1
|
Stocco E, Barbon S, Mammana M, Zambello G, Contran M, Parnigotto PP, Macchi V, Conconi MT, Rea F, De Caro R, Porzionato A. Preclinical and clinical orthotopic transplantation of decellularized/engineered tracheal scaffolds: A systematic literature review. J Tissue Eng 2023; 14:20417314231151826. [PMID: 36874984 PMCID: PMC9974632 DOI: 10.1177/20417314231151826] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/04/2023] [Indexed: 03/07/2023] Open
Abstract
Severe tracheal injuries that cannot be managed by mobilization and end-to-end anastomosis represent an unmet clinical need and an urgent challenge to face in surgical practice; within this scenario, decellularized scaffolds (eventually bioengineered) are currently a tempting option among tissue engineered substitutes. The success of a decellularized trachea is expression of a balanced approach in cells removal while preserving the extracellular matrix (ECM) architecture/mechanical properties. Revising the literature, many Authors report about different methods for acellular tracheal ECMs development; however, only few of them verified the devices effectiveness by an orthotopic implant in animal models of disease. To support translational medicine in this field, here we provide a systematic review on studies recurring to decellularized/bioengineered tracheas implantation. After describing the specific methodological aspects, orthotopic implant results are verified. Furtherly, the only three clinical cases of compassionate use of tissue engineered tracheas are reported with a focus on outcomes.
Collapse
Affiliation(s)
- Elena Stocco
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, Padova, Italy
| | - Silvia Barbon
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, Padova, Italy
| | - Marco Mammana
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University Hospital of Padova, Padova, Italy
| | - Giovanni Zambello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University Hospital of Padova, Padova, Italy
| | - Martina Contran
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Padova, Italy
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, Padova, Italy
| | - Veronica Macchi
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, Padova, Italy
| | - Maria Teresa Conconi
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Federico Rea
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University Hospital of Padova, Padova, Italy
| | - Raffaele De Caro
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, Padova, Italy
| | - Andrea Porzionato
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, Padova, Italy
| |
Collapse
|
2
|
Samat AA, Hamid ZAA, Yahaya BH. Tissue Engineering for Tracheal Replacement: Strategies and Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022:137-163. [PMID: 35389199 DOI: 10.1007/5584_2022_707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The critical feature in trachea replacement is to provide a hollow cylindrical framework that is laterally stable and longitudinally flexible, facilitating cartilage and epithelial tissue formation. Despite advanced techniques and sources of materials used, most inherent challenges are related to the complexity of its anatomy. Limited blood supply leads to insufficient regenerative capacity for cartilage and epithelium. Natural and synthetic scaffolds, different types of cells, and growth factors are part of tissue engineering approaches with varying outcomes. Pre-vascularization remains one of the crucial factors to expedite the regenerative process in tracheal reconstruction. This review discusses the challenges and strategies used in tracheal tissue engineering, focusing on scaffold implantation in clinical and preclinical studies conducted in recent decades.
Collapse
Affiliation(s)
- Asmak Abdul Samat
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
- Fundamental Dental and Medical Sciences, Kulliyyah of Dentistry, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Zuratul Ain Abdul Hamid
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia.
| |
Collapse
|
3
|
Yoshimatsu M, Ohnishi H, Zhao C, Hayashi Y, Kuwata F, Kaba S, Okuyama H, Kawai Y, Hiwatashi N, Kishimoto Y, Sakamoto T, Ikeya M, Omori K. In vivo regeneration of rat laryngeal cartilage with mesenchymal stem cells derived from human induced pluripotent stem cells via neural crest cells. Stem Cell Res 2021; 52:102233. [PMID: 33607469 DOI: 10.1016/j.scr.2021.102233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/21/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022] Open
Abstract
The laryngotracheal cartilage is a cardinal framework for the maintenance of the airway for breathing, which occasionally requires reconstruction. Because hyaline cartilage has a poor intrinsic regenerative ability, various regenerative approaches have been attempted to regenerate laryngotracheal cartilage. The use of autologous mesenchymal stem cells (MSCs) for cartilage regeneration has been widely investigated. However, long-term culture may limit proliferative capacity. Human-induced pluripotent stem cell-derived MSCs (iMSCs) can circumvent this problem due to their unlimited proliferative capacity. This study aimed to investigate the efficacy of iMSCs in the regeneration of thyroid cartilage in immunodeficient rats. Herein, we induced iMSCs through neural crest cell intermediates. For the relevance to prospective future clinical application, induction was conducted under xeno-free/serum-free conditions. Then, clumps fabricated from an iMSC/extracellular matrix complex (C-iMSC) were transplanted into thyroid cartilage defects in immunodeficient rats. Histological examinations revealed cartilage-like regenerated tissue and human nuclear antigen (HNA)-positive surviving transplanted cells in the regenerated lesion. HNA-positive cells co-expressed SOX9, and type II collagen was identified around HNA-positive cells. These results indicated that the transplanted C-iMSCs promoted thyroid cartilage regeneration and some of the iMSCs differentiated into chondrogenic lineage cells. Induced MSCs may be a promising candidate cell therapy for human laryngotracheal reconstruction.
