51
|
Decellularized tongue tissue as an in vitro model for studying tongue cancer and tongue regeneration. Acta Biomater 2017; 58:122-135. [PMID: 28600128 DOI: 10.1016/j.actbio.2017.05.062] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/25/2017] [Accepted: 05/25/2017] [Indexed: 12/15/2022]
Abstract
The decellularization of tissues or organs provides an efficient strategy for preparing functional scaffolds for tissue engineering. The microstructures of native extracellular matrices and biochemical compositions retained in the decellularized matrices provide tissue-specific microenvironments for anchoring cells. Here, we report the tongue extracellular matrix (TEM), which showed favorable cytocompatibility for normal tongue-derived cells and tongue squamous cell carcinoma (TSCC) cells under static or stirring culture conditions. Our results show that TEM retained tongue-specific integrated microstructures and abundant matrix components, which offer mechanical support and spatial signals for regulating cell behavior and function. Reconstructed TSCC by TEM presented characteristics resembling clinical TSCC histopathology, suggesting the possibility for TSCC research. In addition, TEM might be capable of guiding tongue-derived cells to the niche, benefiting cell survival, proliferation and differentiation. STATEMENT OF SIGNIFICANCE In this study, we prepared decellularized tongue extracellular matrix (TEM) and evaluated the possibility for tongue squamous cell carcinoma (TSCC) research and tongue regeneration. TEM has six irreplaceable advantages: (1) tongue-specific intricate structures of TEM, which offer mechanical support for the cells; (2) abundant matrix components and spatial signals benefiting for cell attachment, survival, differentiation, and long-term viability of the highly functional phenotypes of tongue cells or TSCC cells; (3) reconstructed TSCC by TEM exhibited tumor heterogeneity, extremely resembling clinical TSCC histopathology; (4) ideal model to evaluate TSCC movement mode; (5) guiding tongue-derived cells to the site-appropriate niche; and (6) the possibility for static or stirred cell culture. These properties might be considered in TSCC research or tongue regeneration.
Collapse
|
52
|
Wu T, Economopoulos KP, Ott HC. Engineering Bioartificial Lungs for Transplantation. CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0082-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
53
|
Scarritt ME, Pashos NC, Motherwell JM, Eagle ZR, Burkett BJ, Gregory AN, Mostany R, Weiss DJ, Alvarez DF, Bunnell BA. Re-endothelialization of rat lung scaffolds through passive, gravity-driven seeding of segment-specific pulmonary endothelial cells. J Tissue Eng Regen Med 2017; 12:e786-e806. [PMID: 27943597 DOI: 10.1002/term.2382] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 09/27/2016] [Accepted: 12/06/2016] [Indexed: 01/05/2023]
Abstract
Effective re-endothelialization is critical for the use of decellularized scaffolds for ex vivo lung engineering. Current approaches yield insufficiently re-endothelialized scaffolds that haemorrhage and become thrombogenic upon implantation. Herein, gravity-driven seeding coupled with bioreactor culture facilitated widespread distribution and engraftment of endothelial cells throughout rat lung scaffolds. Initially, human umbilical vein endothelial cells were seeded into the pulmonary artery by either gravity-driven, variable flow perfusion seeding or pump-driven, pulsatile flow perfusion seeding. Gravity seeding evenly distributed cells and supported cell survival and re-lining of the vascular walls while perfusion pump-driven seeding led to increased cell fragmentation and death. Using gravity seeding, rat pulmonary artery endothelial cells and rat pulmonary vein endothelial cells attached in intermediate and large vessels, while rat pulmonary microvascular endothelial cells deposited mostly in microvessels. Combination seeding of these cells led to positive vascular endothelial cadherin staining. In addition, combination seeding improved barrier function as assessed by serum albumin extravasation; however, leakage was observed in the distal portions of the re-endothelialized tissue suggesting that recellularization of the alveoli is necessary to complete barrier function of the capillary-alveolar network. Overall, these data indicate that vascular recellularization of rat lung scaffolds is achieved through gravity seeding. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Michelle E Scarritt
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nicholas C Pashos
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA.,Bioinnovation PhD Program, Tulane University, New Orleans, LA, USA
| | | | - Zachary R Eagle
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Brian J Burkett
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ashley N Gregory
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Diego F Alvarez
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
54
|
Culture of iPSCs Derived Pancreatic β-Like Cells In Vitro Using Decellularized Pancreatic Scaffolds: A Preliminary Trial. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4276928. [PMID: 28480220 PMCID: PMC5396430 DOI: 10.1155/2017/4276928] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/30/2016] [Accepted: 01/19/2017] [Indexed: 12/11/2022]
Abstract
Diabetes mellitus is a disease which has affected 415 million patients in 2015. In an effort to replace the significant demands on transplantation and morbidity associated with transplantation, the production of β-like cells differentiated from induced pluripotent stem cells (iPSCs) was evaluated. This approach is associated with promising decellularized scaffolds with natural extracellular matrix (ECM) and ideal cubic environment that will promote cell growth in vivo. Our efforts focused on combining decellularized rat pancreatic scaffolds with mouse GFP+-iPSCs-derived pancreatic β-like cells, to evaluate whether decellularized scaffolds could facilitate the growth and function of β-like cells. β-like cells were differentiated from GFP+-iPSCs and evaluated via cultivating in the dynamic circulation perfusion device. Our results demonstrated that decellularized pancreatic scaffolds display favorable biochemical properties. Furthermore, not only could the scaffolds support the survival of β-like cells, but they also accelerated the expression of the insulin as compared to plate-based cell culture. In conclusion, these results suggest that decellularized pancreatic scaffolds could provide a suitable platform for cellular activities of β-like cells including survival and insulin secretion. This study provides preliminary support for regenerating insulin-secreting organs from the decellularized scaffolds combined with iPSCs derived β-like cells as a potential clinical application.
Collapse
|
55
|
Modification of Rat Lung Decellularization Protocol Based on Dynamic Conductometry of Working Solution. Bull Exp Biol Med 2017; 162:703-706. [DOI: 10.1007/s10517-017-3692-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Indexed: 12/14/2022]
|
56
|
Simões IN, Vale P, Soker S, Atala A, Keller D, Noiva R, Carvalho S, Peleteiro C, Cabral JMS, Eberli D, da Silva CL, Baptista PM. Acellular Urethra Bioscaffold: Decellularization of Whole Urethras for Tissue Engineering Applications. Sci Rep 2017; 7:41934. [PMID: 28165009 PMCID: PMC5292742 DOI: 10.1038/srep41934] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/03/2017] [Indexed: 11/16/2022] Open
Abstract
Patients with stress urinary incontinence mainly suffer from malfunction of the urethra closure mechanism. We established the decellularization of porcine urethras to produce acellular urethra bioscaffolds for future tissue engineering applications, using bioscaffolds or bioscaffold-derived soluble products. Cellular removal was evaluated by H&E, DAPI and DNA quantification. The presence of specific ECM proteins was assessed through immunofluorescence staining and colorimetric assay kits. Human skeletal muscle myoblasts, muscle progenitor cells and adipose-derived stromal vascular fractions were used to evaluate the recellularization of the acellular urethra bioscaffolds. The mechanochemical decellularization system removed ~93% of tissue's DNA, generally preserving ECM's components and microarchitecture. Recellularization was achieved, though methodological advances are required regarding cell seeding strategies and functional assessment. Through microdissection and partial digestion, different urethra ECM-derived coating substrates were formulated (i.e. containing smooth or skeletal muscle ECM) and used to culture MPCs in vitro. The skeletal muscle ECM substrates enhanced fiber formation leading to the expression of the main skeletal muscle-related proteins and genes, as confirmed by immunofluorescence and RT-qPCR. The described methodology produced a urethra bioscaffold that retained vital ECM proteins and was liable to cell repopulation, a crucial first step towards the generation of urethra bioscaffold-based Tissue Engineering products.
Collapse
Affiliation(s)
- Irina N. Simões
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Paulo Vale
- Serviço Urologia, Hospital Garcia de Orta, Almada, Portugal
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Daniel Keller
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Rute Noiva
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Sandra Carvalho
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Conceição Peleteiro
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Daniel Eberli
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Cláudia L. da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro M. Baptista
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Instituto de Investigacion Sanitaria de Aragón (IIS Aragon), Zaragoza, Spain
- CIBERehd, Zaragoza, Spain
| |
Collapse
|
57
|
Uhl FE, Wagner DE, Weiss DJ. Preparation of Decellularized Lung Matrices for Cell Culture and Protein Analysis. Methods Mol Biol 2017; 1627:253-283. [PMID: 28836208 PMCID: PMC7456164 DOI: 10.1007/978-1-4939-7113-8_18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The limited available treatment options for patients with chronic lung diseases, such as fibrosis, lead to poor prognosis after diagnosis and short survival rates. An exciting new bioengineering approach utilizes de- and recellularization of lung tissue to potentially overcome donor organ shortage and immune reactions toward the received transplant. The goal of decellularization is to create a scaffold which contains the necessary framework for stability and functionality for regenerating lung tissue while removing immunomodulatory factors by removal of cells. After decellularization, the scaffold could be re-functionalized by repopulation with the patient's own stem/progenitor cells to create a fully functional organ or can be used as ex vivo models of disease. In this chapter the decellularization of lung tissue from multiple species (i.e., rodents, pigs, and humans) as well as disease states such as fibrosis is described. We discuss and describe the various quality control measures which should be used to characterize decellularized scaffolds, methods for protein analysis of the remaining scaffold, and methods for recellularization of scaffolds.
Collapse
Affiliation(s)
- Franziska E Uhl
- Department of Med-Pulmonary, College of Medicine, University of Vermont, Burlington, VT, USA.
| | - Darcy E Wagner
- Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Daniel J Weiss
- Department of Med-Pulmonary, College of Medicine, University of Vermont, Burlington, VT, USA
| |
Collapse
|
58
|
Poornejad N, Buckmiller E, Schaumann L, Wang H, Wisco J, Roeder B, Reynolds P, Cook A. Re-epithelialization of whole porcine kidneys with renal epithelial cells. J Tissue Eng 2017; 8:2041731417718809. [PMID: 28758007 PMCID: PMC5513523 DOI: 10.1177/2041731417718809] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/13/2017] [Indexed: 01/16/2023] Open
Abstract
Decellularized porcine kidneys were recellularized with renal epithelial cells by three methods: perfusion through the vasculature under high pressure, perfusion through the ureter under high pressure, or perfusion through the ureter under moderate vacuum. Histology, scanning electron microscopy, confocal microscopy, and magnetic resonance imaging were used to assess vasculature preservation and the distribution of cells throughout the kidneys. Cells were detected in the magnetic resonance imaging by labeling them with iron oxide. Perfusion of cells through the ureter under moderate vacuum (40 mmHg) produced the most uniform distribution of cells throughout the kidneys.
