periostin and bone morphogenetic protein.
Collapse
Affiliation(s)
- Eun Young Hwang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Republic of Korea
| | - Mi Suk Jeong
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Republic of Korea
| | - Eun-Kyeong Park
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, College of Medicine, Pusan National University, Busan 602-735, Republic of Korea
| | - Se Bok Jang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Republic of Korea.
| |
Collapse
|
66
|
Abstract
Extracellular matrix (ECM) is not only involved in the maintenance of normal physiological tissue but also in interactions with other ECM components, tissue remodeling, and modulating immune responses. The skin provides a distinctive environment characterized by rich fibroblasts producing various ECM proteins, epithelial-mesenchymal interactions, and immune responses induced by external stimuli. Recently, periostin-a matricellular protein-has been highlighted for its pivotal functions in the skin. Analysis of periostin null mice has revealed that periostin contributes to collagen fibrillogenesis, collagen cross-linking, and the formation of ECM meshwork via interactions with other ECM components. Periostin expression is enhanced by mechanical stress or skin injury; this is indicative of the physiologically protective functions of periostin, which promotes wound repair by acting on keratinocytes and fibroblasts. Along with its physiological functions, periostin plays pathogenic roles in skin fibrosis and chronic allergic inflammation. In systemic sclerosis (SSc) patients, periostin levels reflect the severity of skin fibrosis. Periostin null mice have shown reduced skin fibrosis in a bleomycin-induced SSc mouse model, indicating a key role of periostin in fibrosis. Moreover, in atopic dermatitis (AD), attenuated AD phenotype has been observed in periostin null mice in a house dust mite extract-induced AD mouse model. Th2 cytokine-induced periostin acts on keratinocytes to produce inflammatory cytokines that further enhance the Th2 response, thereby sustaining and amplifying chronic allergic inflammation. Thus, periostin is deeply involved in the pathogenesis of AD and other inflammation-related disorders affecting the skin. Understanding the dynamic actions of periostin would be key to dissecting pathogenesis of skin-related diseases and to developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| |
Collapse
|
67
|
Izuhara K, Arima K, Ohta S, Suzuki S, Inamitsu M, Yamamoto KI. Periostin in allergic inflammation. Allergol Int 2014; 63:143-151. [PMID: 24662806 DOI: 10.2332/allergolint.13-rai-0663] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Indexed: 01/22/2023] Open
Abstract
Periostin, an extracellular matrix protein belonging to the fasciclin family, has been shown to play a critical role in the process of remodeling during tissue/organ development or repair. Periostin functions as a matricellular protein in cell activation by binding to their receptors on cell surface, thereby exerting its biological activities. After we found that periostin is a downstream molecule of interleukin (IL)-4 and IL-13, signature cytokines of type 2 immune responses, we showed that periostin is a component of subepithelial fibrosis in bronchial asthma, the first formal proof that periostin is involved in allergic inflammation. Subsequently, a great deal of evidence has accumulated demonstrating the significance of periostin in allergic inflammation. It is of note that in skin tissues, periostin is critical for amplification and persistence of allergic inflammation by communicating between fibroblasts and keratinocytes. Furthermore, periostin has been applied to development of novel diagnostics or therapeutic agents for allergic diseases. Serum periostin can reflect local production of periostin in inflamed lesions induced by Th2-type immune responses and also can predict the efficacy of Th2 antagonists against bronchial asthma. Blocking the interaction between periostin and its receptor, αv integrin, or down-regulating the periostin expression shows improvement of periostin-induced inflammation in mouse models or in in vitro systems. It is hoped that diagnostics or therapeutic agents targeting periostin will be of practical use in the near future.
Collapse
Affiliation(s)
- Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Kazuhiko Arima
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Shoichiro Ohta
- Department of Laboratory Medicine, Saga Medical School, Saga, Japan
| | - Shoichi Suzuki
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Masako Inamitsu
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Ken-ichi Yamamoto
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| |
Collapse
|
68
|
Mael-Ainin M, Abed A, Conway SJ, Dussaule JC, Chatziantoniou C. Inhibition of periostin expression protects against the development of renal inflammation and fibrosis. J Am Soc Nephrol 2014; 25:1724-36. [PMID: 24578131 DOI: 10.1681/asn.2013060664] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Increased renal expression of periostin, a protein normally involved in embryonic and dental development, correlates with the decline of renal function in experimental models and patient biopsies. Because periostin has been reported to induce cell differentiation, we investigated whether it is also involved in the development of renal disease and whether blocking its abnormal expression improves renal function and/or structure. After unilateral ureteral obstruction in wild-type mice, we observed a progressive increase in the expression and synthesis of periostin in the obstructed kidney that associated with the progression of renal lesions. In contrast, mice lacking the periostin gene showed less injury-induced interstitial fibrosis and inflammation and were protected against structural alterations. This protection was associated with a preservation of the renal epithelial phenotype. In vitro, administration of TGF-β to renal epithelial cells increased the expression of periostin several-fold, leading to subsequent loss of the epithelial phenotype. Furthermore, treatment of these cells with periostin increased the expression of collagen I and stimulated the phosphorylation of FAK, p38, and ERK 42/44. In vivo delivery of antisense oligonucleotides to inhibit periostin expression protected animals from L-NAME-induced renal injury. These data strongly suggest that periostin mediates renal disease in response to TGF-β and that blocking periostin may be a promising therapeutic strategy against the development of CKD.
Collapse
Affiliation(s)
- Mouna Mael-Ainin
- Institut National de la Santé Et de la Recherche Médicale UMRS 702, Tenon Hospital, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Ahmed Abed
- Institut National de la Santé Et de la Recherche Médicale UMRS 702, Tenon Hospital, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Simon J Conway
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Jean-Claude Dussaule
- Institut National de la Santé Et de la Recherche Médicale UMRS 702, Tenon Hospital, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Department of Physiology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christos Chatziantoniou
- Institut National de la Santé Et de la Recherche Médicale UMRS 702, Tenon Hospital, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, Paris, France;
| |
Collapse
|
69
|
Chester AH, El-Hamamsy I, Butcher JT, Latif N, Bertazzo S, Yacoub MH. The living aortic valve: From molecules to function. Glob Cardiol Sci Pract 2014; 2014:52-77. [PMID: 25054122 PMCID: PMC4104380 DOI: 10.5339/gcsp.2014.11] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/28/2014] [Indexed: 12/12/2022] Open
Abstract
The aortic valve lies in a unique hemodynamic environment, one characterized by a range of stresses (shear stress, bending forces, loading forces and strain) that vary in intensity and direction throughout the cardiac cycle. Yet, despite its changing environment, the aortic valve opens and closes over 100,000 times a day and, in the majority of human beings, will function normally over a lifespan of 70–90 years. Until relatively recently heart valves were considered passive structures that play no active role in the functioning of a valve, or in the maintenance of its integrity and durability. However, through clinical experience and basic research the aortic valve can now be characterized as a living, dynamic organ with the capacity to adapt to its complex mechanical and biomechanical environment through active and passive communication between its constituent parts. The clinical relevance of a living valve substitute in patients requiring aortic valve replacement has been confirmed. This highlights the importance of using tissue engineering to develop heart valve substitutes containing living cells which have the ability to assume the complex functioning of the native valve.