Collapse
Affiliation(s)
- Masayoshi Yoshimatsu
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hiroe Ohnishi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chengzhu Zhao
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yasuyuki Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Kuwata
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Kaba
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideaki Okuyama
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshitaka Kawai
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nao Hiwatashi
- Department of Otolaryngology, Kyoto-Katsura Hospital, Kyoto, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Tatsunori Sakamoto
- Department of Otorhinolaryngology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
4
|
Zhou Q, Ye X, Ran Q, Kitahara A, Matsumoto Y, Moriyama M, Ajioka Y, Saijo Y. Trachea Engineering Using a Centrifugation Method and Mouse-Induced Pluripotent Stem Cells. Tissue Eng Part C Methods 2019; 24:524-533. [PMID: 30101671 DOI: 10.1089/ten.tec.2018.0115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The outcomes of tracheal transplantation for the treatment of airway stenosis are unsatisfactory. We investigated the feasibility of regeneration of the trachea using a rat decellularized tracheal scaffold and mouse-induced pluripotent stem (iPS) cells for in vivo transplantation. The rat trachea was first decellularized using a detergent/enzymatic treatment method. We successfully established a centrifugation method that can transplant cells onto the luminal surface of the decellularized rat tracheal scaffold circumferentially. Two types of mouse iPS cells were differentiated into definitive endoderm cells and transplanted onto the luminal surface of the decellularized tracheal matrix scaffold using this centrifugation method. For in vivo study, normal rat tracheas, no-cell rat tracheal scaffolds, or rat tracheal scaffolds recellularized with rat tracheal epithelial cells (EGV-4T) were orthotopically transplanted on F344 rats, and rat tracheal scaffolds recellularized with mouse iPS cells were transplanted on F344/NJc1-rnu/rnu rats. Rats transplanted with no-cell scaffolds or scaffolds recellularized with EGV-4T survived for 1 month, although airway stenosis was observed. One of the F344/NJc1-rnu/rnu rats transplanted with rat trachea regenerated using mouse iPS cells survived over 5 weeks. Histological analysis indicated the cause of death was airway stenosis due to colonic cellular proliferation of undifferentiated iPS cells. Re-epithelialization with numerous ciliated epithelial cells was observed in one of the rats transplanted with trachea bioengineered using iPS cells. In this study, we present a simple and efficient tracheal tissue engineering model using a centrifugation method in a small-animal model. Tissue-engineered trachea using decellularized tracheal scaffolds and iPS cells is potentially applicable for tracheal transplantation.