Collapse
Affiliation(s)
- Nafiseh Poornejad
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Evan Buckmiller
- Department of Genetics and Biotechnology, Brigham Young University, Provo, UT, USA
| | - Lara Schaumann
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Haonan Wang
- Department of Electrical Engineering, Brigham Young University, Provo, UT, USA
| | - Jonathan Wisco
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT, USA
| | - Beverly Roeder
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Paul Reynolds
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT, USA
| | - Alonzo Cook
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| |
Collapse
|
59
|
Calle EA, Hill RC, Leiby KL, Le AV, Gard AL, Madri JA, Hansen KC, Niklason LE. Targeted proteomics effectively quantifies differences between native lung and detergent-decellularized lung extracellular matrices. Acta Biomater 2016; 46:91-100. [PMID: 27693690 DOI: 10.1016/j.actbio.2016.09.043] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 08/25/2016] [Accepted: 09/28/2016] [Indexed: 12/16/2022]
Abstract
Extracellular matrix is a key component of many products in regenerative medicine. Multiple regenerative medicine products currently in the clinic are comprised of human or xenogeneic extracellular matrix. In addition, whole-organ regeneration exploits decellularized native organs as scaffolds for organotypic cell culture. However, precise understanding of the constituents of such extracellular matrix-based implants and scaffolds has sorely lagged behind their use. We present here an advanced protein extraction method using known quantities of proteotypic 13C-labeled peptides to quantify matrix proteins in native and decellularized lung tissues. Using quantitative proteomics that produce picomole-level measurements of a large number of matrix proteins, we show that a mild decellularization technique ("Triton/SDC") results in near-native retention of laminins, proteoglycans, and other basement membrane and ECM-associated proteins. Retention of these biologically important glycoproteins and proteoglycans is quantified to be up to 27-fold higher in gently-decellularized lung scaffolds compared to scaffolds generated using a previously published decellularization regimen. Cells seeded onto this new decellularized matrix also proliferate robustly, showing positive staining for proliferating cell nuclear antigen (PCNA). The high fidelity of the gently decellularized scaffold as compared to the original lung extracellular matrix represents an important step forward in the ultimate recapitulation of whole organs using tissue-engineering techniques. This method of ECM and scaffold protein analysis allows for better understanding, and ultimately quality control, of matrices that are used for tissue engineering and human implantation. These results should advance regenerative medicine in general, and whole organ regeneration in particular. STATEMENT OF SIGNIFICANCE The extracellular matrix (ECM) in large part defines the biochemical and mechanical properties of tissues and organs; these inherent cues make acellular ECM scaffolds potent substrates for tissue regeneration. As such, they are increasingly prevalent in the clinic and the laboratory. However, the exact composition of these scaffolds has been difficult to ascertain. This paper uses targeted proteomics to definitively quantify 71 proteins present in acellular lung ECM scaffolds. We use this technique to compare two decellularization methods and demonstrate superior retention of ECM proteins important for cell adhesion, migration, proliferation, and differentiation in scaffolds treated with low-concentration detergent solutions. In the long term, the ability to acquire quantitative biochemical data about biological substrates will facilitate the rational design of engineered tissues and organs based on precise cell-matrix interactions.
Collapse
|
60
|
Hun M, Barsanti M, Wong K, Ramshaw J, Werkmeister J, Chidgey AP. Native thymic extracellular matrix improves in vivo thymic organoid T cell output, and drives in vitro thymic epithelial cell differentiation. Biomaterials 2016; 118:1-15. [PMID: 27940379 DOI: 10.1016/j.biomaterials.2016.11.054] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 12/22/2022]
Abstract
Although the thymus is a primary lymphoid organ, its function is compromised by an age-induced loss of resident epithelial cells, which results in reduced naïve T cell output. This has important implications for immune recovery in aged and elderly patients following damage from cytoablative therapies. As thymic architecture plays a crucial role in naïve T cell development, a tissue specific scaffold that provides essential supporting matrix may assist in stem cell-based thymus regeneration to recreate complex organoids. Here we investigate thymus decellularization approaches that preserve major extracellular matrix components and support thymic epithelial cells for the generation of a functional thymic microenvironment with improved T cell output. We also established an in vitro, serum-free culture system that both maintains a progenitor thymic epithelial cell pool and drives their differentiation in the presence of decellularized thymic matrix. This approach enables further dissection of key cellular and niche components involved in thymic epithelial stem cell maintenance and T cell production.
Collapse
Affiliation(s)
- Michael Hun
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Level 3, 15 Innovation Walk, Monash University, Clayton, Victoria 3800, Australia
| | - Marco Barsanti
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Level 3, 15 Innovation Walk, Monash University, Clayton, Victoria 3800, Australia
| | - Kahlia Wong
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Level 3, 15 Innovation Walk, Monash University, Clayton, Victoria 3800, Australia
| | - John Ramshaw
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | | | - Ann P Chidgey
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Level 3, 15 Innovation Walk, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
61
|
Nonaka PN, Uriarte JJ, Campillo N, Oliveira VR, Navajas D, Farré R. Lung bioengineering: physical stimuli and stem/progenitor cell biology interplay towards biofabricating a functional organ. Respir Res 2016; 17:161. [PMID: 27894293 PMCID: PMC5126992 DOI: 10.1186/s12931-016-0477-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/22/2016] [Indexed: 01/18/2023] Open
Abstract
A current approach to obtain bioengineered lungs as a future alternative for transplantation is based on seeding stem cells on decellularized lung scaffolds. A fundamental question to be solved in this approach is how to drive stem cell differentiation onto the different lung cell phenotypes. Whereas the use of soluble factors as agents to modulate the fate of stem cells was established from an early stage of the research with this type of cells, it took longer to recognize that the physical microenvironment locally sensed by stem cells (e.g. substrate stiffness, 3D architecture, cyclic stretch, shear stress, air-liquid interface, oxygenation gradient) also contributes to their differentiation. The potential role played by physical stimuli would be particularly relevant in lung bioengineering since cells within the organ are physiologically subjected to two main stimuli required to facilitate efficient gas exchange: air ventilation and blood perfusion across the organ. The present review focuses on describing how the cell mechanical microenvironment can modulate stem cell differentiation and how these stimuli could be incorporated into lung bioreactors for optimizing organ bioengineering.
Collapse
Affiliation(s)
- Paula N Nonaka
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Juan J Uriarte
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Noelia Campillo
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Vinicius R Oliveira
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.,CIBER Enfermedades Respiratorias, Madrid, Spain.,Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain. .,CIBER Enfermedades Respiratorias, Madrid, Spain. .,Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain.
| |
Collapse
|
62
|
Agmon G, Christman KL. Controlling stem cell behavior with decellularized extracellular matrix scaffolds. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2016; 20:193-201. [PMID: 27524932 PMCID: PMC4979580 DOI: 10.1016/j.cossms.2016.02.001] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Decellularized tissues have become a common regenerative medicine platform with multiple materials being researched in academic laboratories, tested in animal studies, and used clinically. Ideally, when a tissue is decellularized the native cell niche is maintained with many of the structural and biochemical cues that naturally interact with the cells of that particular tissue. This makes decellularized tissue materials an excellent platform for providing cells with the signals needed to initiate and maintain differentiation into tissue-specific lineages. The extracellular matrix (ECM) that remains after the decellularization process contains the components of a tissue specific microenvironment that is not possible to create synthetically. The ECM of each tissue has a different composition and structure and therefore has unique properties and potential for affecting cell behavior. This review describes the common methods for preparing decellularized tissue materials and the effects that decellularized materials from different tissues have on cell phenotype.
Collapse
|
63
|
Platz J, Bonenfant NR, Uhl FE, Coffey AL, McKnight T, Parsons C, Sokocevic D, Borg ZD, Lam YW, Deng B, Fields JG, DeSarno M, Loi R, Hoffman AM, Bianchi J, Dacken B, Petersen T, Wagner DE, Weiss DJ. Comparative Decellularization and Recellularization of Wild-Type and Alpha 1,3 Galactosyltransferase Knockout Pig Lungs: A Model for Ex Vivo Xenogeneic Lung Bioengineering and Transplantation. Tissue Eng Part C Methods 2016; 22:725-39. [PMID: 27310581 DOI: 10.1089/ten.tec.2016.0109] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND A novel potential approach for lung transplantation could be to utilize xenogeneic decellularized pig lung scaffolds that are recellularized with human lung cells. However, pig tissues express several immunogenic proteins, notably galactosylated cell surface glycoproteins resulting from alpha 1,3 galactosyltransferase (α-gal) activity, that could conceivably prevent effective use. Use of lungs from α-gal knock out (α-gal KO) pigs presents a potential alternative and thus comparative de- and recellularization of wild-type and α-gal KO pig lungs was assessed. METHODS Decellularized lungs were compared by histologic, immunohistochemical, and mass spectrometric techniques. Recellularization was assessed following compartmental inoculation of human lung bronchial epithelial cells, human lung fibroblasts, human bone marrow-derived mesenchymal stromal cells (all via airway inoculation), and human pulmonary vascular endothelial cells (CBF) (vascular inoculation). RESULTS No obvious differences in histologic structure was observed but an approximate 25% difference in retention of residual proteins was determined between decellularized wild-type and α-gal KO pig lungs, including retention of α-galactosylated epitopes in acellular wild-type pig lungs. However, robust initial recellularization and subsequent growth and proliferation was observed for all cell types with no obvious differences between cells seeded into wild-type versus α-gal KO lungs. CONCLUSION These proof of concept studies demonstrate that decellularized wild-type and α-gal KO pig lungs can be comparably decellularized and comparably support initial growth of human lung cells, despite some differences in retained proteins. α-Gal KO pig lungs are a suitable platform for further studies of xenogeneic lung regeneration.