Collapse
|
70
|
Ghatak S, Misra S, Norris RA, Moreno-Rodriguez RA, Hoffman S, Levine RA, Hascall VC, Markwald RR. Periostin induces intracellular cross-talk between kinases and hyaluronan in atrioventricular valvulogenesis. J Biol Chem 2014; 289:8545-61. [PMID: 24469446 DOI: 10.1074/jbc.m113.539882] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Periostin (PN), a novel fasciclin-related matricellular protein, has been implicated in cardiac development and postnatal remodeling, but the mechanism remains unknown. We examined the role of PN in mediating intracellular kinase activation for atrioventricular valve morphogenesis using well defined explant cultures, gene transfection systems, and Western blotting. The results show that valve progenitor (cushion) cells secrete PN into the extracellular matrix, where it can bind to INTEGRINs and activate INTEGRIN/focal adhesion kinase signaling pathways and downstream kinases, PI3K/AKT and ERK. Functional assays with prevalvular progenitor cells showed that activating these signaling pathways promoted adhesion, migration, and anti-apoptosis. Through activation of PI3K/ERK, PN directly enhanced collagen expression. Comparing PN-null to WT mice also revealed that expression of hyaluronan (HA) and activation of hyaluronan synthase-2 (Has2) are also enhanced upon PN/INTEGRIN/focal adhesion kinase-mediated activation of PI3K and/or ERK, an effect confirmed by the reduction of HA synthase-2 in PN-null mice. We also identified in valve progenitor cells a potential autocrine signaling feedback loop between PN and HA through PI3K and/or ERK. Finally, in a three-dimensional assay to simulate normal valve maturation in vitro, PN promoted collagen compaction in a kinase-dependent fashion. In summary, this study provides the first direct evidence that PN can act to stimulate a valvulogenic signaling pathway.
Collapse
Affiliation(s)
- Shibnath Ghatak
- From the Department of Regenerative Medicine and Cell Biology
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Kain KH, Miller JWI, Jones-Paris CR, Thomason RT, Lewis JD, Bader DM, Barnett JV, Zijlstra A. The chick embryo as an expanding experimental model for cancer and cardiovascular research. Dev Dyn 2013; 243:216-28. [PMID: 24357262 DOI: 10.1002/dvdy.24093] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 10/28/2013] [Accepted: 10/28/2013] [Indexed: 12/17/2022] Open
Abstract
A long and productive history in biomedical research defines the chick as a model for human biology. Fundamental discoveries, including the description of directional circulation propelled by the heart and the link between oncogenes and the formation of cancer, indicate its utility in cardiac biology and cancer. Despite the more recent arrival of several vertebrate and invertebrate animal models during the last century, the chick embryo remains a commonly used model for vertebrate biology and provides a tractable biological template. With new molecular and genetic tools applied to the avian genome, the chick embryo is accelerating the discovery of normal development and elusive disease processes. Moreover, progress in imaging and chick culture technologies is advancing real-time visualization of dynamic biological events, such as tissue morphogenesis, angiogenesis, and cancer metastasis. A rich background of information, coupled with new technologies and relative ease of maintenance, suggest an expanding utility for the chick embryo in cardiac biology and cancer research.
Collapse
|
72
|
Tang M, Yuan W, Bodmer R, Wu X, Ocorr K. The role of pygopus in the differentiation of intracardiac valves in Drosophila. Genesis 2013; 52:19-28. [PMID: 24265259 DOI: 10.1002/dvg.22724] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 12/14/2022]
Abstract
Cardiac valves serve an important function; they support unidirectional blood flow and prevent blood regurgitation. Wnt signaling plays an important role in the formation of mouse cardiac valves and cardiac valve proliferation in Zebrafish, but identification of the specific signaling components involved has not been addressed systematically. Of the components involved in Wnt signal transduction, pygopus (pygo), first identified as a core component of Wnt signaling in Drosophila, has not yet to be investigated with respect to valve development and differentiation. Here, we take advantage of the Drosophila heart model to study the role of pygo in formation of valves between the cardiac chambers. We found that cardiac-specific pygo knockdown in the Drosophila heart causes dilation in the region of these cardiac valves, and their characteristic dense mesh of myofibrils does not form and resembles that of neighboring cardiomyocytes. In contrast, heart-specific knockdown of the transcription factors, arm/β-Cat, lgs/BCL9, or pan/TCF, which mediates canonical Wnt signal transduction, shows a much weaker valve differentiation defect. Double-heterozygous combinations of mutants for pygo and the Wnt-signaling components have no additional effect on heart function compared with pygo heterozygotes alone. These results are consistent with the idea that pygo functions independently of canonical Wnt signaling in the differentiation of the adult interchamber cardiac valves.
Collapse
Affiliation(s)
- Min Tang
- The Center for Heart Development, Key Laboratory of MOE for Developmental Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, People's Republic of China; Development and Aging Program, Sanford-Burnham Medical Research Institute, La Jolla, California, 92037
| | | | | | | | | |
Collapse
|
73
|
Roy R, Kukucka M, Messroghli D, Kunkel D, Brodarac A, Klose K, Geißler S, Becher PM, Kang SK, Choi YH, Stamm C. Epithelial-to-Mesenchymal Transition Enhances the Cardioprotective Capacity of Human Amniotic Epithelial Cells. Cell Transplant 2013; 24:985-1002. [PMID: 24256742 DOI: 10.3727/096368913x675151] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The amniotic epithelium consists of cells exhibiting mature epithelial cell characteristics, but also varying degrees of stemness. We tested the hypothesis that induction of epithelial-to-mesenchymal transition (EMT) in amniotic epithelial cells (AECs) derived from human placenta enhances their capacity to support the ischemic myocardium. In response to incubation with transforming growth factor-β1 (TGF-β1) protein, AECs lost their cobblestone morphology and acquired a fibroblastoid shape, associated with downregulation of E-cadherin, upregulation of N-cadherin, Akt phosphorylation, and intracellular periostin translocation. EMT-AECs displayed greatly enhanced mobility and secreted gelatinase activity compared with naive AECs. The surface presentation of CD105 and CD73 decreased, and RNA microarray analysis mirrored the loss of epithelial characteristics and transcriptional profile. Unmodified AECs and EMT-AECs were then injected intramyocardially in fully immunocompetent mice after permanent LAD ligation, and heart function was followed by MRI as well as 2D speckle tracking echocardiography after 4 weeks. EMT-AEC-treated infarct hearts displayed better global systolic function and improved longitudinal strain rate in the area of interest. Although no signals of human cells were detectable by histology, infarct size was smaller in EMT-AEC-treated hearts, associated with fewer TUNEL-positive cells and upregulation of periostin, while blood vessel density was increased in both ACE- and EMT-AEC-treated hearts. We conclude that EMT enhances the cardioprotective effects of human AECs.
Collapse
Affiliation(s)
- Rajika Roy
- Berlin Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Inai K, Burnside JL, Hoffman S, Toole BP, Sugi Y. BMP-2 induces versican and hyaluronan that contribute to post-EMT AV cushion cell migration. PLoS One 2013; 8:e77593. [PMID: 24147033 PMCID: PMC3795687 DOI: 10.1371/journal.pone.0077593] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/09/2013] [Indexed: 11/24/2022] Open
Abstract
Distal outgrowth and maturation of mesenchymalized endocardial cushions are critical morphogenetic events during post-EMT atrioventricular (AV) valvuloseptal morphogenesis. We explored the role of BMP-2 in the regulation of valvulogenic extracellular matrix (ECM) components, versican and hyaluronan (HA), and cell migration during post-EMT AV cushion distal outgrowth/expansion. We observed intense staining of versican and HA in AV cushion mesenchyme from the early cushion expansion stage, Hamburger and Hamilton (HH) stage-17 to the cushion maturation stage, HH stage-29 in the chick. Based on this expression pattern we examined the role of BMP-2 in regulating versican and HA using 3D AV cushion mesenchymal cell (CMC) aggregate cultures on hydrated collagen gels. BMP-2 induced versican expression and HA deposition as well as mRNA expression of versican and Has2 by CMCs in a dose dependent manner. Noggin, an antagonist of BMP, abolished BMP-2-induced versican and HA as well as mRNA expression of versican and Has2. We further examined whether BMP-2-promoted cell migration was associated with expression of versican and HA. BMP-2- promoted cell migration was significantly impaired by treatments with versican siRNA and HA oligomer. In conclusion, we provide evidence that BMP-2 induces expression of versican and HA by AV CMCs and that these ECM components contribute to BMP-2-induced CMC migration, indicating critical roles for BMP-2 in distal outgrowth/expansion of mesenchymalized AV cushions.