Collapse
Affiliation(s)
- Qiliang Zhou
- 1 Department of Medical Oncology and Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| | - Xulu Ye
- 1 Department of Medical Oncology and Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| | - Qingsong Ran
- 1 Department of Medical Oncology and Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| | - Akihiko Kitahara
- 2 Department of Thoracic Surgery, Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| | - Yoshifumi Matsumoto
- 1 Department of Medical Oncology and Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| | - Masato Moriyama
- 1 Department of Medical Oncology and Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| | - Yoichi Ajioka
- 3 Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| | - Yasuo Saijo
- 1 Department of Medical Oncology and Niigata University Graduate School of Medical and Dental Sciences , Niigata, Japan
| |
Collapse
|
5
|
Kehl D, Görtz S, Wang C, Hoerstrup SP, Bleul U, Weber B. Gestational Age-Dependent Fetal Fluid Dynamics in the Ovine Developmental Model: Establishment of Surrogate Markers for the Differentiation of Stem Cell Origin. Cells Tissues Organs 2019; 206:208-217. [PMID: 31079095 DOI: 10.1159/000499504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/26/2019] [Indexed: 11/19/2022] Open
Abstract
The ovine developmental model represents the standard in vivo model for studies involving maternofetal physiology, amniotic fluid (AF) research, and fetal cell therapy prior to human clinical use. Although being close to the human fetal anatomy, 2 separate extraembryonic fluid compartments remain during gestation, known as the amnion and the allantois. A clear distinction between AF versus allantoic fluid (AL) is therefore indispensable for correct scientific conclusions with regard to human translation. In the presented study, the biochemical composition of AF and AL was evaluated in ovine gravid uteri postmortem (n = 31) over the entire gestation. Four parameters, consisting of Na+, Cl-, Mg2+, and total protein, have been found to allow for specific discrimination of the 2 fetal fluids at all gestational phases and therefore as potential surrogate parameters for gestational age. In addition, volumetric changes of the developing fetus and the 2 fetal fluid cavities were analyzed by contrast-enhanced computed tomography (n = 12). AF showed a significant, linear volumetric increase over gestation, whereas AL volume maintained relatively static independent of gestational age. These results serve as a basis for future studies by providing surrogate markers enabling a reliable distinction of isolated fetal fluids and contained cells in the ovine developmental model over the entire gestation.
Collapse
Affiliation(s)
- Debora Kehl
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Sabrina Görtz
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Craig Wang
- Department of Mathematics, University of Zurich, Zurich, Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Wyss Zurich, University of Zurich, and ETH Zurich, Zurich, Switzerland
| | - Ulrich Bleul
- Clinic of Reproductive Medicine, Department of Farm Animals, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Benedikt Weber
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland, .,Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland, .,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland, .,Skin and Endothelium Research Division (SERD), Department of Dermatology, Medical University of Vienna, Vienna, Austria,
| |
Collapse
|
6
|
Barboni B, Russo V, Berardinelli P, Mauro A, Valbonetti L, Sanyal H, Canciello A, Greco L, Muttini A, Gatta V, Stuppia L, Mattioli M. Placental Stem Cells from Domestic Animals: Translational Potential and Clinical Relevance. Cell Transplant 2019; 27:93-116. [PMID: 29562773 PMCID: PMC6434480 DOI: 10.1177/0963689717724797] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The field of regenerative medicine is moving toward clinical practice in veterinary science. In this context, placenta-derived stem cells isolated from domestic animals have covered a dual role, acting both as therapies for patients and as a valuable cell source for translational models. The biological properties of placenta-derived cells, comparable among mammals, make them attractive candidates for therapeutic approaches. In particular, stemness features, low immunogenicity, immunomodulatory activity, multilineage plasticity, and their successful capacity for long-term engraftment in different host tissues after autotransplantation, allo-transplantation, or xenotransplantation have been demonstrated. Their beneficial regenerative effects in domestic animals have been proven using preclinical studies as well as clinical trials starting to define the mechanisms involved. This is, in particular, for amniotic-derived cells that have been thoroughly studied to date. The regenerative role arises from a mutual tissue-specific cell differentiation and from the paracrine secretion of bioactive molecules that ultimately drive crucial repair processes in host tissues (e.g., anti-inflammatory, antifibrotic, angiogenic, and neurogenic factors). The knowledge acquired so far on the mechanisms of placenta-derived stem cells in animal models represent the proof of concept of their successful use in some therapeutic treatments such as for musculoskeletal disorders. In the next future, legislation in veterinary regenerative medicine will be a key element in order to certify those placenta-derived cell-based protocols that have already demonstrated their safety and efficacy using rigorous approaches and to improve the degree of standardization of cell-based treatments among veterinary clinicians.