Collapse
Affiliation(s)
- Joseph Platz
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Nicholas R Bonenfant
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Franziska E Uhl
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Amy L Coffey
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Tristan McKnight
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Charles Parsons
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Dino Sokocevic
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Zachary D Borg
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Ying-Wai Lam
- 2 Department of Biology and VGN Proteomics Facility, University of Vermont College of Arts and Sciences , Burlington, Vermont
| | - Bin Deng
- 2 Department of Biology and VGN Proteomics Facility, University of Vermont College of Arts and Sciences , Burlington, Vermont
| | - Julia G Fields
- 2 Department of Biology and VGN Proteomics Facility, University of Vermont College of Arts and Sciences , Burlington, Vermont
| | - Michael DeSarno
- 3 Biostatistics Unit, University of Vermont College of Medicine , Burlington, Vermont
| | - Roberto Loi
- 4 Department of Biomedical Sciences, University of Cagliari , Cagliari, Italy
| | - Andrew M Hoffman
- 5 Department of Clinical Sciences, Tufts University , Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| | | | | | - Thomas Petersen
- 8 United Therapeutics Corp., Research Triangle Park , Durham, North Carolina
| | - Darcy E Wagner
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont.,9 Comprehensive Pneumonology Center, Helmholtz Center Munich, Ludwig Maximilians University Munich , Munich, Germany
| | - Daniel J Weiss
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| |
Collapse
|
64
|
Zhao L, Chen HL, Xie LQ, Wang M, Li XF, Feng ZW, Li M. Modification of decellularized vascular scaffold with conditioned medium to enhance cell reseeding. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2016; 27:1115-25. [DOI: 10.1080/09205063.2016.1183333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Liang Zhao
- College of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, PR China
- Key Laboratory of Biomedical Materials, Xinxiang Medical University, Xinxiang, PR China
| | - Hong-Li Chen
- College of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, PR China
- Key Laboratory of Biomedical Materials, Xinxiang Medical University, Xinxiang, PR China
| | - Li-Qin Xie
- College of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, PR China
- Key Laboratory of Biomedical Materials, Xinxiang Medical University, Xinxiang, PR China
| | - Mian Wang
- College of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, PR China
- Key Laboratory of Biomedical Materials, Xinxiang Medical University, Xinxiang, PR China
| | - Xia-Fei Li
- Basic Medical College, Xinxiang Medical University, Xinxiang, PR China
| | - Zhi-Wei Feng
- College of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, PR China
- Key Laboratory of Biomedical Materials, Xinxiang Medical University, Xinxiang, PR China
| | - Min Li
- College of Life Sciences, Fujian Normal University, Fuzhou, PR China
| |
Collapse
|
65
|
|
66
|
Pouliot RA, Link PA, Mikhaiel NS, Schneck MB, Valentine MS, Kamga Gninzeko FJ, Herbert JA, Sakagami M, Heise RL. Development and characterization of a naturally derived lung extracellular matrix hydrogel. J Biomed Mater Res A 2016; 104:1922-35. [PMID: 27012815 DOI: 10.1002/jbm.a.35726] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 03/10/2016] [Accepted: 03/22/2016] [Indexed: 01/15/2023]
Abstract
The complexity and rapid clearance mechanisms of lung tissue make it difficult to develop effective treatments for many chronic pathologies. We are investigating lung derived extracellular matrix (ECM) hydrogels as a novel approach for delivery of cellular therapies to the pulmonary system. The main objectives of this study include effective decellularization of porcine lung tissue, development of a hydrogel from the porcine ECM, and characterization of the material's composition, mechanical properties, and ability to support cellular growth. Our evaluation of the decellularized tissue indicated successful removal of cellular material and immunogenic remnants in the ECM. The self-assembly of the lung ECM hydrogel was rapid, reaching maximum modulus values within 3 min at 37°C. Rheological characterization showed the lung ECM hydrogel to have a concentration dependent storage modulus between 15 and 60 Pa. The purpose of this study was to evaluate our novel ECM derived hydrogel and measure its ability to support 3D culture of MSCs in vitro and in vivo delivery of MSCs. Our in vitro experiments using human mesenchymal stem cells demonstrated our novel ECM hydrogel's ability to enhance cellular attachment and viability. Our in vivo experiments demonstrated that rat MSC delivery in pre-gel solution significantly increased cell retention in the lung over 24 h in an emphysema rat model. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1922-1935, 2016.
Collapse
Affiliation(s)
- Robert A Pouliot
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Patrick A Link
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Nabil S Mikhaiel
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Matthew B Schneck
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Michael S Valentine
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | | | - Joseph A Herbert
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Masahiro Sakagami
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia.,Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
67
|
Stabler CT, Caires LC, Mondrinos MJ, Marcinkiewicz C, Lazarovici P, Wolfson MR, Lelkes PI. Enhanced Re-Endothelialization of Decellularized Rat Lungs. Tissue Eng Part C Methods 2016; 22:439-50. [PMID: 26935764 DOI: 10.1089/ten.tec.2016.0012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Decellularized lung tissue has been recognized as a potential platform to engineer whole lung organs suitable for transplantation or for modeling a variety of lung diseases. However, many technical hurdles remain before this potential may be fully realized. Inability to efficiently re-endothelialize the pulmonary vasculature with a functional endothelium appears to be the primary cause of failure of recellularized lung scaffolds in early transplant studies. Here, we present an optimized approach for enhanced re-endothelialization of decellularized rodent lung scaffolds with rat lung microvascular endothelial cells (ECs). This was achieved by adjusting the posture of the lung to a supine position during cell seeding through the pulmonary artery. The supine position allowed for significantly more homogeneous seeding and better cell retention in the apex regions of all lobes than the traditional upright position, especially in the right upper and left lobes. Additionally, the supine position allowed for greater cell retention within large diameter vessels (proximal 100-5000 μm) than the upright position, with little to no difference in the small diameter distal vessels. EC adhesion in the proximal regions of the pulmonary vasculature in the decellularized lung was dependent on the binding of EC integrins, specifically α1β1, α2β1, and α5β1 integrins to, respectively, collagen type-I, type-IV, and fibronectin in the residual extracellular matrix. Following in vitro maturation of the seeded constructs under perfusion culture, the seeded ECs spread along the vascular wall, leading to a partial reestablishment of endothelial barrier function as inferred from a custom-designed leakage assay. Our results suggest that attention to cellular distribution within the whole organ is of paramount importance for restoring proper vascular function.
Collapse
Affiliation(s)
- Collin T Stabler
- 1 Department of Bioengineering, College of Engineering, Temple University , Philadelphia, Pennsylvania
| | - Luiz C Caires
- 1 Department of Bioengineering, College of Engineering, Temple University , Philadelphia, Pennsylvania.,2 Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, MG, Brazil
| | - Mark J Mondrinos
- 1 Department of Bioengineering, College of Engineering, Temple University , Philadelphia, Pennsylvania
| | - Cezary Marcinkiewicz
- 1 Department of Bioengineering, College of Engineering, Temple University , Philadelphia, Pennsylvania
| | - Philip Lazarovici
- 1 Department of Bioengineering, College of Engineering, Temple University , Philadelphia, Pennsylvania
| | - Marla R Wolfson
- 3 Department of Physiology, Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Peter I Lelkes
- 1 Department of Bioengineering, College of Engineering, Temple University , Philadelphia, Pennsylvania
| |
Collapse
|
68
|
Zvarova B, Uhl FE, Uriarte JJ, Borg ZD, Coffey AL, Bonenfant NR, Weiss DJ, Wagner DE. Residual Detergent Detection Method for Nondestructive Cytocompatibility Evaluation of Decellularized Whole Lung Scaffolds. Tissue Eng Part C Methods 2016; 22:418-28. [PMID: 26905643 DOI: 10.1089/ten.tec.2015.0439] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The development of reliable tissue engineering methods using decellularized cadaveric or donor lungs could potentially provide a new source of lung tissue. The vast majority of current lung decellularization protocols are detergent based and incompletely removed residual detergents may have a deleterious impact on subsequent scaffold recellularization. Detergent removal and quality control measures that rigorously and reliably confirm removal, ideally utilizing nondestructive methods, are thus critical for generating optimal acellular scaffolds suitable for potential clinical translation. Using a modified and optimized version of a methylene blue-based detergent assay, we developed a straightforward, noninvasive method for easily and reliably detecting two of the most commonly utilized anionic detergents, sodium deoxycholate (SDC) and sodium dodecyl sulfate (SDS), in lung decellularization effluents. In parallel studies, we sought to determine the threshold of detergent concentration that was cytotoxic using four different representative human cell types utilized in the study of lung recellularization: human bronchial epithelial cells, human pulmonary vascular endothelial cells (CBF12), human lung fibroblasts, and human mesenchymal stem cells. Notably, different cells have varying thresholds for either SDC or SDS-based detergent-induced cytotoxicity. These studies demonstrate the importance of reliably removing residual detergents and argue that multiple cell lines should be tested in cytocompatibility-based assessments of acellular scaffolds. The detergent detection assay presented here is a useful nondestructive tool for assessing detergent removal in potential decellularization schemes or for use as a potential endpoint in future clinical schemes, generating acellular lungs using anionic detergent-based decellularization protocols.