Collapse
Affiliation(s)
- Kei Inai
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jessica L. Burnside
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Stanley Hoffman
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Bryan P. Toole
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yukiko Sugi
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
75
|
Wang X, Liu J, Wang Z, Huang Y, Liu W, Zhu X, Cai Y, Fang X, Lin S, Yuan L, Ouyang G. Periostin contributes to the acquisition of multipotent stem cell-like properties in human mammary epithelial cells and breast cancer cells. PLoS One 2013; 8:e72962. [PMID: 24009721 PMCID: PMC3756944 DOI: 10.1371/journal.pone.0072962] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 07/21/2013] [Indexed: 12/22/2022] Open
Abstract
Periostin (POSTN), a recently characterised matricellular protein, is frequently dysregulated in various malignant cancers and promotes tumor metastatic growth. POSTN plays a critical role in the crosstalk between murine breast cancer stem cells (CSCs) and their niche to permit metastatic colonization. However, whether pro-metastatic capability of POSTN is associated with multipotent potentials of mesenchymal stem cells (MSCs) has not been documented. Here we demonstrate that POSTN promotes a stem cell-like trait and a mesenchymal phenotype in human mammary epithelial cells and breast cancer cells. Interestingly, ectopic overexpression of POSTN or recombinant POSTN treatment can induce human mammary epithelial cells and breast cancer cells differentiation into multiple cell lineages that recapitulate part of the multilineage differentiation potentials of MSCs. Moreover, POSTN is highly expressed in bone marrow-derived MSCs and their derived adipocytes, chondrocytes, and osteoblasts in vitro. Furthermore, POSTN promotes the growth of xenograft tumors in vivo. POSTN-overexpressing human mammary epithelial cells enhance breast tumor growth and metastasis. These data thus provide evidence of a new role for POSTN in mammary epithelial neoplasia and metastasis, suggesting that epithelial cancer cells might acquire CSC-like traits and a mesenchymal phenotype, as well as the multipotent potentials of MSCs to promote tumorigenesis and metastasis. Therefore, targeting POSTN and other extracellular matrix components of tumor microenvironment may help to develop new therapeutical strategies to inhibit tumor metastasis.
Collapse
Affiliation(s)
- Xiaowei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jia Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhe Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yangmei Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Weiping Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiao Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yao Cai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiaoguang Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Shuyong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Li Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Gaoliang Ouyang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
- * E-mail:
| |
Collapse
|
76
|
Beigi F, Schmeckpeper J, Pow-Anpongkul P, Payne JA, Zhang L, Zhang Z, Huang J, Mirotsou M, Dzau VJ. C3orf58, a novel paracrine protein, stimulates cardiomyocyte cell-cycle progression through the PI3K-AKT-CDK7 pathway. Circ Res 2013; 113:372-80. [PMID: 23784961 DOI: 10.1161/circresaha.113.301075] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
RATIONALE The regenerative capacity of the heart is markedly diminished shortly after birth, coinciding with overall withdrawal of cardiomyocytes from cell cycle. Consequently, the adult mammalian heart has limited capacity to regenerate after injury. The discovery of factors that can induce cardiomyocyte proliferation is, therefore, of high interest and has been the focus of extensive investigation throughout the past years. OBJECTIVE We have recently identified C3orf58 as a novel hypoxia and Akt induced stem cell factor (HASF) secreted from mesenchymal stem cells, which can promote cardiac repair through cytoprotective mechanisms. Here, we tested the hypothesis that HASF can also contribute to cardiac regeneration by stimulating cardiomyocyte division and proliferation. METHODS AND RESULTS Neonatal ventricular cardiomyocytes were stimulated in culture for 7 days with purified recombinant HASF protein. Compared with control untreated cells, HASF-treated neonatal cardiomyocytes exhibited 60% increase in DNA synthesis as measured by bromodeoxyuridine incorporation. These results were confirmed by immunofluorescence confocal microscopy showing a 50% to 100% increase in the number of cardiomyocytes in the mitotic and cytokinesis phases. Importantly, in vivo cardiac overexpression of HASF in a transgenic mouse model resulted in enhanced level of DNA synthesis and cytokinesis in neonatal and adult cardiomyocytes. These proliferative effects were modulated by a phosphoinositide 3-kinase-protein kinase B-cycle-dependent kinase 7 pathway as revealed by the use of phosphoinositide 3-kinase -pathway-specific inhibitors and silencing of the Cdk7 gene. CONCLUSIONS Our studies support the hypothesis that HASF induces cardiomyocyte proliferation via a phosphoinositide 3-kinase-protein kinase B-cycle-dependent kinase 7 pathway. The implications of this finding may be significant for cardiac regeneration biology and therapeutics.
Collapse
Affiliation(s)
- Farideh Beigi
- Department of Medicine, Mandel Center for Hypertension and Atherosclerosis Research, Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Roy R, Brodarac A, Kukucka M, Kurtz A, Becher PM, Jülke K, Choi YH, Pinzur L, Chajut A, Tschöpe C, Stamm C. Cardioprotection by placenta-derived stromal cells in a murine myocardial infarction model. J Surg Res 2013; 185:70-83. [PMID: 23830369 DOI: 10.1016/j.jss.2013.05.084] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/15/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Autologous cells for cell therapy of ischemic cardiomyopathy often display age- and disease-related functional impairment, whereas an allogenic immunotolerant cell product would allow off-the-shelf application of uncompromised donor cells. We investigated the cardiac regeneration potential of a novel, clinical-grade placenta-derived human stromal cell product (PLX-PAD). METHODS PLX-PAD cells derived from human donor placentas and expanded in a three-dimensional bioreactor system were tested for surface marker expression, proangiogenic, anti-inflammatory, and immunomodulatory properties in vitro. In BALB/C mice, the left anterior descending artery was ligated and PLX-PAD cells (n = 10) or vehicle (n = 10) were injected in the infarct border zone. Four weeks later, heart function was analyzed by two-dimensional and M-mode echocardiography. Scar size, microvessel density, extracellular matrix composition, myocyte apoptosis, and PLX-PAD cell retention were studied by histology. RESULTS In vitro, PLX-PAD cells displayed both proangiogenesis and anti-inflammatory properties, represented by the secretion of both vascular endothelial growth factor and angiopoietin-1 that was upregulated by hypoxia, as well as by the capacity to suppress T-cell proliferation and augment IL-10 secretion when co-cultured with peripheral blood mononuclear cells. Compared with control mice, PLX-PAD-treated hearts had better contractile function, smaller infarct size, greater regional left ventricular wall thickness, and less apoptosis after 4 wk. PLX-PAD stimulated both angiogenesis and arteriogenesis in the infarct border zone, and periostin expression was upregulated in PLX-PAD-treated hearts. CONCLUSIONS Clinical-grade PLX-PAD cells exert beneficial effects on ischemic myocardium that are associated with improved contractile function, and may be suitable for further evaluation aiming at clinical pilot trials of cardiac cell therapy.