Collapse
Affiliation(s)
- B Barboni
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Russo
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - P Berardinelli
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Mauro
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Valbonetti
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - H Sanyal
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Canciello
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Greco
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Muttini
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Gatta
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Stuppia
- 2 Medical Genetics, University "G. d'Annunzio" of Chieti Pescara, Chieti, Italy
| | - M Mattioli
- 3 Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale," Teramo, Italy
| |
Collapse
|
7
|
Decellularization Concept in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:71-85. [DOI: 10.1007/5584_2019_338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
8
|
Abbaspanah B, Momeni M, Ebrahimi M, Mousavi SH. Advances in perinatal stem cells research: a precious cell source for clinical applications. Regen Med 2018; 13:595-610. [PMID: 30129876 DOI: 10.2217/rme-2018-0019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Perinatal tissues possess numerous types of stem (stromal) cells, which are considered effective candidates for cell therapy. These tissues possess common characteristics of both embryonic and adult stem cells, and cell therapists have begun to use perinatal stem cells to treat several diseases. Despite their benefits, these cells are considered biological waste and usually discarded after delivery. This review highlights the characteristics and potential clinical applications in regenerative medicine of perinatal stem cell sources - cord blood hematopoietic stem cells, umbilical cord mesenchymal stem cells, amniotic membrane stem cells, amniotic fluid stem cells, amniotic epithelial cells and chorionic mesenchymal stem cells.
Collapse
Affiliation(s)
| | - Maryam Momeni
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran.,Department of Stem Cells & Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran
| | - Seyed Hadi Mousavi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
|
10
|
Abstract
Trachea replacement for nonoperable defects remains an unsolved problem due to complications with stenosis and mechanical insufficiency. While native trachea has anisotropic mechanical properties, the vast majority of engineered constructs focus on uniform cartilaginous-like conduits. These conduits often lack quantitative mechanical analysis at the construct level, which limits analysis of functional outcomes in vivo, as well as comparisons across studies. This review aims to present a clear picture of native tracheal mechanics at the tissue and organ level, as well as loading conditions to establish design criteria for trachea replacements. We further explore the implications of failing to match native properties with regards to implant collapse, stenosis, and infection.
Collapse
Affiliation(s)
- Elizabeth M Boazak
- Department of Biomedical Engineering, The City College of New York, Steinman Hall, 160 Convent Avenue, New York, New York 10031, United States
| | - Debra T Auguste
- Department of Biomedical Engineering, The City College of New York, Steinman Hall, 160 Convent Avenue, New York, New York 10031, United States.,Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
11
|
Lee JS, Choi YS, Cho SW. Decellularized Tissue Matrix for Stem Cell and Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:161-180. [DOI: 10.1007/978-981-13-0445-3_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
12
|
Boazak EM, Benson JM, Auguste DT. R- and Z-Axis Patterned Scaffolds Mimic Tracheal Circumferential Compliance and Longitudinal Extensibility. ACS Biomater Sci Eng 2017; 3:3222-3229. [DOI: 10.1021/acsbiomaterials.7b00641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Elizabeth M. Boazak
- Department of Biomedical
Engineering, The City College of New York, Steinman Hall, 160 Convent Avenue, New York, New York 10031, United States
| | - Jamie M. Benson
- Department of Biomedical
Engineering, The City College of New York, Steinman Hall, 160 Convent Avenue, New York, New York 10031, United States
| | - Debra T. Auguste
- Department of Biomedical
Engineering, The City College of New York, Steinman Hall, 160 Convent Avenue, New York, New York 10031, United States
| |
Collapse
|
13
|
Abstract
Purpose of Review There is no consensus on the best technology to be employed for tracheal replacement. One particularly promising approach is based upon tissue engineering and involves applying autologous cells to transplantable scaffolds. Here, we present the reported pre-clinical and clinical data exploring the various options for achieving such seeding. Recent Findings Various cell combinations, delivery strategies, and outcome measures are described. Mesenchymal stem cells (MSCs) are the most widely employed cell type in tracheal bioengineering. Airway epithelial cell luminal seeding is also widely employed, alone or in combination with other cell types. Combinations have thus far shown the greatest promise. Chondrocytes may improve mechanical outcomes in pre-clinical models, but have not been clinically tested. Rapid or pre-vascularization of scaffolds is an important consideration. Overall, there are few published objective measures of post-seeding cell viability, survival, or overall efficacy. Summary There is no clear consensus on the optimal cell-scaffold combination and mechanisms for seeding. Systematic in vivo work is required to assess differences between tracheal grafts seeded with combinations of clinically deliverable cell types using objective outcome measures, including those for functionality and host immune response. Electronic supplementary material The online version of this article (10.1007/s40778-017-0108-2) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Kehl D, Generali M, Görtz S, Geering D, Slamecka J, Hoerstrup SP, Bleul U, Weber B. Amniotic Fluid Cells Show Higher Pluripotency-Related Gene Expression Than Allantoic Fluid Cells. Stem Cells Dev 2017; 26:1424-1437. [DOI: 10.1089/scd.2016.0352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Debora Kehl