Collapse
Affiliation(s)
- Barbora Zvarova
- 1 Department of Medicine, University of Vermont , Burlington, Vermont
| | - Franziska E Uhl
- 1 Department of Medicine, University of Vermont , Burlington, Vermont
| | - Juan J Uriarte
- 2 Unit of Biophysics and Bioengineering, University of Barcelona, Barcelona, Spain .,3 CIBER Enfermedades Respiratorias , Madrid, Spain
| | - Zachary D Borg
- 1 Department of Medicine, University of Vermont , Burlington, Vermont
| | - Amy L Coffey
- 1 Department of Medicine, University of Vermont , Burlington, Vermont
| | | | - Daniel J Weiss
- 1 Department of Medicine, University of Vermont , Burlington, Vermont
| | - Darcy E Wagner
- 1 Department of Medicine, University of Vermont , Burlington, Vermont.,4 Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern , Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
69
|
Hoganson DM, Owens GE, Meppelink AM, Bassett EK, Bowley CM, Hinkel CJ, Finkelstein EB, Goldman SM, Vacanti JP. Decellularized extracellular matrix microparticles as a vehicle for cellular delivery in a model of anastomosis healing. J Biomed Mater Res A 2016; 104:1728-35. [PMID: 26946064 DOI: 10.1002/jbm.a.35703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/31/2015] [Accepted: 03/01/2016] [Indexed: 01/09/2023]
Abstract
Extracellular matrix (ECM) materials from animal and human sources have become important materials for soft tissue repair. Microparticles of ECM materials have increased surface area and exposed binding sites compared to sheet materials. Decellularized porcine peritoneum was mechanically dissociated into 200 µm microparticles, seeded with fibroblasts and cultured in a low gravity rotating bioreactor. The cells avidly attached and maintained excellent viability on the microparticles. When the seeded microparticles were placed in a collagen gel, the cells quickly migrated off the microparticles and through the gel. Cells from seeded microparticles migrated to and across an in vitro anastomosis model, increasing the tensile strength of the model. Cell seeded microparticles of ECM material have potential for paracrine and cellular delivery therapies when delivered in a gel carrier. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1728-1735, 2016.
Collapse
Affiliation(s)
- David M Hoganson
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Massachusetts
| | - Gwen E Owens
- Graduate Option in Biochemistry and Molecular Biophysics, California Institute of Technology, Pasadena, California
| | - Amanda M Meppelink
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Erik K Bassett
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Cameron J Hinkel
- Department of Biomedical Engineering, Washington University in St. Louis, Missouri
| | - Eric B Finkelstein
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York
| | | | - Joseph P Vacanti
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
70
|
Behavior of vascular resistance undergoing various pressure insufflation and perfusion on decellularized lungs. J Biomech 2016; 49:1230-1232. [PMID: 26949099 DOI: 10.1016/j.jbiomech.2016.02.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/17/2016] [Accepted: 02/21/2016] [Indexed: 11/22/2022]
Abstract
Bioengineering of functional lung tissue by using whole lung scaffolds has been proposed as a potential alternative for patients awaiting lung transplant. Previous studies have demonstrated that vascular resistance (Rv) could be altered to optimize the process of obtaining suitable lung scaffolds. Therefore, this work was aimed at determining how lung inflation (tracheal pressure) and perfusion (pulmonary arterial pressure) affect vascular resistance. This study was carried out using the lungs excised from 5 healthy male Sprague-Dawley rats. The trachea was cannulated and connected to a continuous positive airway pressure (CPAP) device to provide a tracheal pressure ranging from 0 to 15cmH2O. The pulmonary artery was cannulated and connected to a controlled perfusion system with continuous pressure (gravimetric level) ranging from 5 to 30cmH2O. Effective Rv was calculated by ratio of pulmonary artery pressure (PPA) by pulmonary artery flow (V'PA). Rv in the decellularized lungs scaffolds decreased at increasing V'PA, stabilizing at a pulmonary arterial pressure greater than 20cmH2O. On the other hand, CPAP had no influence on vascular resistance in the lung scaffolds after being subjected to pulmonary artery pressure of 5cmH2O. In conclusion, compared to positive airway pressure, arterial lung pressure markedly influences the mechanics of vascular resistance in decellularized lungs.
Collapse
|
71
|
An official American Thoracic Society workshop report: stem cells and cell therapies in lung biology and diseases. Ann Am Thorac Soc 2016; 12:S79-97. [PMID: 25897748 DOI: 10.1513/annalsats.201502-086st] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The University of Vermont College of Medicine and the Vermont Lung Center, in collaboration with the NHLBI, Alpha-1 Foundation, American Thoracic Society, European Respiratory Society, International Society for Cell Therapy, and the Pulmonary Fibrosis Foundation, convened a workshop, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," held July 29 to August 1, 2013 at the University of Vermont. The conference objectives were to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are all rapidly expanding areas of study that both provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, discuss and debate current controversies, and identify future research directions and opportunities for both basic and translational research in cell-based therapies for lung diseases. This conference was a follow-up to four previous biennial conferences held at the University of Vermont in 2005, 2007, 2009, and 2011. Each of those conferences, also sponsored by the National Institutes of Health, American Thoracic Society, and Respiratory Disease Foundations, has been important in helping guide research and funding priorities. The major conference recommendations are summarized at the end of the report and highlight both the significant progress and major challenges in these rapidly progressing fields.
Collapse
|
72
|
Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches. Int J Mol Sci 2016; 17:ijms17010128. [PMID: 26797607 PMCID: PMC4730369 DOI: 10.3390/ijms17010128] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/25/2022] Open
Abstract
The tissue turnover of unperturbed adult lung is remarkably slow. However, after injury or insult, a specialised group of facultative lung progenitors become activated to replenish damaged tissue through a reparative process called regeneration. Disruption in this process results in healing by fibrosis causing aberrant lung remodelling and organ dysfunction. Post-insult failure of regeneration leads to various incurable lung diseases including chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis. Therefore, identification of true endogenous lung progenitors/stem cells, and their regenerative pathway are crucial for next-generation therapeutic development. Recent studies provide exciting and novel insights into postnatal lung development and post-injury lung regeneration by native lung progenitors. Furthermore, exogenous application of bone marrow stem cells, embryonic stem cells and inducible pluripotent stem cells (iPSC) show evidences of their regenerative capacity in the repair of injured and diseased lungs. With the advent of modern tissue engineering techniques, whole lung regeneration in the lab using de-cellularised tissue scaffold and stem cells is now becoming reality. In this review, we will highlight the advancement of our understanding in lung regeneration and development of stem cell mediated therapeutic strategies in combating incurable lung diseases.
Collapse
|
73
|
Ebrahimi A, Kardar GA, Teimoori-Toolabi L, Toolabi L, Ghanbari H, Sadroddiny E. Inducible expression of indoleamine 2,3-dioxygenase attenuates acute rejection of tissue-engineered lung allografts in rats. Gene 2015; 576:412-20. [PMID: 26506443 DOI: 10.1016/j.gene.2015.10.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 10/06/2015] [Accepted: 10/21/2015] [Indexed: 12/19/2022]
Abstract
Lung disease remains one of the principal causes of death worldwide and the incidence of pulmonary diseases is increasing. Complexity in treatments and shortage of donors leads us to develop new ways for lung disease treatment. One promising strategy is preparing engineered lung for transplantation. In this context, employing new immunosuppression strategies which suppresses immune system locally rather than systemic improves transplant survival. This tends to reduce the difficulties in transplant rejection and the systemic impact of the use of immunosuppressive drugs which causes side effects such as serious infections and malignancies. In our study examining the immunosuppressive effects of IDO expression, we produced rat lung tissues with the help of decellularized tissue, differentiating medium and rat mesenchymal stem cells. Transduction of these cells by IDO expressing lentiviruses provided inducible and local expression of this gene. To examine immunosuppressive properties of IDO expression by these tissues, we transplanted these allografts into rats and, subsequently, evaluated cytokine expression and histopathological properties. Expression of inflammatory cytokines IFNγ and TNFα were significantly downregulated in IDO expressing allograft. Moreover, acute rejection score of this experimental group was also lower comparing other two groups and mRNA levels of FOXP3, a regulatory T cell marker, upregulated in IDO expressing group. However, infiltrating lymphocyte counting did not show significant difference between groups. This study demonstrates that IDO gene transfer into engineered lung allograft tissues significantly attenuates acute allograft damage suggesting local therapy with IDO as a strategy to reduce the need for systemic immunosuppression and, thereby, its side effects.
Collapse
Affiliation(s)
- Ammar Ebrahimi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholam Ali Kardar
- Immunology, Asthma and Allergy Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - LadanTeimoori Toolabi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Iran
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
74
|
Stabler CT, Lecht S, Mondrinos MJ, Goulart E, Lazarovici P, Lelkes PI. Revascularization of decellularized lung scaffolds: principles and progress. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1273-85. [PMID: 26408553 DOI: 10.1152/ajplung.00237.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/23/2015] [Indexed: 02/07/2023] Open
Abstract
There is a clear unmet clinical need for novel biotechnology-based therapeutic approaches to lung repair and/or replacement, such as tissue engineering of whole bioengineered lungs. Recent studies have demonstrated the feasibility of decellularizing the whole organ by removal of all its cellular components, thus leaving behind the extracellular matrix as a complex three-dimensional (3D) biomimetic scaffold. Implantation of decellularized lung scaffolds (DLS), which were recellularized with patient-specific lung (progenitor) cells, is deemed the ultimate alternative to lung transplantation. Preclinical studies demonstrated that, upon implantation in rodent models, bioengineered lungs that were recellularized with airway and vascular cells were capable of gas exchange for up to 14 days. However, the long-term applicability of this concept is thwarted in part by the failure of current approaches to reconstruct a physiologically functional, quiescent endothelium lining the entire vascular tree of reseeded lung scaffolds, as inferred from the occurrence of hemorrhage into the airway compartment and thrombosis in the vasculature in vivo. In this review, we explore the idea that successful whole lung bioengineering will critically depend on 1) preserving and/or reestablishing the integrity of the subendothelial basement membrane, especially of the ultrathin respiratory membrane separating airways and capillaries, during and following decellularization and 2) restoring vascular physiological functionality including the barrier function and quiescence of the endothelial lining following reseeding of the vascular compartment. We posit that physiological reconstitution of the pulmonary vascular tree in its entirety will significantly promote the clinical translation of the next generation of bioengineered whole lungs.