Collapse
Affiliation(s)
- Rajika Roy
- Berlin-Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Ahlfeld SK, Gao Y, Wang J, Horgusluoglu E, Bolanis E, Clapp DW, Conway SJ. Periostin downregulation is an early marker of inhibited neonatal murine lung alveolar septation. ACTA ACUST UNITED AC 2013; 97:373-85. [PMID: 23723163 DOI: 10.1002/bdra.23149] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/12/2013] [Accepted: 04/18/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Extreme preterm birth exposes the saccular lung to multiple teratogens, which ultimately retard alveolar development. Specifically, therapeutic high level oxygen supplementation adversely affects the premature lungs and results in blunted alveolarization. Prolonged hyperoxic lung injury has previously been shown to upregulate the matricellular protein Periostin (Postn) and stimulate ectopic accumulation of alpha smooth muscle actin (αSMA) myofibroblasts. Therapies that promote lung septation are lacking largely due to a lack of reliable early biomarkers of injury. Thus, we determined if Postn expression correlated with the initial appearance of myofibroblasts in the saccular lung and was required for early alveolar development. METHODS Lung development in C57BL/6J mice following room-air (RA, 21%-O₂) or continuous hyperoxia (85%-O₂) from birth (P0) through postnatal day P14 was correlated with Postn and αSMA expression. Alveolarization in Postn knockout mice exposed to room-air, 60%-, and 85%-O₂ was also examined. RESULTS Postn was widely expressed in distal lung septa through P2 to P4 and peak expression coincided with accumulation of saccular myofibroblasts. Initially, 85%-O₂ prematurely downregulated Postn and αSMA expression and suppressed proliferation before the first evidence of distal lung simplification at P4. By P14, chronic 85%-O₂ resulted in secondary upregulation of Postn and αSMA in blunted septa. Myofibroblast differentiation and alveolar development was unaffected in Postn null mice and acute 85%-O₂ exposure equally inhibited septal formation in Postn null and wild-type littermates. CONCLUSION Postn expression is tightly correlated with the presence of αSMA-myofibroblasts and is a novel early biomarker of acutely inhibited alveolar septation during a crucial window of lung development.
Collapse
Affiliation(s)
- Shawn K Ahlfeld
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
79
|
Johansson MW, Annis DS, Mosher DF. α(M)β(2) integrin-mediated adhesion and motility of IL-5-stimulated eosinophils on periostin. Am J Respir Cell Mol Biol 2013; 48:503-10. [PMID: 23306834 DOI: 10.1165/rcmb.2012-0150oc] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Periostin is an extracellular matrix protein that is up-regulated by T helper cell type 2 cytokines in the asthmatic airway and implicated in mouse studies as promoting eosinophil recruitment. We asked whether periostin modulates eosinophil adhesion and motility in vitro. Periostin adsorbed to polystyrene supported adhesion of purified human blood eosinophils stimulated by IL-5, IL-3, or granulocyte/macrophage colony-stimulating factor, but did not support adhesion of eosinophils treated with IL-4 or IL-13. The degree of adhesion depended on the concentrations of periostin during coating and activating cytokine during the adhesion assay. Both full-length periostin and alternatively spliced periostin, lacking C-terminal exons 17, 18, 19, and 21, supported adhesion. Adhesion was inhibited by monoclonal antibody to α(M) or β(2) integrin subunits, but not by antibodies to other eosinophil integrin subunits. Adsorbed periostin also supported α(M)β(2)-dependent random motility of IL-5-stimulated eosinophils with optimal movement at an intermediate coating concentration. In the presence of IL-5, eosinophils adherent on periostin formed punctate structures positive for filamentous actin, gelsolin, and phosphotyrosine. These structures fit the criteria for podosomes, highly dynamic adhesive contacts that are distinct from classical focal adhesions. The results establish α(M)β(2) (CD11b/CD18, Mac-1) as an adhesive and promigratory periostin receptor on cytokine-stimulated eosinophils, and suggest that periostin may function as a haptotactic stimulus able to guide eosinophils to areas of high periostin density in the asthmatic airway.
Collapse
Affiliation(s)
- Mats W Johansson
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53706, USA.
| | | | | |
Collapse
|
80
|
Barczyk MM, Lu N, Popova SN, Bolstad AI, Gullberg D. α11β1 integrin-mediated MMP-13-dependent collagen lattice contraction by fibroblasts: evidence for integrin-coordinated collagen proteolysis. J Cell Physiol 2013; 228:1108-19. [PMID: 23065814 DOI: 10.1002/jcp.24261] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 10/03/2012] [Indexed: 02/01/2023]
Abstract
We have previously determined that integrin α11β1 is required on mouse periodontal ligament (PDL) fibroblasts to generate the force needed for incisor eruption. As part of the phenotype of α11(-/-) mice, the incisor PDL (iPDL) is thickened, due to disturbed matrix remodeling. To determine the molecular mechanism behind the disturbed matrix dynamics in the PDL we crossed α11(-/-) mice with the Immortomouse and isolated immortalized iPDL cells. Microarray analysis of iPDL cells cultured inside a 3D collagen gel demonstrated downregulated expression of a number of genes in α11-deficient iPDL cells, including matrix metalloproteinase-13 (MMP-13) and cathepsin K. α11(-/-) iPDL cells in vitro displayed disturbed interactions with collagen I during contraction of attached and floating collagen lattices and furthermore displayed reduced MMP-13 protein expression levels. The MMP-13 specific inhibitor WAY 170523 and the Cathepsin K Inhibitor II both blocked part of the α11 integrin-mediated collagen remodeling. In summary, our data demonstrate that in iPDL fibroblasts the mechanical strain generated by α11β1 integrin regulates molecules involved in collagen matrix dynamics. The positive regulation of α11β1-dependent matrix remodeling, involving MMP-13 and cathepsin K, might also occur in other types of fibroblasts and be an important regulatory mechanism for coordinated extracellular and intracellular collagen turnover in tissue homeostasis.
Collapse
|
81
|
Wong GS, Habibollahi P, Heidari P, Lee JS, Klein-Szanto AJ, Waldron TJ, Gimotty P, Nakagawa H, Taylor PR, Wang TC, Mahmood U, Rustgi AK. Optical imaging of periostin enables early endoscopic detection and characterization of esophageal cancer in mice. Gastroenterology 2013; 144:294-297. [PMID: 23085486 PMCID: PMC3624041 DOI: 10.1053/j.gastro.2012.10.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 12/29/2022]
Abstract
Imaging strategies that detect early stage esophageal squamous cell carcinoma (ESCC) could improve clinical outcomes, when combined with endoscopic approaches. Periostin is an integrin-binding protein that is important in the tumor microenvironment. We created a fluorescent-labeled antibody that recognizes periostin and binds specifically to ESCC xenograft tumors in mice. In L2-cre;p120ctnLoxP/LoxP mice, which develop squamous cell cancers that resemble human ESCC, we visualized the probe in preneoplastic and neoplastic esophageal lesions using near-infrared fluorescent imaging with upper-gastrointestinal endoscopy. Periostin might be a biomarker of the esophageal tumor microenvironment that can be used to detect preneoplastic lesions.
Collapse
Affiliation(s)
- Gabrielle S. Wong
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peiman Habibollahi
- ) Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Pedram Heidari
- ) Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ju-Seog Lee
- ) Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Andres J. Klein-Szanto
- ) Department of Pathology and Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, 19104, USA
| | - Todd J. Waldron
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Phyllis Gimotty
- ) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA,) Division of Biostatistics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hiroshi Nakagawa
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Philip R. Taylor
- ) Genetic Epidemiology Branch, DCEG, NCI, NIH, Bethesda, MD 20892, USA
| | - Timothy C. Wang
- ) Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA,) Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, 10032, USA
| | - Umar Mahmood
- ) Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Anil K. Rustgi
- ) Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA,) Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA,) Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
82
|
Abstract
Regenerative medicine seeks to understand tissue development and homeostasis and build on that knowledge to enhance regeneration of injured tissues. By replenishing lost functional tissues and cells, regenerative medicine could change the treatment paradigm for a broad range of degenerative and ischemic diseases. Multipotent cells hold promise as potential building blocks for regenerating lost tissues, but successful tissue regeneration will depend on comprehensive control of multipotent cells-differentiation into a target cell type, delivery to a desired tissue, and integration into a durable functional structure. At each step of this process, proteins and small molecules provide essential signals and, in some cases, may themselves act as effective therapies. Identifying these signals is thus a fundamental goal of regenerative medicine. In this review we discuss current progress using proteins and small molecules to regulate tissue regeneration, both in combination with cellular therapies and as monotherapy.