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Melanie Generali
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Sabrina Görtz
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Diego Geering
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Jaroslav Slamecka
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Ulrich Bleul
- Clinic of Reproductive Medicine, Department of Farm Animals, Vetsuisse-Faculty University of Zurich, Zurich, Switzerland
| | - Benedikt Weber
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Jakobsen KK, Grønhøj C, Jensen DH, Fischer-Nielsen A, Hjuler T, von Buchwald C. Mesenchymal stem cell therapy for laryngotracheal stenosis: A systematic review of preclinical studies. PLoS One 2017; 12:e0185283. [PMID: 28934345 PMCID: PMC5608394 DOI: 10.1371/journal.pone.0185283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/08/2017] [Indexed: 11/18/2022] Open
Abstract
Background Laryngotracheal stenosis (LTS) can be either congenital or acquired. Laryngeal stenosis is most often encountered after prolonged intubation. The mechanism for stenosis following intubation is believed to be hypertrophic scarring. Mesenchymal stem cells (MSCs) therapy has shown promising results in regenerative medicine. We aimed to systematically review the literature on MSC therapy for stenosis of the conductive airways. Methods PubMed, EMBASE, Google Scholar and the Cochrane Library were systematically searched from January 1980–January 2017 with the purpose of identifying all studies addressing the effect of MSC therapy on the airway. We assessed effect on inflammation, fibrosis, and MSC as a component in tissue engineering for treating defects in the airway. Results We identified eleven studies (n = 256 animals) from eight countries evaluating the effect of MSCs as a regenerative therapy in the upper airways. The studies indicate that MSC therapy may lead to a more constructive inflammatory response as well as support tissue regeneration. Conclusion There may be a favorable effect of MSCs in inhibiting inflammation and as a component in tissue engineering. Given the heterogeneous nature of the included animal studies, any clear conclusion regarding the effect of tracheal stenosis in human subjects cannot be drawn. The included preclinical studies are however encouraging for further research.
Collapse
Affiliation(s)
- Kathrine Kronberg Jakobsen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Christian Grønhøj
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - David H. Jensen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Cell Therapy Facility, Blood Bank, Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Hjuler
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Christian von Buchwald
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
16
|
Xu Y, Li D, Yin Z, He A, Lin M, Jiang G, Song X, Hu X, Liu Y, Wang J, Wang X, Duan L, Zhou G. Tissue-engineered trachea regeneration using decellularized trachea matrix treated with laser micropore technique. Acta Biomater 2017; 58:113-121. [PMID: 28546133 DOI: 10.1016/j.actbio.2017.05.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 04/15/2017] [Accepted: 05/04/2017] [Indexed: 01/12/2023]
Abstract
Tissue-engineered trachea provides a promising approach for reconstruction of long segmental tracheal defects. However, a lack of ideal biodegradable scaffolds greatly restricts its clinical translation. Decellularized trachea matrix (DTM) is considered a proper scaffold for trachea cartilage regeneration owing to natural tubular structure, cartilage matrix components, and biodegradability. However, cell residual and low porosity of DTM easily result in immunogenicity and incomplete cartilage regeneration. To address these problems, a laser micropore technique (LMT) was applied in the current study to modify trachea sample porosity to facilitate decellular treatment and cell ingrowth. Decellularization processing demonstrated that cells in LMT treated samples were more easily removed compared with untreated native trachea. Furthermore, after optimizing the protocols of LMT and decellular treatments, the LMT-treated DTM (LDTM) could retain their original tubular shape with only mild extracellular matrix damage. After seeding with chondrocytes and culture in vitro for 8 weeks, the cell-LDTM constructs formed tubular cartilage with relatively homogenous cell distribution in both micropores and bilateral surfaces. In vivo results further confirmed that the constructs could form mature tubular cartilage with increased DNA and cartilage matrix contents, as well as enhanced mechanical strength, compared with native trachea. Collectively, these results indicate that LDTM is an ideal scaffold for tubular cartilage regeneration and, thus, provides a promising strategy for functional reconstruction of trachea cartilage. STATEMENT OF SIGNIFICANCE Lacking ideal biodegradable scaffolds greatly restricts development of tissue-engineered trachea. Decellularized trachea matrix (DTM) is considered a proper scaffold for trachea cartilage regeneration. However, cell residual and low porosity of DTM easily result in immunogenicity and incomplete cartilage regeneration. By laser micropore technique (LMT), the current study efficiently enhanced the porosity and decellularized efficacy of DTM. The LMT-treated DTM basically retained the original tubular shape with mild matrix damage. After chondrocyte seeding followed by in vitro culture and in vivo implantation, the constructs formed mature tubular cartilage with matrix content and mechanical strength similar to native trachea. The current study provides an ideal scaffold and a promising strategy for cartilage regeneration and functional reconstruction of trachea.