Collapse
Affiliation(s)
- Collin T Stabler
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Shimon Lecht
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Mark J Mondrinos
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ernesto Goulart
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; and
| | - Philip Lazarovici
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania;
| |
Collapse
|
75
|
Price AP, Godin LM, Domek A, Cotter T, D'Cunha J, Taylor DA, Panoskaltsis-Mortari A. Automated decellularization of intact, human-sized lungs for tissue engineering. Tissue Eng Part C Methods 2015; 21:94-103. [PMID: 24826875 DOI: 10.1089/ten.tec.2013.0756] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We developed an automated system that can be used to decellularize whole human-sized organs and have shown lung as an example. Lungs from 20 to 30 kg pigs were excised en bloc with the trachea and decellularized with our established protocol of deionized water, detergents, sodium chloride, and porcine pancreatic DNase. A software program was written to control a valve manifold assembly that we built for selection and timing of decellularization fluid perfusion through the airway and the vasculature. This system was interfaced with a prototypic bioreactor chamber that was connected to another program, from a commercial source, which controlled the volume and flow pressure of fluids. Lung matrix that was decellularized by the automated method was compared to a manual method previously used by us and others. Automation resulted in more consistent acellular matrix preparations as demonstrated by measuring levels of DNA, hydroxyproline (collagen), elastin, laminin, and glycosaminoglycans. It also proved highly beneficial in saving time as the decellularization procedure was reduced from days down to just 24 h. Developing a rapid, controllable, automated system for production of reproducible matrices in a closed system is a major step forward in whole-organ tissue engineering.
Collapse
Affiliation(s)
- Andrew P Price
- 1 Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota Cancer Center, University of Minnesota , Minneapolis, Minnesota
| | | | | | | | | | | | | |
Collapse
|
76
|
Qi C, Yan X, Huang C, Melerzanov A, Du Y. Biomaterials as carrier, barrier and reactor for cell-based regenerative medicine. Protein Cell 2015; 6:638-53. [PMID: 26088192 PMCID: PMC4537472 DOI: 10.1007/s13238-015-0179-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/11/2015] [Indexed: 01/24/2023] Open
Abstract
Cell therapy has achieved tremendous success in regenerative medicine in the past several decades. However, challenges such as cell loss, death and immune-rejection after transplantation still persist. Biomaterials have been designed as carriers to deliver cells to desirable region for local tissue regeneration; as barriers to protect transplanted cells from host immune attack; or as reactors to stimulate host cell recruitment, homing and differentiation. With the assistance of biomaterials, improvement in treatment efficiency has been demonstrated in numerous animal models of degenerative diseases compared with routine free cell-based therapy. Emerging clinical applications of biomaterial assisted cell therapies further highlight their great promise in regenerative therapy and even cure for complex diseases, which have been failed to realize by conventional therapeutic approaches.
Collapse
Affiliation(s)
- Chunxiao Qi
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
| | - Xiaojun Yan
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
| | - Chenyu Huang
- />Department of Plastic and Reconstructive Surgery, Beijing Tsinghua Changgung Hospital; Medical Center, Tsinghua University, Beijing, 102218 China
| | - Alexander Melerzanov
- />Cellular and Molecular Technologies Laboratory, MIPT, Dolgoprudny, 141701 Russia
| | - Yanan Du
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
- />Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 310003 China
| |
Collapse
|
77
|
Welman T, Michel S, Segaren N, Shanmugarajah K. Bioengineering for Organ Transplantation: Progress and Challenges. Bioengineered 2015; 6:257-61. [PMID: 26259720 DOI: 10.1080/21655979.2015.1081320] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Organ transplantation can offer a curative option for patients with end stage organ failure. Unfortunately the treatment is severely limited by the availability of donor organs. Organ bioengineering could provide a solution to the worldwide critical organ shortage. The majority of protocols to date have employed the use of decellularization-recellularization technology of naturally occurring tissues and organs with promising results in heart, lung, liver, pancreas, intestine and kidney engineering. Successful decellularization has provided researchers with suitable scaffolds to attempt cell reseeding. Future work will need to focus on the optimization of organ specific recellularization techniques before organ bioengineering can become clinically translatable. This review will examine the current progress in organ bioengineering and highlight future challenges in the field.
Collapse
Affiliation(s)
- Ted Welman
- a Ashford and St Peter's Hospitals NHS Foundation Trust ; Chertsey, Surrey , United Kingdom
| | - Sebastian Michel
- b Department of Cardiac Surgery ; Ludwig-Maximilians-Universtät München ; Munich , Germany
| | - Nicholas Segaren
- c Department of Plastic Surgery ; Addenbrooke's Hospital ; Cambridge , United Kingdom
| | | |
Collapse
|
78
|
Prakash YS, Tschumperlin DJ, Stenmark KR. Coming to terms with tissue engineering and regenerative medicine in the lung. Am J Physiol Lung Cell Mol Physiol 2015; 309:L625-38. [PMID: 26254424 DOI: 10.1152/ajplung.00204.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/04/2015] [Indexed: 01/10/2023] Open
Abstract
Lung diseases such as emphysema, interstitial fibrosis, and pulmonary vascular diseases cause significant morbidity and mortality, but despite substantial mechanistic understanding, clinical management options for them are limited, with lung transplantation being implemented at end stages. However, limited donor lung availability, graft rejection, and long-term problems after transplantation are major hurdles to lung transplantation being a panacea. Bioengineering the lung is an exciting and emerging solution that has the ultimate aim of generating lung tissues and organs for transplantation. In this article we capture and review the current state of the art in lung bioengineering, from the multimodal approaches, to creating anatomically appropriate lung scaffolds that can be recellularized to eventually yield functioning, transplant-ready lungs. Strategies for decellularizing mammalian lungs to create scaffolds with native extracellular matrix components vs. de novo generation of scaffolds using biocompatible materials are discussed. Strengths vs. limitations of recellularization using different cell types of various pluripotency such as embryonic, mesenchymal, and induced pluripotent stem cells are highlighted. Current hurdles to guide future research toward achieving the clinical goal of transplantation of a bioengineered lung are discussed.
Collapse
Affiliation(s)
- Y S Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota;
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Division of Pulmonary Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| |
Collapse
|
79
|
Peloso A, Dhal A, Zambon JP, Li P, Orlando G, Atala A, Soker S. Current achievements and future perspectives in whole-organ bioengineering. Stem Cell Res Ther 2015; 6:107. [PMID: 26028404 PMCID: PMC4450459 DOI: 10.1186/s13287-015-0089-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 05/06/2015] [Indexed: 12/11/2022] Open
Abstract
Irreversible end-stage organ failure represents one of the leading causes of death, and organ transplantation is currently the only curative solution. Donor organ shortage and adverse effects of immunosuppressive regimens are the major limiting factors for this definitive practice. Recent developments in bioengineering and regenerative medicine could provide a solid base for the future creation of implantable, bioengineered organs. Whole-organ detergent-perfusion protocols permit clinicians to gently remove all the cells and at the same time preserve the natural three-dimensional framework of the native organ. Several decellularized organs, including liver, kidney, and pancreas, have been created as a platform for further successful seeding. These scaffolds are composed of organ-specific extracellular matrix that contains growth factors important for cellular growth and function. Macro- and microvascular tree is entirely maintained and can be incorporated in the recipient's vascular system after the implant. This review will emphasize recent achievements in the whole-organ scaffolds and at the same time underline complications that the scientific community has to resolve before reaching a functional bioengineered organ.
Collapse
Affiliation(s)
- Andrea Peloso
- IRCCS Policlinico San Matteo, Department of General Surgery, University of Pavia, Viale Golgi 19, Pavia, 27100, Italy. .,Wake Forest Institute for Regenerative Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27157, USA.
| | - Abritee Dhal
- Wake Forest Institute for Regenerative Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27157, USA.
| | - Joao P Zambon
- Wake Forest Institute for Regenerative Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27157, USA.
| | - Peng Li
- Wake Forest Institute for Regenerative Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27157, USA. .,Department of General Surgery Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu, 226001, China.
| | - Giuseppe Orlando
- Wake Forest Institute for Regenerative Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27157, USA. .,Wake Forest School of Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27517, USA.
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27157, USA. .,Wake Forest School of Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27517, USA.
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Medical Centre Boulevard, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
80
|
Chen G, Chen J, Yang B, Li L, Luo X, Zhang X, Feng L, Jiang Z, Yu M, Guo W, Tian W. Combination of aligned PLGA/Gelatin electrospun sheets, native dental pulp extracellular matrix and treated dentin matrix as substrates for tooth root regeneration. Biomaterials 2015; 52:56-70. [PMID: 25818413 DOI: 10.1016/j.biomaterials.2015.02.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/25/2015] [Accepted: 02/01/2015] [Indexed: 02/05/2023]
Abstract
In tissue engineering, scaffold materials provide effective structural support to promote the repair of damaged tissues or organs through simulating the extracellular matrix (ECM) microenvironments for stem cells. This study hypothesized that simulating the ECM microenvironments of periodontium and dental pulp/dentin complexes would contribute to the regeneration of tooth root. Here, aligned PLGA/Gelatin electrospun sheet (APES), treated dentin matrix (TDM) and native dental pulp extracellular matrix (DPEM) were fabricated and combined into APES/TDM and DPEM/TDM for periodontium and dental pulp regeneration, respectively. This study firstly examined the physicochemical properties and biocompatibilities of both APES and DPEM in vitro, and further investigated the degradation of APES and revascularization of DPEM in vivo. Then, the potency of APES/TDM and DPEM/TDM in odontogenic induction was evaluated via co-culture with dental stem cells. Finally, we verified the periodontium and dental pulp/dentin complex regeneration in the jaw of miniature swine. Results showed that APES possessed aligned fiber orientation which guided cell proliferation while DPEM preserved the intrinsic fiber structure and ECM proteins. Importantly, both APES/TDM and DPEM/TDM facilitated the odontogenic differentiation of dental stem cells in vitro. Seeded with stem cells, the sandwich composites (APES/TDM/DPEM) generated tooth root-like tissues after being transplanted in porcine jaws for 12 w. In dental pulp/dentin complex-like tissues, columnar odontoblasts-like layer arranged along the interface between newly-formed predentin matrix and dental pulp-like tissues in which blood vessels could be found; in periodontium complex-like tissues, cellular cementum and periodontal ligament (PDL)-like tissues were generated on the TDM surface. Thus, above results suggest that APES and DPEM exhibiting appropriate physicochemical properties and well biocompatibilities, in accompany with TDM, could make up an ECM microenvironment for tooth root regeneration, which also offers a strategy for complex tissue or organ regeneration.
Collapse
Affiliation(s)
- Gang Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jinlong Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Bo Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Lei Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xiangyou Luo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xuexin Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Lian Feng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Zongting Jiang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Mei Yu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Pedodontics, West China College of Stomatology, Sichuan University, No.14, 3rd Section, Renmin South Road, Chengdu, 610041, PR China.