Collapse
Affiliation(s)
- Eric M Green
- Harvard Stem Cell Institute and the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
83
|
Zelarayán LC, Zafiriou MP, Zimmermann WH. Emerging Concepts in Myocardial Pharmacoregeneration. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
84
|
Abstract
Eosinophilic esophagitis (EoE) is a recently recognized, immune-mediated disease characterized clinically by symptoms of esophageal dysfunction and histologically by eosinophil-predominant inflammation. The chronic esophageal eosinophilia of EoE is associated with tissue remodeling that includes epithelial hyperplasia, subepithelial fibrosis, and hypertrophy of esophageal smooth muscle. This remodeling causes the esophageal rings and strictures that frequently complicate EoE and underlies the mucosal fragility that predisposes to painful mucosal tears in the EoE esophagus. The pathogenesis of tissue remodeling in EoE is not completely understood, but emerging studies suggest that secretory products of eosinophils and mast cells, as well as cytokines produced by other inflammatory cells, epithelial cells, and stromal cells in the esophagus, all contribute to the process. Interleukin (IL)-4 and IL-13, Th2 cytokines overproduced in allergic disorders, have direct profibrotic and remodeling effects in EoE. The EoE esophagus exhibits increased expression of transforming growth factor (TGF)-β1, which is a potent activator of fibroblasts and a strong inducer of epithelial-mesenchymal transition. In addition, IL-4, IL-13, and TGF-β all have a role in regulating periostin, an extracellular matrix protein that might influence remodeling by acting as a ligand for integrins, by its effects on eosinophils or by activating fibrogenic genes in the esophagus. Presently, few treatments have been shown to affect the tissue remodeling that causes EoE complications. This report reviews the potential roles of fibroblasts, eosinophils, mast cells, and profibrotic cytokines in esophageal remodeling in EoE and identifies potential targets for future therapies that might prevent EoE complications.
Collapse
Affiliation(s)
| | - Rhonda F. Souza
- 2Internal Medicine, Children's Medical Center and the VA North Texas Health Care System, Harold C. Simmons Comprehensive Cancer Center, and the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stuart J. Spechler
- 2Internal Medicine, Children's Medical Center and the VA North Texas Health Care System, Harold C. Simmons Comprehensive Cancer Center, and the University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
85
|
Sauls K, de Vlaming A, Harris BS, Williams K, Wessels A, Levine RA, Slaugenhaupt SA, Goodwin RL, Pavone LM, Merot J, Schott JJ, Le Tourneau T, Dix T, Jesinkey S, Feng Y, Walsh C, Zhou B, Baldwin S, Markwald RR, Norris RA. Developmental basis for filamin-A-associated myxomatous mitral valve disease. Cardiovasc Res 2012; 96:109-19. [PMID: 22843703 DOI: 10.1093/cvr/cvs238] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS We hypothesized that the structure and function of the mature valves is largely dependent upon how these tissues are built during development, and defects in how the valves are built can lead to the pathological progression of a disease phenotype. Thus, we sought to uncover potential developmental origins and mechanistic underpinnings causal to myxomatous mitral valve disease. We focus on how filamin-A, a cytoskeletal binding protein with strong links to human myxomatous valve disease, can function as a regulatory interface to control proper mitral valve development. METHODS AND RESULTS Filamin-A-deficient mice exhibit abnormally enlarged mitral valves during foetal life, which progresses to a myxomatous phenotype by 2 months of age. Through expression studies, in silico modelling, 3D morphometry, biochemical studies, and 3D matrix assays, we demonstrate that the inception of the valve disease occurs during foetal life and can be attributed, in part, to a deficiency of interstitial cells to efficiently organize the extracellular matrix (ECM). This ECM organization during foetal valve gestation is due, in part, to molecular interactions between filamin-A, serotonin, and the cross-linking enzyme, transglutaminase-2 (TG2). Pharmacological and genetic perturbations that inhibit serotonin-TG2-filamin-A interactions lead to impaired ECM remodelling and engender progression to a myxomatous valve phenotype. CONCLUSIONS These findings illustrate a molecular mechanism by which valve interstitial cells, through a serotonin, TG, and filamin-A pathway, regulate matrix organization during foetal valve development. Additionally, these data indicate that disrupting key regulatory interactions during valve development can set the stage for the generation of postnatal myxomatous valve disease.
Collapse
Affiliation(s)
- Kimberly Sauls
- Department of Regenerative Medicine and Cell Biology, School of Medicine, Cardiovascular Developmental Biology Center, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Abstract
The term matricellular proteins describes a family of structurally unrelated extracellular macromolecules that, unlike structural matrix proteins, do not play a primary role in tissue architecture, but are induced following injury and modulate cell-cell and cell-matrix interactions. When released to the matrix, matricellular proteins associate with growth factors, cytokines, and other bioactive effectors and bind to cell surface receptors transducing signaling cascades. Matricellular proteins are upregulated in the injured and remodeling heart and play an important role in regulation of inflammatory, reparative, fibrotic and angiogenic pathways. Thrombospondin (TSP)-1, -2, and -4 as well as tenascin-C and -X secreted protein acidic and rich in cysteine (SPARC), osteopontin, periostin, and members of the CCN family (including CCN1 and CCN2/connective tissue growth factor) are involved in a variety of cardiac pathophysiological conditions, including myocardial infarction, cardiac hypertrophy and fibrosis, aging-associated myocardial remodeling, myocarditis, diabetic cardiomyopathy, and valvular disease. This review discusses the properties and characteristics of the matricellular proteins and presents our current knowledge on their role in cardiac adaptation and disease. Understanding the role of matricellular proteins in myocardial pathophysiology and identification of the functional domains responsible for their actions may lead to design of peptides with therapeutic potential for patients with heart disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Ave., Forchheimer G46B, Bronx, NY 10461, USA.
| |
Collapse
|
87
|
Elliott CG, Kim SS, Hamilton DW. Functional significance of periostin in excisional skin repair: is the devil in the detail? Cell Adh Migr 2012; 6:319-26. [PMID: 22983194 DOI: 10.4161/cam.20879] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the past year, three papers have been published exploring the role of the matricellular protein periostin in excisional skin repair. These papers all show a delay in wound closure and the kinetics of this delay are strikingly similar across the three reports. The similarities between these papers end, however, when each investigates the mechanism through which periostin influences skin repair. Three proposed mechanisms have been identified: (1) myofibroblast differentiation, (2) keratinocyte proliferation and (3) fibroblast proliferation and migration. The aim of this commentary is to compare and contrast the three studies performed to date in an attempt to decipher the role of periostin in the repair of full-thickness skin wounds.