Collapse
|
17
|
Virk JS, Zhang H, Nouraei R, Sandhu G. Prosthetic reconstruction of the trachea: A historical perspective. World J Clin Cases 2017; 5:128-133. [PMID: 28470004 PMCID: PMC5395980 DOI: 10.12998/wjcc.v5.i4.128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/17/2017] [Accepted: 02/20/2017] [Indexed: 02/05/2023] Open
Abstract
This review discusses the history of tracheal reconstruction; from early work to future challenges. The focus is primarily on prosthetic tracheal reconstruction in the form of intraluminal stents, patch repairs, circumferential repairs and replacement of the trachea. A historical perspective of materials used such as foreign materials, autografts, allografts, xenografts and techniques, along with their advantages and disadvantages, is provided.
Collapse
|
18
|
Chiang T, Pepper V, Best C, Onwuka E, Breuer CK. Clinical Translation of Tissue Engineered Trachea Grafts. Ann Otol Rhinol Laryngol 2016; 125:873-885. [PMID: 27411362 DOI: 10.1177/0003489416656646] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To provide a state-of-the-art review discussing recent achievements in tissue engineered tracheal reconstruction. DATA SOURCES AND REVIEW METHODS A structured PubMed search of the current literature up to and including October 2015. Representative articles that discuss the translation of tissue engineered tracheal grafts (TETG) were reviewed. CONCLUSIONS The integration of a biologically compatible support with autologous cells has resulted in successful regeneration of respiratory epithelium, cartilage, and vascularization with graft patency, although the optimal construct composition has yet to be defined. Segmental TETG constructs are more commonly complicated by stenosis and delayed epithelialization when compared to patch tracheoplasty. IMPLICATIONS FOR PRACTICE The recent history of human TETG recipients represents revolutionary proof of principle studies in regenerative medicine. Application of TETG remains limited to a compassionate use basis; however, defining the mechanisms of cartilage formation, epithelialization, and refinement of in vivo regeneration will advance the translation of TETG from the bench to the bedside.
Collapse
Affiliation(s)
- Tendy Chiang
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA Department of Pediatric Otolaryngology, Nationwide Children's Hospital, Columbus, Ohio, USA Department of Otolaryngology-Head & Neck Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Victoria Pepper
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Cameron Best
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Ekene Onwuka
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher K Breuer
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
19
|
Mansfield EG, Greene VK, Auguste DT. Patterned, tubular scaffolds mimic longitudinal and radial mechanics of the neonatal trachea. Acta Biomater 2016; 33:176-82. [PMID: 26821338 DOI: 10.1016/j.actbio.2016.01.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 01/08/2016] [Accepted: 01/23/2016] [Indexed: 11/30/2022]
Abstract
Tracheal damage, abnormality or absence can result from the growth of tumors or from Congenital High Airway Obstruction Syndrome. No optimal or routine treatment has been established for tracheal repair, despite numerous attempts with natural and artificial prostheses. The fetal trachea is comprised of cartilaginous rings connected by an elastomeric tissue. In an effort to design an engineered trachea replacement, we have synthesized 2-hydroxyethyl methacrylate hydrogels with moduli of 67 ± 3.1 kPa (soft) and 13.0 ± 1.8 MPa (hard). Given the criteria for longitudinal extensibility and lateral rigidity applied during respiration, we evaluated a series of patterned hydrogels with different sizes of hard and soft segments to mimic fetal tracheas. A 1:2 ratio of soft:hard segments resulted in a construct capable of 11.0 ± 1% extension within the elastic range. Tubular constructs with this ratio required similar load/length for cyclic compression as ovine trachea samples. Achieving biomimetic mechanical properties in a trachea replacement may be essential for achieving normal respiration in recipient patients. STATEMENT OF SIGNIFICANCE Fetal abnormalities or tumors can result in tracheal absence or damage. Despite numerous attempts with natural and artificial replacements, there is still no routine treatment for tracheal repair. The literature recognizes the importance of tracheal lateral rigidity and longitudinal extensibility for normal respiration. Achieving closely matched mechanical properties may provide proper function and help decrease implant fibrosis and subsequent occlusion. In this study, we evaluated the mechanics of a series of patterned, tubular hydrogels with different ratios of hard and soft segments to mimic alternating cartilage and ligament sections in fetal tracheas. We compared our results to that of sheep trachea. This is the first report to assess both radial rigidity and longitudinal extensibility in an engineered trachea construct.