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
81
|
Rapid porcine lung decellularization using a novel organ regenerative control acquisition bioreactor. ASAIO J 2015; 61:71-7. [PMID: 25303798 DOI: 10.1097/mat.0000000000000159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To regenerate discarded lungs that would not normally be used for transplant, ex vivo reseeding after decellularization may produce organs suitable for clinical transplantation and therefore close the donor gap. Organ regenerative control acquisition (Harvard Biosciences, Holliston, MA), a novel bioreactor system that simulates physiological conditions, was used to evaluate a method of rapid decellularization. Although most current decellularization methods are 24-72 hours, we hypothesized that perfusing porcine lungs with detergents at higher pressures for less time would yield comparable bioscaffolds suitable for future experimentation. Methods involved perfusion of 1% Triton X-100 (Triton) and 0.1% sodium dodecyl sulfate at varied physiological flow rates. Architecture of native and decellularized lungs was analyzed with hematoxylin and eosin (H&E) staining, transmission electron microscopy (TEM), and scanning electron microscopy (SEM). Dry gas and liquid ventilation techniques were introduced. Our 7 hour decellularization procedure removes nuclear material while maintaining architecture. Bioscaffolds have the microarchitecture for reseeding of stem cells. Hematoxylin and eosin staining suggested removal of nuclear material, whereas SEM and TEM imaging demonstrated total removal of cells with structural architecture preserved. This process can lead to clinical implementation, thereby increasing the availability of human lungs for transplantation.
Collapse
|
82
|
Crabbé A, Liu Y, Sarker SF, Bonenfant NR, Barrila J, Borg ZD, Lee JJ, Weiss DJ, Nickerson CA. Recellularization of decellularized lung scaffolds is enhanced by dynamic suspension culture. PLoS One 2015; 10:e0126846. [PMID: 25962111 PMCID: PMC4427280 DOI: 10.1371/journal.pone.0126846] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 04/08/2015] [Indexed: 12/20/2022] Open
Abstract
Strategies are needed to improve repopulation of decellularized lung scaffolds with stromal and functional epithelial cells. We demonstrate that decellularized mouse lungs recellularized in a dynamic low fluid shear suspension bioreactor, termed the rotating wall vessel (RWV), contained more cells with decreased apoptosis, increased proliferation and enhanced levels of total RNA compared to static recellularization conditions. These results were observed with two relevant mouse cell types: bone marrow-derived mesenchymal stromal (stem) cells (MSCs) and alveolar type II cells (C10). In addition, MSCs cultured in decellularized lungs under static but not bioreactor conditions formed multilayered aggregates. Gene expression and immunohistochemical analyses suggested differentiation of MSCs into collagen I-producing fibroblast-like cells in the bioreactor, indicating enhanced potential for remodeling of the decellularized scaffold matrix. In conclusion, dynamic suspension culture is promising for enhancing repopulation of decellularized lungs, and could contribute to remodeling the extracellular matrix of the scaffolds with subsequent effects on differentiation and functionality of inoculated cells.
Collapse
Affiliation(s)
- Aurélie Crabbé
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Yulong Liu
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Shameema F. Sarker
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Nicholas R. Bonenfant
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Jennifer Barrila
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Zachary D. Borg
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - James J. Lee
- Division of Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - Daniel J. Weiss
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Cheryl A. Nickerson
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- * E-mail:
| |
Collapse
|
83
|
da Palma RK, Campillo N, Uriarte JJ, Oliveira LVF, Navajas D, Farré R. Pressure- and flow-controlled media perfusion differently modify vascular mechanics in lung decellularization. J Mech Behav Biomed Mater 2015; 49:69-79. [PMID: 26002417 DOI: 10.1016/j.jmbbm.2015.04.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 04/18/2015] [Accepted: 04/23/2015] [Indexed: 01/07/2023]
Abstract
Organ biofabrication is a potential future alternative for obtaining viable organs for transplantation. Achieving intact scaffolds to be recellularized is a key step in lung bioengineering. Perfusion of decellularizing media through the pulmonary artery has shown to be effective. How vascular perfusion pressure and flow vary throughout lung decellularization, which is not well known, is important for optimizing the process (minimizing time) while ensuring scaffold integrity (no barotrauma). This work was aimed at characterizing the pressure/flow relationship at the pulmonary vasculature and at how effective vascular resistance depends on pressure- and flow-controlled variables when applying different methods of media perfusion for lung decellularization. Lungs from 43 healthy mice (C57BL/6; 7-8 weeks old) were investigated. After excision and tracheal cannulation, lungs were inflated at 10 cmH2O airway pressure and subjected to conventional decellularization with a solution of 1% sodium dodecyl sulfate (SDS). Pressure (PPA) and flow (V'PA) at the pulmonary artery were continuously measured. Decellularization media was perfused through the pulmonary artery: (a) at constant PPA=20 cmH2O or (b) at constant V'PA=0.5 and 0.2 ml/min. Effective vascular resistance was computed as Rv=PPA/V'PA. Rv (in cmH2O/(ml/min)); mean±SE) considerably varied throughout lung decellularization, particularly for pressure-controlled perfusion (from 29.1±3.0 in baseline to a maximum of 664.1±164.3 (p<0.05), as compared with flow-controlled perfusion (from 49.9±3.3 and 79.5±5.1 in baseline to a maximum of 114.4±13.9 and 211.7±70.5 (p<0.05, both), for V'PA of 0.5 and 0.2 ml/min respectively. Most of the media infused to the pulmonary artery throughout decellularization circulated to the airways compartment across the alveolar-capillary membrane. This study shows that monitoring perfusion mechanics throughout decellularization provides information relevant for optimizing the process time while ensuring that vascular pressure is kept within a safety range to preserve the organ scaffold integrity.
Collapse
Affiliation(s)
- Renata K da Palma
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Master's and Doctoral Degree Programs in Rehabilitation Sciences, Nove de Julho University, Sao Paulo, Brazil
| | - Noelia Campillo
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Juan J Uriarte
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain
| | - Luis V F Oliveira
- Master's and Doctoral Degree Programs in Rehabilitation Sciences, Nove de Julho University, Sao Paulo, Brazil
| | - Daniel Navajas
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut de Bioenginyeria de Catalunya, Barcelona, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain
| | - Ramon Farré
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Respiratorias, Madrid, Spain; Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.
| |
Collapse
|
84
|
Calle EA, Mendez JJ, Ghaedi M, Leiby KL, Bove PF, Herzog EL, Sundaram S, Niklason LE. Fate of distal lung epithelium cultured in a decellularized lung extracellular matrix. Tissue Eng Part A 2015; 21:1916-28. [PMID: 25789725 DOI: 10.1089/ten.tea.2014.0511] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Type II cells are the defenders of the alveolus. They produce surfactant to prevent alveolar collapse, they actively transport water to prevent filling of the air sacs that would otherwise prevent gas exchange, and they differentiate to type I epithelial cells. They are an indispensable component of functional lung tissue. To understand the functionality of type II cells in isolation, we sought to track their fate in decellularized matrices and to assess their ability to contribute to barrier function by differentiation to type I alveolar epithelial cells. Rat type II cells were isolated from neonatal rat lungs by labeling with the RTII-70 surface marker and separation using a magnetic column. This produced a population of ∼50% RTII-70-positive cells accompanied by few type I epithelial cells or α-actin-positive mesenchymal cells. This population was seeded into decellularized rat lung matrices and cultured for 1 or 7 days. Culture in Dulbecco's modified Eagle's medium +10% fetal bovine serum (FBS) resulted in reduced expression of epithelial markers and increased expression of mesenchymal markers. By 7 days, no epithelial markers were visible by immunostaining; nearly all cells were α-actin positive. Gene expression for the mesenchymal markers, α-actin, vimentin, and TGF-βR, was significantly upregulated on day 1 (p=0.0005, 0.0005, and 2.342E-5, respectively). Transcript levels of α-actin and TGF-βR remained high at 7 days (p=1.364E-10 and 0.0002). Interestingly, human type II cells cultured under the same conditions showed a similar trend in the loss of epithelial markers, but did not display high expression of mesenchymal markers. Rat cells additionally showed the ability to produce and degrade the basement membrane and extracellular matrix components, such as fibronectin, collagen IV, and collagen I. Quantitative real-time reverse transcription polymerase chain reaction (RT-PCR) showed significant increases in expression of the fibronectin and matrix metalloprotease-2 (MMP-2) genes after 1 day in culture (p=0.0135 and 0.0128, respectively) and elevated collagen I expression at 7 days (p=0.0016). These data suggest that the original type II-enriched population underwent a transition to increased expression of mesenchymal markers, perhaps as part of a survival or wound-healing program. These results suggest that additional medium components and/or the application of physiologically appropriate stimuli such as ventilation may be required to promote lung-specific epithelial phenotypes.
Collapse
Affiliation(s)
- Elizabeth A Calle
- 1Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Julio J Mendez
- 2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| | - Mahboobe Ghaedi
- 2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| | - Katherine L Leiby
- 1Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Peter F Bove
- 3Cystic Fibrosis/Pulmonary Research Treatment Center, University of North Carolina, Chapel Hill, North Carolina
| | - Erica L Herzog
- 4Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sumati Sundaram
- 2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| | - Laura E Niklason
- 1Department of Biomedical Engineering, Yale University, New Haven, Connecticut.,2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
85
|
Hill RC, Calle EA, Dzieciatkowska M, Niklason LE, Hansen KC. Quantification of extracellular matrix proteins from a rat lung scaffold to provide a molecular readout for tissue engineering. Mol Cell Proteomics 2015; 14:961-73. [PMID: 25660013 PMCID: PMC4390273 DOI: 10.1074/mcp.m114.045260] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/06/2015] [Indexed: 01/20/2023] Open
Abstract
The use of extracellular matrix (ECM) scaffolds, derived from decellularized tissues for engineered organ generation, holds enormous potential in the field of regenerative medicine. To support organ engineering efforts, we developed a targeted proteomics method to extract and quantify extracellular matrix components from tissues. Our method provides more complete and accurate protein characterization than traditional approaches. This is accomplished through the analysis of both the chaotrope-soluble and -insoluble protein fractions and using recombinantly generated stable isotope labeled peptides for endogenous protein quantification. Using this approach, we have generated 74 peptides, representing 56 proteins to quantify protein in native (nondecellularized) and decellularized lung matrices. We have focused on proteins of the ECM and additional intracellular proteins that are challenging to remove during the decellularization procedure. Results indicate that the acellular lung scaffold is predominantly composed of structural collagens, with the majority of these proteins found in the insoluble ECM, a fraction that is often discarded using widely accepted proteomic methods. The decellularization procedure removes over 98% of intracellular proteins evaluated and retains, to varying degrees, proteoglycans and glycoproteins of the ECM. Accurate characterization of ECM proteins from tissue samples will help advance organ engineering efforts by generating a molecular readout that can be correlated with functional outcome to drive the next generation of engineered organs.