Collapse
Affiliation(s)
- Christopher G Elliott
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON Canada
| | | | | |
Collapse
|
88
|
Hur DG, Khalmuratova R, Ahn SK, Ha YS, Min YG. Roles of periostin in symptom manifestation and airway remodeling in a murine model of allergic rhinitis. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2012; 4:222-30. [PMID: 22754716 PMCID: PMC3378929 DOI: 10.4168/aair.2012.4.4.222] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 04/18/2012] [Accepted: 05/03/2012] [Indexed: 11/20/2022]
Abstract
PURPOSE Periostin was originally identified as a secreted factor during screening of a mouse osteoblastic library. In a recent study, periostin was found to directly regulate eosinophil accumulation in allergic mucosal inflammation. Chronic eosinophilic inflammation is related to the development of remodeling. The present study examined the expression of periostin and evaluated its role in the inflammatory process and remodeling associated with allergic rhinitis. METHODS A murine model of allergic rhinitis was established in periostin knockout mice. We analyzed the expression of periostin, manifestation of nasal symptoms, eosinophilic inflammation, and subepithelial fibrosis as well as the expression of MMP-2, TIMP-1, and type 1 collagen in nasal tissue. RESULTS Periostin was mainly distributed in the subepithelial tissue of the nasal mucosa. The subepithelial tissue was thinner in the knockout group than in the control group. No differences in the expression of MMP-2 or TIMP-1 were found in the knockout group. However, after a month of allergen challenge, type I collagen in the nasal tissue was lower in the knockout group than in the control group. The number of eosinophils and the symptom score were also lower in the knockout group. CONCLUSIONS Periostin is expressed in nasal tissues of murine models of allergic rhinitis. Periostin deficiency may affect the remodeling of nasal tissue with reduced subepithelial fibrosis, and lead to less eosinophilic inflammation.
Collapse
Affiliation(s)
- Dong Gu Hur
- Department of Otorhinolaryngology, School of Medicine, Gyeongsang National University, Jinju, Korea
| | | | | | | | | |
Collapse
|
89
|
de Vlaming A, Sauls K, Hajdu Z, Visconti RP, Mehesz AN, Levine RA, Slaugenhaupt SA, Hagège A, Chester AH, Markwald RR, Norris RA. Atrioventricular valve development: new perspectives on an old theme. Differentiation 2012; 84:103-16. [PMID: 22579502 DOI: 10.1016/j.diff.2012.04.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 03/26/2012] [Accepted: 04/01/2012] [Indexed: 11/19/2022]
Abstract
Atrioventricular valve development commences with an EMT event whereby endocardial cells transform into mesenchyme. The molecular events that induce this phenotypic change are well understood and include many growth factors, signaling components, and transcription factors. Besides their clear importance in valve development, the role of these transformed mesenchyme and the function they serve in the developing prevalve leaflets is less understood. Indeed, we know that these cells migrate, but how and why do they migrate? We also know that they undergo a transition to a mature, committed cell, largely defined as an interstitial fibroblast due to their ability to secrete various matrix components including collagen type I. However, we have yet to uncover mechanisms by which the matrix is synthesized, how it is secreted, and how it is organized. As valve disease is largely characterized by altered cell number, cell activation, and matrix disorganization, answering questions of how the valves are built will likely provide us with information of real clinical relevance. Although expression profiling and descriptive or correlative analyses are insightful, to advance the field, we must now move past the simplicity of these assays and ask fundamental, mechanistic based questions aimed at understanding how valves are "built". Herein we review current understandings of atrioventricular valve development and present what is known and what isn't known. In most cases, basic, biological questions and hypotheses that were presented decades ago on valve development still are yet to be answered but likely hold keys to uncovering new discoveries with relevance to both embryonic development and the developmental basis of adult heart valve diseases. Thus, the goal of this review is to remind us of these questions and provide new perspectives on an old theme of valve development.
Collapse
Affiliation(s)
- Annemarieke de Vlaming
- Department of Regenerative Medicine and Cell Biology, School of Medicine, Cardiovascular Developmental Biology Center, Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Merle B, Garnero P. The multiple facets of periostin in bone metabolism. Osteoporos Int 2012; 23:1199-212. [PMID: 22310955 DOI: 10.1007/s00198-011-1892-7] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 12/22/2011] [Indexed: 01/17/2023]
Abstract
Periostin is a matricellular glutamate-containing protein expressed during ontogenesis and in adult connective tissues submitted to mechanical strains including bone and, more specifically, the periosteum, periodontal ligaments, tendons, heart valves, or skin. It is also expressed in neoplastic tissues, cardiovascular and fibrotic diseases, and during wound repair. Its biological functions are extensively investigated in fields such as cardiovascular physiology or oncology. Despite its initial identification in bone, investigations of periostin functions in bone-related physiopathology are less abundant. Recently, several studies have analyzed the potential role of periostin in bone biology and suggest that periostin may be an important regulator of bone formation. The aim of this article is to provide an extensive review on the implications of periostin in bone biology and its potential use in benign and metabolic bone diseases.
Collapse
Affiliation(s)
- B Merle
- INSERM Research Unit 1033, Pavillon F, Hopital E. Herriot, Place d'Arsonval, 69437, Lyon cédex 03, France.
| | | |
Collapse
|
91
|
Periostin: a putative mediator involved in tumour resistance to anti-angiogenic therapy? Cell Biol Int 2012; 35:1085-8. [PMID: 21999314 DOI: 10.1042/cbi20110171] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Despite advances in the development of anti-angiogenic agents for cancer treatment, the increase in the survival duration of cancer patients is still rather modest. One major obstacle in anti-angiogenic therapy is the emergence of drug resistance. Understanding the molecular mechanisms that enable a tumour to evade anti-angiogenic treatment is valuable to improve therapeutic efficacy. Targeting blood supply usually causes hypoxic responses of tumours that trigger a series of adaptive changes leading to a resistant phenotype. Periostin, a secreted ECM (extracellular matrix) protein, is mainly produced by CAFs (cancer-associated fibroblasts) on hypoxic stress. As CAFs have been casually linked to tumour resistance to angiogenesis blockade and periostin can influence many aspects of tumour biology, we hypothesized that periostin might be a crucial mediator involved anti-angiogenic resistance in cancer treatment. This hypothesis is indirectly supported by the following facts: (a) high levels of periostin promote tumour angiogenesis; (b) periostin improves cancer cell survival under hypoxic conditions; and (c) genetic modulation of periostin induces EMT (epithelial-mesenchymal transition) and enhances cancer cell invasion and metastasis, which represents an escape mechanism from anticancer treatment. Testing and confirmation of this hypothesis will give more insight into the resistance mechanisms and provide the rationale for improvement of therapeutic outcome of anti-angiogenic therapy.
Collapse
|
92
|
Cho YH, Cha MJ, Song BW, Kim IK, Song H, Chang W, Lim S, Ham O, Lee SY, Choi E, Kwon HM, Hwang KC. Enhancement of MSC adhesion and therapeutic efficiency in ischemic heart using lentivirus delivery with periostin. Biomaterials 2012; 33:1376-1385. [PMID: 22112759 DOI: 10.1016/j.biomaterials.2011.10.078] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 10/27/2011] [Indexed: 02/03/2023]
Abstract
Many approaches have shown beneficial effects of modified mesenchymal stem cells (MSCs) for treatment of infarcted myocardium, but have primarily focused on enhancing the survival of transplanted MSCs. Here, we show the dual benefits of periostin-overexpressing MSCs (p-MSCs) for infarcted myocardium. P-MSCs led to the marked histological and functional recovery of infarcted myocardium by enhancing survival of MSCs and directly preventing apoptosis of cardiomyocytes. Survival of p-MSCs themselves and cardiomyocytes co-cultured with p-MSCs or treated with the conditioned media from p-MSCs was significantly increased under hypoxic conditions. Decreases in adhesion-related integrins were reversed in cardiomyocytes co-cultured with p-MSCs, followed by increases in p-PI3K and Akt, indicating that periostin activates the PI3K pathway through adhesion-related integrins. When p-MSCs were injected into myocardial infarcted rats, histological pathology and cardiac function were significantly improved compared to MSC-injected controls. Thus, periostin might be a new target of therapeutic treatments using MSCs as carriers for infarcted myocardium.