Collapse
Affiliation(s)
- Elizabeth G Mansfield
- The City College of New York, Department of Biomedical Engineering, Steinman Hall Room 508, 160 Convent Avenue, New York, NY 10031, USA.
| | - Vaughn K Greene
- The City College of New York, Department of Biomedical Engineering, Steinman Hall Room 508, 160 Convent Avenue, New York, NY 10031, USA.
| | - Debra T Auguste
- The City College of New York, Department of Biomedical Engineering, Steinman Hall Room 508, 160 Convent Avenue, New York, NY 10031, USA.
| |
Collapse
|
20
|
Tian Y, Tao L, Zhao S, Tai D, Liu D, Liu P. Isolation and morphological characterization of ovine amniotic fluid mesenchymal stem cells. Exp Anim 2015; 65:125-34. [PMID: 26616638 PMCID: PMC4873481 DOI: 10.1538/expanim.15-0031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the most promising cell populations for tissue engineering and regenerative medicine. Of utmost importance to MSC research is identification of MSC sources that are easily obtainable and stable. Several studies have shown that MSCs can be isolated from amniotic fluid. The sheep is one of the main types of farm animal, and it has many biophysical and biochemical similarities to humans. Here, we obtained MSCs from ovine amniotic fluid and determined the expansion capacity, surface and intracellular marker expression, karyotype, and multilineage differentiation ability of these ovine amniotic fluid mesenchymal stem cells (oAF-MSCs). Moreover, expression levels of differentiation markers were measured using reverse transcription-qPCR (RT-qPCR). Our phenotypic analysis shows that the isolated oAF-MSCs are indeed MSCs.
Collapse
Affiliation(s)
- Yunyun Tian
- College of Life Sciences, Inner Mongolia University, Inner Mongolia, Hohhot 010021, P.R.China
| | | | | | | | | | | |
Collapse
|
21
|
Stem cells from amniotic fluid--Potential for regenerative medicine. Best Pract Res Clin Obstet Gynaecol 2015; 31:45-57. [PMID: 26542929 DOI: 10.1016/j.bpobgyn.2015.08.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Abstract
Regenerative medicine has recently been established as an emerging field focussing on repair, replacement or regeneration of cells, tissues and whole organs. The significant recent advances in the field have intensified the search for novel sources of stem cells with potential for therapy. Recently, researchers have identified the amniotic fluid as an untapped source of stem cells that are multipotent, possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. Stem cells from the amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumours, which make them an ideal candidate for tissue engineering applications. In addition, their ability to engraft in injured organs and modulate immune and repair responses of host tissues suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases affecting major tissues/organs. This review summarises the evidence on amniotic fluid cells over the past 15 years and explores the potential therapeutic applications of amniotic fluid stem cells and amniotic fluid mesenchymal stem cells.