Collapse
Affiliation(s)
- Ryan C Hill
- ‡Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045
| | | | - Monika Dzieciatkowska
- ‡Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045
| | - Laura E Niklason
- §Department of Biomedical Engineering and Anesthesiology, ¶Yale University, New Haven, CT 06519
| | - Kirk C Hansen
- ‡Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045,
| |
Collapse
|
86
|
Scarritt ME, Pashos NC, Bunnell BA. A review of cellularization strategies for tissue engineering of whole organs. Front Bioeng Biotechnol 2015; 3:43. [PMID: 25870857 PMCID: PMC4378188 DOI: 10.3389/fbioe.2015.00043] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/16/2015] [Indexed: 12/22/2022] Open
Abstract
With the advent of whole organ decellularization, extracellular matrix scaffolds suitable for organ engineering were generated from numerous tissues, including the heart, lung, liver, kidney, and pancreas, for use as alternatives to traditional organ transplantation. Biomedical researchers now face the challenge of adequately and efficiently recellularizing these organ scaffolds. Herein, an overview of whole organ decellularization and a thorough review of the current literature for whole organ recellularization are presented. The cell types, delivery methods, and bioreactors employed for recellularization are discussed along with commercial and clinical considerations, such as immunogenicity, biocompatibility, and Food and Drug Administartion regulation.
Collapse
Affiliation(s)
- Michelle E Scarritt
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine , New Orleans, LA , USA
| | - Nicholas C Pashos
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine , New Orleans, LA , USA ; Bioinnovation PhD Program, Tulane University , New Orleans, LA , USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine , New Orleans, LA , USA ; Department of Pharmacology, Tulane University School of Medicine , New Orleans, LA , USA
| |
Collapse
|
87
|
Fujita M. New therapies for chronic obstructive pulmonary disease, lung regeneration. World J Respirol 2015; 5:34-39. [DOI: 10.5320/wjr.v5.i1.34] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/15/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by the presence of airflow limitations that are not fully reversible and is a major cause of chronic morbidity and mortality worldwide. Although there has been extensive research examining the molecular mechanisms underlying the development of COPD, there is no proven clinically effective treatment for promoting recovery from established COPD. At present, regeneration is the only hope for a cure in patients with COPD. In this article, we review current treatments for COPD, focusing particularly on recent advances in lung regeneration based on two major approaches: regeneration-promoting agents and cell therapy. Retinoic acids are the major focus among regeneration-promoting agents, while mesenchymal stem cells are the main topic in the field of cell-based therapy. This article aims to provide valuable information for developing new therapies for COPD.
Collapse
|
88
|
O'Leary C, Gilbert JL, O'Dea S, O'Brien FJ, Cryan SA. Respiratory Tissue Engineering: Current Status and Opportunities for the Future. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:323-44. [PMID: 25587703 DOI: 10.1089/ten.teb.2014.0525] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Currently, lung disease and major airway trauma constitute a major global healthcare burden with limited treatment options. Airway diseases such as chronic obstructive pulmonary disease and cystic fibrosis have been identified as the fifth highest cause of mortality worldwide and are estimated to rise to fourth place by 2030. Alternate approaches and therapeutic modalities are urgently needed to improve clinical outcomes for chronic lung disease. This can be achieved through tissue engineering of the respiratory tract. Interest is growing in the use of airway tissue-engineered constructs as both a research tool, to further our understanding of airway pathology, validate new drugs, and pave the way for novel drug therapies, and also as regenerative medical devices or as an alternative to transplant tissue. This review provides a concise summary of the field of respiratory tissue engineering to date. An initial overview of airway anatomy and physiology is given, followed by a description of the stem cell populations and signaling processes involved in parenchymal healing and tissue repair. We then focus on the different biomaterials and tissue-engineered systems employed in upper and lower respiratory tract engineering and give a final perspective of the opportunities and challenges facing the field of respiratory tissue engineering.
Collapse
Affiliation(s)
- Cian O'Leary
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland , Dublin, Ireland .,2 School of Pharmacy, Royal College of Surgeons in Ireland , Dublin, Ireland .,3 Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin , Dublin, Ireland
| | - Jennifer L Gilbert
- 4 Department of Biology, Institute of Immunology, University of Ireland , Maynooth, Ireland
| | - Shirley O'Dea
- 4 Department of Biology, Institute of Immunology, University of Ireland , Maynooth, Ireland
| | - Fergal J O'Brien
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland , Dublin, Ireland .,3 Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin , Dublin, Ireland .,5 Trinity Centre of Bioengineering, Trinity College Dublin , Dublin, Ireland
| | - Sally-Ann Cryan
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland , Dublin, Ireland .,2 School of Pharmacy, Royal College of Surgeons in Ireland , Dublin, Ireland .,5 Trinity Centre of Bioengineering, Trinity College Dublin , Dublin, Ireland
| |
Collapse
|
89
|
|
90
|
Weiss DJ. Concise review: current status of stem cells and regenerative medicine in lung biology and diseases. Stem Cells 2014; 32:16-25. [PMID: 23959715 DOI: 10.1002/stem.1506] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 07/24/2013] [Indexed: 12/29/2022]
Abstract
Lung diseases remain a significant and devastating cause of morbidity and mortality worldwide. In contrast to many other major diseases, lung diseases notably chronic obstructive pulmonary diseases (COPDs), including both asthma and emphysema, are increasing in prevalence and COPD is expected to become the third leading cause of disease mortality worldwide by 2020. New therapeutic options are desperately needed. A rapidly growing number of investigations of stem cells and cell therapies in lung biology and diseases as well as in ex vivo lung bioengineering have offered exciting new avenues for advancing knowledge of lung biology as well as providing novel potential therapeutic approaches for lung diseases. These initial observations have led to a growing exploration of endothelial progenitor cells and mesenchymal stem (stromal) cells in clinical trials of pulmonary hypertension and COPD with other clinical investigations planned. Ex vivo bioengineering of the trachea, larynx, diaphragm, and the lung itself with both biosynthetic constructs as well as decellularized tissues have been used to explore engineering both airway and vascular systems of the lung. Lung is thus a ripe organ for a variety of cell therapy and regenerative medicine approaches. Current state-of-the-art progress for each of the above areas will be presented as will discussion of current considerations for cell therapy-based clinical trials in lung diseases.
Collapse
Affiliation(s)
- Daniel J Weiss
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
91
|
Vadasz S, Jensen T, Moncada C, Girard E, Zhang F, Blanchette A, Finck C. Second and third trimester amniotic fluid mesenchymal stem cells can repopulate a de-cellularized lung scaffold and express lung markers. J Pediatr Surg 2014; 49:1554-63. [PMID: 25475793 DOI: 10.1016/j.jpedsurg.2014.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND/PURPOSE This study examined the potential of amniotic fluid mesenchymal stem cells (AF-MSCs) to generate lung precursor cells in vitro and on a xenologous three-dimensional de-cellularized lung scaffold. METHODS AF-MSCs were isolated from human amniotic fluid obtained from 17-37 weeks gestation. Lung differentiation was induced on Matrigel or on de-cellularized rat lungs intra-tracheally injected with AF-MSCs by culturing with a modification of small airway growth medium (mSAGM) lacking retinoic acid (RA) and triodothyronine (T3) with addition of fibroblast growth factor-10 (FGF10). Cells and scaffolds were characterized by immunofluorescence and RT-PCR for markers of viability, proliferation, and lung distal airway differentiation (TTF-1(+) and SPC(+)) in the absence of markers of brain (TuJ1(-)) and thyroid (Pax8(-)). RESULTS After culture in mSAGM on either Matrigel or lung scaffolds, there were TTF-1(+)/TuJ1(-)/Pax8(-) cells, indicating a lung precursor phenotype. In addition, SPC(+) cells also evolved suggesting a more mature lung phenotype. CONCLUSIONS We demonstrate that mid- to late-trimester AF-MSCs can be induced to develop into lung precursor cells when cultured on the appropriate extracellular matrix (ECM), making them a viable source for use in cell therapy or development of an ex vivo tissue engineered lung.
Collapse
Affiliation(s)
- Stephanie Vadasz
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Todd Jensen
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Camilo Moncada
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Eric Girard
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030; Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106
| | - Fan Zhang
- Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106
| | - Alex Blanchette
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Christine Finck
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030; Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106.
| |
Collapse
|
92
|
Foster WS, Suen CM, Stewart DJ. Regenerative Cell and Tissue-based Therapies for Pulmonary Arterial Hypertension. Can J Cardiol 2014; 30:1350-60. [DOI: 10.1016/j.cjca.2014.08.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/13/2014] [Accepted: 08/24/2014] [Indexed: 12/21/2022] Open
|
93
|
Kajbafzadeh AM, Sabetkish N, Sabetkish S, Tavangar SM, Hossein Beigi RS, Talebi MA, Akbarzadeh A, Nikfarjam L. Lung tissue engineering and preservation of alveolar microstructure using a novel casting method. Biotech Histochem 2014; 90:111-23. [PMID: 25268847 DOI: 10.3109/10520295.2014.957724] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We used a rat model to decellularize and seed alveolar cells on a three-dimensional lung scaffold to preserve alveolar microarchitecture. We verified the preservation of terminal respiratory structure by casting and by scanning electron microscopy (SEM) of the casts after decellularization. Whole lungs were obtained from 12 healthy Sprague-Dawley rats, cannulated through the trachea under sterile conditions, and decellularized using a detergent-based method. Casting of both natural and decellularized lungs was performed to verify preservation of the inner microstructure of scaffolds for further cell seeding. Alveolar cell seeding was performed using green fluorescent protein (GFP) lung cells and non-GFP lung cells, and a peristaltic pump. We assessed cell seeding using histological and immunohistochemical staining, and enzymatic evaluation. All cellular components were removed completely from the scaffolds, and histological staining and SEM of casts were used to verify the preservation of tissue structure. Tensile tests verified conservation of biomechanical properties. The hydroxyproline content of decellularized lungs was similar to native lung. Histological and immunohistochemical evaluations showed effective cell seeding on decellularized matrices. Enzymatic measurement of trypsin and alpha 1 antitrypsin suggested the potential functional properties of the regenerated lungs. Casts produced by our method have satisfactory geometrical properties for further cell seeding of lung scaffolds. Preservation of micro-architecture and terminal alveoli that was confirmed by SEM of lung casts increases the probability of an effective cell seeding process.