Collapse
Affiliation(s)
- Yun-Hyeong Cho
- Cardiology Division, Myongji Hospital, Kwandong University College of Medicine, Goyangsi, Gyeonggido, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Elliott CG, Wang J, Guo X, Xu SW, Eastwood M, Guan J, Leask A, Conway SJ, Hamilton DW. Periostin modulates myofibroblast differentiation during full-thickness cutaneous wound repair. J Cell Sci 2012; 125:121-32. [PMID: 22266908 DOI: 10.1242/jcs.087841] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The matricellular protein periostin is expressed in the skin. Although periostin has been hypothesized to contribute to dermal homeostasis and repair, this has not been directly tested. To assess the contribution of periostin to dermal healing, 6 mm full-thickness excisional wounds were created in the skin of periostin-knockout and wild-type, sex-matched control mice. In wild-type mice, periostin was potently induced 5-7 days after wounding. In the absence of periostin, day 7 wounds showed a significant reduction in myofibroblasts, as visualized by expression of α-smooth muscle actin (α-SMA) within the granulation tissue. Delivery of recombinant human periostin by electrospun collagen scaffolds restored α-SMA expression. Isolated wild-type and knockout dermal fibroblasts did not differ in in vitro assays of adhesion or migration; however, in 3D culture, periostin-knockout fibroblasts showed a significantly reduced ability to contract a collagen matrix, and adopted a dendritic phenotype. Recombinant periostin restored the defects in cell morphology and matrix contraction displayed by periostin-deficient fibroblasts in a manner that was sensitive to a neutralizing anti-β1-integrin and to the FAK and Src inhibitor PP2. We propose that periostin promotes wound contraction by facilitating myofibroblast differentiation and contraction.
Collapse
Affiliation(s)
- Christopher G Elliott
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Lopez-Guisa JM, Powers C, File D, Cochrane E, Jimenez N, Debley JS. Airway epithelial cells from asthmatic children differentially express proremodeling factors. J Allergy Clin Immunol 2012; 129:990-7.e6. [PMID: 22227417 DOI: 10.1016/j.jaci.2011.11.035] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 11/17/2011] [Accepted: 11/22/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND The airway epithelium can express factors that drive subepithelial airway remodeling. TGF-β2, vascular epithelial growth factor (VEGF), a disintegrin and metalloprotease 33 (ADAM33), and periostin are hypothesized to be involved in subepithelial remodeling and are overexpressed in adult asthmatic airways. Epidemiologic data suggest that lung function deficits in asthmatic patients are acquired in childhood. OBJECTIVES We sought to determine whether airway epithelial cells (AECs) from asthmatic children differentially express TGF-β2, VEGF, ADAM33, or periostin compared with cells from atopic nonasthmatic and healthy children intrinsically or in response to IL-4/IL-13 stimulation. METHODS Bronchial and nasal epithelial cells were obtained from brushings from well-characterized asthmatic (n = 16), atopic nonasthmatic (n = 9), and healthy (n = 15) children after achievement of anesthesia for elective procedures. After differentiation at an air-liquid interface (ALI) for 3 weeks, conditioned media were sampled and RNA was extracted from unstimulated and IL-4/IL-13-stimulated cultures. TGF-β2 and VEGF levels were measured with ELISA. ADAM33 and periostin expression was assessed by using real-time PCR. RESULTS TGF-β2 and VEGF production was significantly greater in bronchial and nasal ALI cultures from asthmatic children than in cultures from atopic nonasthmatic and healthy children. TGF-β2 levels increased significantly in asthmatic cultures after IL-4/IL-13 stimulation. Within-subject correlation between nasal and bronchial ALI production of TGF-β2 (r = 0.64, P = .001) and VEGF (r = 0.73, P < .001) was good. Periostin expression was 3.7-fold higher in bronchial cells (P < .001) and 3.9-fold higher in nasal cells (P < .004) from asthmatic children than in cells from atopic nonasthmatic or healthy children. ADAM33 was not differentially expressed by AECs from asthmatic patients compared with that from cells from atopic nonasthmatic or healthy children. CONCLUSION AECs from asthmatic children differentially express TGF-β2, VEGF, and periostin compared with cells from atopic nonasthmatic and healthy children. Nasal epithelial cells might be a suitable surrogate for bronchial cells that could facilitate investigation of the airway epithelium in future longitudinal pediatric studies.
Collapse
Affiliation(s)
- Jesus M Lopez-Guisa
- Center for Tissue and Cell Sciences, Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | | | | | | |
Collapse
|
95
|
Sugi Y, Kern MJ, Markwald RR, Burnside JL. Periostin Expression is Altered in Aortic Valves in Smad6 Mutant Mice. ACTA ACUST UNITED AC 2012; 1. [PMID: 25383261 PMCID: PMC4224111 DOI: 10.4172/2167-0897.1000101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Smad6 is known to predominantly inhibit BMP signaling by negatively regulating the BMP signaling process. Therefore, Smad6 mutation potentially provides an important genetic model for investigating the role of BMP signaling in vivo. Periostin is a 90-kDA secreted extracellular matrix (ECM) protein and implicated in cardiac valve progenitor cell differentiation, maturation and adult aortic valve calcification in mice. We have previously reported periostin expression patterns during AV valve development in mice. Because periostin can play critical roles in aortic valve interstitial cell differentiation and can be correlated with adult valve disease pathogenesis, in the present study we specifically focused on periostin expression during outflow tract (OT) development and its expression within the adult mouse valves. We previously reported that periostin expression in valve progenitor cells was altered by exogenously adding BMP-2 in culture. In this study, we investigated whether expression of periostin and other valvulogenic ECM proteins was altered in Smad6-mutant newborn mice in vivo. Periostin protein was localized within OT during embryonic development in mice. At embryonic day (ED) 13.5, robust periostin expression was detected within the developing pulmonary trunk and developing pulmonary and aortic valves. Periostin expression remained intense in pulmonary and aortic valves up to the adult stage. Our immunohistochemical and immunointensity analyses revealed that periostin expression was significantly reduced in the aortic valves in Smad6−/− neonatal hearts. Versican expression was also significantly reduced in Smad6−/− aortic valves, whereas, hyaluronan deposition was not significantly altered in the Smad6−/− neonatal valves. Expression of periostin and versican was less prominently affected in AV valves compared to the aortic valves, suggesting that a cell lineage/origin-dependent response to regulatory molecules may play a critical role in valve interstitial cell development and ECM protein expression.
Collapse
Affiliation(s)
- Yukiko Sugi
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| | - Michael J Kern
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| | - Roger R Markwald
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| | - Jessica L Burnside
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| |
Collapse
|
96
|
Abstract
Abstract
The formation and remodeling of the embryonic valves is a complex and dynamic process that occurs within a constantly changing hemodynamic environment. Defects in embryonic and fetal valve remodeling are the leading cause of congenital heart defects, yet very little is known about how fibrous leaflet tissue is created from amorphous gelatinous masses called cushions. Microenvironmental cues such as mechanical forces and extracellular matrix composition play major roles in cell differentiation, but almost all research efforts in valvulogenesis center around genetics and molecular approaches. This review summarizes what is known about the dynamic mechanical and extracellular matrix microenvironment of the atrioventricular and semilunar valves during embryonic development and their possible guidance roles. A variety of new computational tools and sophisticated experimental techniques are progressing that enable precise microenvironmental alterations that are critical to complement genetic gain and loss of function approaches. Studies at the interface of mechanical and genetic signaling in embryonic valvulogenesis will likely pay significant dividends, not only in terms of increasing our mechanistic understanding, but also lead to the development of novel therapeutic strategies for patients with congenital valve abnormalities.