Collapse
Affiliation(s)
- A-M Kajbafzadeh
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | | | | | | | | | | | | | | |
Collapse
|
94
|
Momtahan N, Sukavaneshvar S, Roeder BL, Cook AD. Strategies and processes to decellularize and recellularize hearts to generate functional organs and reduce the risk of thrombosis. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:115-32. [PMID: 25084164 DOI: 10.1089/ten.teb.2014.0192] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Heart failure is one of the leading causes of death in the United States. Current therapies, such as heart transplants and bioartificial hearts, are helpful, but not optimal. Decellularization of porcine whole hearts followed by recellularization with patient-specific human cells may provide the ultimate solution for patients with heart failure. Great progress has been made in the development of efficient processes for decellularization, and the design of automated bioreactors. Challenges remain in selecting and culturing cells, growing the cells on the decellularized scaffolds without contamination, characterizing the regenerated organs, and preventing thrombosis. Various strategies have been proposed to prevent thrombosis of blood-contacting devices, including reendothelization and the creation of nonfouling surfaces using surface modification technologies. This review discusses the progress and remaining challenges involved with recellularizing whole hearts, focusing on the prevention of thrombosis.
Collapse
Affiliation(s)
- Nima Momtahan
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | | | | | | |
Collapse
|
95
|
Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell populations. Nat Med 2014; 20:822-32. [PMID: 25100528 PMCID: PMC4229034 DOI: 10.1038/nm.3642] [Citation(s) in RCA: 383] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/24/2014] [Indexed: 12/15/2022]
Abstract
Recent studies have shown that the respiratory system has an extensive ability to respond to injury and regenerate lost or damaged cells. The unperturbed adult lung is remarkably quiescent, but after insult or injury progenitor populations can be activated or remaining cells can re-enter the cell cycle. Techniques including cell-lineage tracing and transcriptome analysis have provided novel and exciting insights into how the lungs and trachea regenerate in response to injury and have allowed the identification of pathways important in lung development and regeneration. These studies are now informing approaches for modulating the pathways that may promote endogenous regeneration as well as the generation of exogenous lung cell lineages from pluripotent stem cells. The emerging advances, highlighted in this Review, are providing new techniques and assays for basic mechanistic studies as well as generating new model systems for human disease and strategies for cell replacement.
Collapse
Affiliation(s)
- Darrell N Kotton
- 1] Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts, USA. [2] Pulmonary Center, Boston University, Boston, Massachusetts, USA. [3] Department of Medicine, Boston University, Boston, Massachusetts, USA
| | - Edward E Morrisey
- 1] Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA. [2] Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA. [3] Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA. [4] Institute for Regenerative Medicine, University of Pennsylvania Philadelphia, Pennsylvania, USA
| |
Collapse
|
96
|
Garreta E, Melo E, Navajas D, Farré R. Low oxygen tension enhances the generation of lung progenitor cells from mouse embryonic and induced pluripotent stem cells. Physiol Rep 2014; 2:2/7/e12075. [PMID: 25347858 PMCID: PMC4187564 DOI: 10.14814/phy2.12075] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Whole-organ decellularization technology has emerged as a new alternative for the fabrication of bioartificial lungs. Embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) are potentially useful for recellularization since they can be directed to express phenotypic marker genes of lung epithelial cells. Normal pulmonary development takes place in a low oxygen environment ranging from 1 to 5%. By contrast, in vitro ESC and iPSC differentiation protocols are usually carried out at room-air oxygen tension. Here, we sought to determine the role played by oxygen tension on the derivation of Nkx2.1+ lung/thyroid progenitor cells from mouse ESC and iPSC. A step-wise differentiation protocol was used to generate Nkx2.1+ lung/thyroid progenitors under 20% and 5% oxygen tension. On day 12, gene expression analysis revealed that Nkx2.1 and Foxa2 (endodermal and early lung epithelial cell marker) were significantly upregulated at 5% oxygen tension in ESC and iPSC differentiated cultures compared to 20% oxygen conditions. In addition, quantification of Foxa2+Nkx2.1+Pax8- cells corresponding to the lung field, with exclusion of the potential thyroid fate identified by Pax8 expression, confirmed that the low physiologic oxygen tension exerted a significant positive effect on early pulmonary differentiation of ESC and iPSC. In conclusion, we found that 5% oxygen tension enhanced the derivation of lung progenitors from mouse ESC and iPSC compared to 20% room-air oxygen tension.
Collapse
Affiliation(s)
- Elena Garreta
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centre de Medicina Regenerativa de Barcelona (CMRB), Parc de Recerca Biomèdica de Barcelona (PRBB), Dr. Aiguader88 7ª Planta, Barcelona, 08003, Spain
| | - Esther Melo
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain F. Hoffmann-La Roche, AG, NORD DTABldg. 69/331, Basel, CH-4070, Switzerland
| | - Daniel Navajas
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
97
|
Stem cells, cell therapies, and bioengineering in lung biology and diseases. Comprehensive review of the recent literature 2010-2012. Ann Am Thorac Soc 2014; 10:S45-97. [PMID: 23869446 DOI: 10.1513/annalsats.201304-090aw] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A conference, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," was held July 25 to 28, 2011 at the University of Vermont to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are rapidly expanding areas of study that provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, to discuss and debate current controversies, and to identify future research directions and opportunities for basic and translational research in cell-based therapies for lung diseases. The goal of this article, which accompanies the formal conference report, is to provide a comprehensive review of the published literature in lung regenerative medicine from the last conference report through December 2012.
Collapse
|
98
|
Wagner DE, Bonvillain RW, Jensen T, Girard ED, Bunnell BA, Finck CM, Hoffman AM, Weiss DJ. Can stem cells be used to generate new lungs? Ex vivo lung bioengineering with decellularized whole lung scaffolds. Respirology 2014; 18:895-911. [PMID: 23614471 DOI: 10.1111/resp.12102] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 03/26/2013] [Indexed: 01/06/2023]
Abstract
For patients with end-stage lung diseases, lung transplantation is the only available therapeutic option. However, the number of suitable donor lungs is insufficient and lung transplants are complicated by significant graft failure and complications of immunosuppressive regimens. An alternative to classic organ replacement is desperately needed. Engineering of bioartificial organs using either natural or synthetic scaffolds is an exciting new potential option for generation of functional pulmonary tissue for human clinical application. Natural organ scaffolds can be generated by decellularization of native tissues; these acellular scaffolds retain the native organ ultrastructure and can be seeded with autologous cells towards the goal of regenerating functional tissues. Several decellularization strategies have been employed for lungs; however, there is no consensus on the optimal approach. A variety of cell types have been investigated as potential candidates for effective recellularization of acellular lung scaffolds. Candidate cells that might be best utilized are those which can be easily and reproducibly isolated, expanded in vitro, seeded onto decellularized matrices, induced to differentiate into pulmonary lineage cells, and which survive to functional maturity. Whole lung cell suspensions, endogenous progenitor cells, embryonic and adult stem cells and induced pluripotent stem (iPS) cells have been investigated for their applicability to repopulate acellular lung matrices. Ideally, patient-derived autologous cells would be used for lung recellularization as they have the potential to reduce the need for post-transplant immunosuppression. Several studies have performed transplantation of rudimentary bioengineered lung scaffolds in animal models with limited, short-term functionality but much further study is needed.
Collapse
Affiliation(s)
- Darcy E Wagner
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Suki B. Assessing the Functional Mechanical Properties of Bioengineered Organs With Emphasis on the Lung. J Cell Physiol 2014; 229:1134-40. [DOI: 10.1002/jcp.24600] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/03/2014] [Indexed: 01/16/2023]
Affiliation(s)
- Béla Suki
- Department of Biomedical Engineering; Boston University; Boston Massachusetts
| |
Collapse
|
100
|
Crabbé A, Ledesma MA, Nickerson CA. Mimicking the host and its microenvironment in vitro for studying mucosal infections by Pseudomonas aeruginosa. Pathog Dis 2014; 71:1-19. [PMID: 24737619 DOI: 10.1111/2049-632x.12180] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 02/01/2023] Open
Abstract
Why is a healthy person protected from Pseudomonas aeruginosa infections, while individuals with cystic fibrosis or damaged epithelium are particularly susceptible to this opportunistic pathogen? To address this question, it is essential to thoroughly understand the dynamic interplay between the host microenvironment and P. aeruginosa. Therefore, using model systems that represent key aspects of human mucosal tissues in health and disease allows recreating in vivo host-pathogen interactions in a physiologically relevant manner. In this review, we discuss how factors of mucosal tissues, such as apical-basolateral polarity, junctional complexes, extracellular matrix proteins, mucus, multicellular complexity (including indigenous microbiota), and other physicochemical factors affect P. aeruginosa pathogenesis and are thus important to mimic in vitro. We highlight in vitro cell and tissue culture model systems of increasing complexity that have been used over the past 35 years to study the infectious disease process of P. aeruginosa, mainly focusing on lung models, and their respective advantages and limitations. Continued improvements of in vitro models based on our expanding knowledge of host microenvironmental factors that participate in P. aeruginosa pathogenesis will help advance fundamental understanding of pathogenic mechanisms and increase the translational potential of research findings from bench to the patient's bedside.
Collapse
Affiliation(s)
- Aurélie Crabbé
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, AZ, USA
| | | | | |
Collapse
|