Collapse
Affiliation(s)
| | - Jonathan T. Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
97
|
Tholen S, Biniossek ML, Geßler AL, Müller S, Weißer J, Kizhakkedathu JN, Reinheckel T, Schilling O. Contribution of cathepsin L to secretome composition and cleavage pattern of mouse embryonic fibroblasts. Biol Chem 2011; 392:961-71. [DOI: 10.1515/bc.2011.162] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The endolysosomal cysteine endoprotease cathepsin L is secreted from cells in a variety of pathological conditions such as cancer and arthritis. We compared the secretome composition and extracellular proteolytic cleavage events in cell supernatants of cathepsin L-deficient and wild-type mouse embryonic fibroblasts (MEFs). Quantitative proteomic comparison of cell conditioned media indicated that cathepsin L deficiency affects, albeit in a limited manner, the abundances of extracellular matrix (ECM) components, signaling proteins, and further proteases as well as endogenous protease inhibitors. Immunodetection corroborated that cathepsin L deficiency results in decreased abundance of the ECM protein periostin and elevated abundance of matrix metalloprotease (MMP)-2. While mRNA levels of MMP-2 were not affected by cathepsin L ablation, periostin mRNA levels were reduced, potentially indicating a downstream effect. To characterize cathepsin L contribution to extracellular proteolysis, we performed terminal amine isotopic labeling of substrates (TAILS), an N-terminomic technique for the identification and quantification of native and proteolytically generated protein N-termini. TAILS identified >1500 protein N-termini. Cathepsin L deficiency predominantly reduced the magnitude of collagenous cleavage sites C-terminal to a proline residue. This contradicts cathepsin L active site specificity and indicates altered activity of further proteases as a result of cathepsin L ablation.
Collapse
|
98
|
Tholen S, Biniossek ML, Gessler AL, Müller S, Weisser J, Kizhakkedathu JN, Reinheckel T, Schilling O. Contribution of cathepsin L to secretome composition and cleavage pattern of mouse embryonic fibroblasts. Biol Chem 2011. [PMID: 21972973 DOI: 10.1515/bc-2011-162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The endolysosomal cysteine endoprotease cathepsin L is secreted from cells in a variety of pathological conditions such as cancer and arthritis. We compared the secretome composition and extracellular proteolytic cleavage events in cell supernatants of cathepsin L-deficient and wild-type mouse embryonic fibroblasts (MEFs). Quantitative proteomic comparison of cell conditioned media indicated that cathepsin L deficiency affects, albeit in a limited manner, the abundances of extracellular matrix (ECM) components, signaling proteins, and further proteases as well as endogenous protease inhibitors. Immunodetection corroborated that cathepsin L deficiency results in decreased abundance of the ECM protein periostin and elevated abundance of matrix metalloprotease (MMP)-2. While mRNA levels of MMP-2 were not affected by cathepsin L ablation, periostin mRNA levels were reduced, potentially indicating a downstream effect. To characterize cathepsin L contribution to extracellular proteolysis, we performed terminal amine isotopic labeling of substrates (TAILS), an N-terminomic technique for the identification and quantification of native and proteolytically generated protein N-termini. TAILS identified >1500 protein N-termini. Cathepsin L deficiency predominantly reduced the magnitude of collagenous cleavage sites C-terminal to a proline residue. This contradicts cathepsin L active site specificity and indicates altered activity of further proteases as a result of cathepsin L ablation.
Collapse
Affiliation(s)
- Stefan Tholen
- Institute for Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Wang F, Song Y, Jiang Y, Yang C, Ding Z. Associations among periostin gene polymorphisms, clinical parameters and heart failure: a case-control study in 1104 Chinese individuals. J Cardiovasc Med (Hagerstown) 2011; 12:469-74. [PMID: 21558869 DOI: 10.2459/jcm.0b013e328347e48c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS To determine the associations among periostin gene polymorphisms, clinical parameters and heart failure in a Chinese population. METHODS In total, 464 patients with heart failure and 640 control individuals were included in this study. rs3829365 and rs1028728 were genotyped through PCR and restriction fragment length polymorphism. Multivariate logistic regression was employed to analyze the independent strength of association among clinical parameters, genotypes and heart failure. RESULTS rs3829365 was associated with heart failure (P = 0.043), whereas rs1028728 was not (P = 0.188). After adjusting for age, sex, hypertension, diabetes mellitus, smoking and hypertriglyceridemia in multivariate logistic regression, we found that CG or GG of rs3829365 [P = 0.015, odds ratio (OR) = 1.88] was an independent risk factor for heart failure and that CG or GG of rs3829365 (P = 0.039, OR = 0.94) and AT or TT of rs1028728 (P = 0.011, OR = 0.68) were significantly protective factors for heart failure in patients who smoked and in patients with hypertriglyceridemia, respectively. Moreover, heart failure in patients with CG or GG genotype of rs3829365 tended to be more severe than in those with CC genotype. CONCLUSION These findings suggest that rs3829365 of the periostin gene may be helpful to determine the susceptibility to, and severity of, heart failure. The interactions between rs3829365 and smoking and between rs1028728 and hypertriglyceridemia warrant further investigations for underlying mechanisms.
Collapse
Affiliation(s)
- Fangfang Wang
- Department of Cardiovascular Disease, Changzhou No. 2 People's Hospital, Affiliated Nanjing Medical University, Changzhou, Jiangsu Province, China
| | | | | | | | | |
Collapse
|
100
|
PERIOSTIN regulates MMP-2 expression via the αvβ3 integrin/ERK pathway in human periodontal ligament cells. Arch Oral Biol 2011; 57:52-9. [PMID: 21885032 DOI: 10.1016/j.archoralbio.2011.07.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/14/2011] [Accepted: 07/28/2011] [Indexed: 01/05/2023]
Abstract
OBJECTIVE During orthodontic tooth movement, activation of the vascular system in the compressed periodontal ligament (PDL), which becomes hypoxic, is essential for periodontal tissue remodelling. PERIOSTIN, an extracellular matrix protein, is expressed in PDL and its concentration is increased on the compressive side during orthodontic tooth movement. PERIOSTIN promotes angiogenesis through upregulation of matrix metalloproteinase (MMP)-2, which has been shown to be expressed via αvβ3 integrin/extracellular signal-related kinase (ERK) signalling pathway and vascular endothelial growth factor (VEGF). Therefore, we hypothesized that hypoxia-induced PERIOSTIN promotes MMP-2 expression via αvβ3 integrin/ERK signalling and VEGF in PDL cells. METHODS Human PDL cells were cultured in condition medium containing desferrioxamine (DFO) to mimic hypoxia. The total RNA, cell lysates or supernatant were collected, and MMP2 and VEGF expression, PERIOSTIN expression and ERK phosphorylation, and MMP-2 activity were analysed by real-time RT-PCR, western blot analysis, and zymography, respectively. A recombinant human PERIOSTIN or PERIOSTIN siRNA was applied to the cells, then the total RNA was extracted to measure MMP-2 and VEGF expression. The cells were treated with αvβ3 integrin-blocking antibody or ERK inhibitor followed by PERIOSTIN stimulation. MMP-2 expression was measured by real-time RT-PCR. RESULTS PERIOSTIN was upregulated in a time-dependent manner in human PDL cells treated with DFO, a chemical hypoxia mimic. MMP-2 and VEGF expression, and MMP-2 activity were increased by DFO or PERIOSTIN treatment, and decreased by PERIOSTIN silencing. PERIOSTIN treatment also induced ERK phosphorylation, and PERIOSTIN-induced MMP-2 was reduced by αvβ3 integrin-blocking antibody or ERK inhibitor. CONCLUSION These data suggest that PERIOSTIN upregulates MMP-2 expression via the αvβ3 integrin/ERK signalling pathway and VEGF expression in human PDL cells.
Collapse
|