Collapse
|
22
|
Rana D, Zreiqat H, Benkirane-Jessel N, Ramakrishna S, Ramalingam M. Development of decellularized scaffolds for stem cell-driven tissue engineering. J Tissue Eng Regen Med 2015; 11:942-965. [PMID: 26119160 DOI: 10.1002/term.2061] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/22/2015] [Accepted: 05/04/2015] [Indexed: 12/19/2022]
Abstract
Organ transplantation is an effective treatment for chronic organ dysfunctioning conditions. However, a dearth of available donor organs for transplantation leads to the death of numerous patients waiting for a suitable organ donor. The potential of decellularized scaffolds, derived from native tissues or organs in the form of scaffolds has been evolved as a promising approach in tissue-regenerative medicine for translating functional organ replacements. In recent years, donor organs, such as heart, liver, lung and kidneys, have been reported to provide acellular extracellular matrix (ECM)-based scaffolds through the process called 'decellularization' and proved to show the potential of recellularization with selected cell populations, particularly with stem cells. In fact, decellularized stem cell matrix (DSCM) has also emerged as a potent biological scaffold for controlling stem cell fate and function during tissue organization. Despite the proven potential of decellularized scaffolds in tissue engineering, the molecular mechanism responsible for stem cell interactions with decellularized scaffolds is still unclear. Stem cells interact with, and respond to, various signals/cues emanating from their ECM. The ability to harness the regenerative potential of stem cells via decellularized ECM-based scaffolds has promising implications for tissue-regenerative medicine. Keeping these points in view, this article reviews the current status of decellularized scaffolds for stem cells, with particular focus on: (a) concept and various methods of decellularization; (b) interaction of stem cells with decellularized scaffolds; (c) current recellularization strategies, with associated challenges; and (iv) applications of the decellularized scaffolds in stem cell-driven tissue engineering and regenerative medicine. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Deepti Rana
- Centre for Stem Cell Research (CSCR), Institute for Stem Cell Biology and Regenerative Medicine (Bengaluru) Christian Medical College Campus, Vellore, India
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, Faculty of Engineering and Bosch Institute, University of Sydney, NSW, Australia
| | - Nadia Benkirane-Jessel
- INSERM, Osteoarticular and Dental Regenerative Nanomedicine Laboratory, UMR 1109, Faculté de Médecine, Strasbourg, France
| | - Seeram Ramakrishna
- Centre for Nanofibres and Nanotechnology, Department of Mechanical Engineering, National University of Singapore
| | - Murugan Ramalingam
- Centre for Stem Cell Research (CSCR), Institute for Stem Cell Biology and Regenerative Medicine (Bengaluru) Christian Medical College Campus, Vellore, India
- WPI Advanced Institute for Materials Research, Tohoku University, Sendai, Japan
| |
Collapse
|
23
|
Iacono E, Rossi B, Merlo B. Stem cells from foetal adnexa and fluid in domestic animals: an update on their features and clinical application. Reprod Domest Anim 2015; 50:353-64. [PMID: 25703812 DOI: 10.1111/rda.12499] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/15/2015] [Indexed: 12/25/2022]
Abstract
Over the past decade, stem cell research has emerged as an area of major interest for its potential in regenerative medicine applications. This is in constant need of new cell sources to conceive regenerative medicine approaches for diseases that are still without therapy. Scientists drew the attention towards alternative sources such as foetal adnexa and fluid, as these sources possess many advantages: first of all, cells can be extracted from discarded foetal material and it is non-invasive and inexpensive for the patient; secondly, abundant stem cells can be obtained; and finally, these stem cell sources are free from ethical considerations. Cells derived from foetal adnexa and fluid preserve some of the characteristics of the primitive embryonic layers from which they originate. Many studies have demonstrated the differentiation potential in vitro and in vivo towards mesenchymal and non-mesenchymal cell types; in addition, the immune-modulatory properties make these cells a good candidate for allo- and xenotransplantation. Naturally occurring diseases in domestic animals can be more ideal as disease model of human genetic and acquired diseases and could help to define the potential therapeutic use efficiency and safety of stem cells therapies. This review offers an update on the state of the art of characterization of domestic animals' MSCs derived from foetal adnexa and fluid and on the latest findings in pre-clinical or clinical setting of the stem cell populations isolated from these sources.
Collapse
Affiliation(s)
- E Iacono
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia (Bo), Italy
| | | | | |
Collapse
|
24
|
Pennington EC, Dionigi B, Gray FL, Ahmed A, Brazzo J, Dolinko A, Calderon N, Darrah T, Zurakowski D, Nazarian A, Snyder B, Fauza DO. Limb reconstruction with decellularized, non-demineralized bone in a young leporine model. ACTA ACUST UNITED AC 2015; 10:015021. [PMID: 25668190 DOI: 10.1088/1748-6041/10/1/015021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Limb salvage from a variety of pathological processes in children is often limited by the unavailability of optimal allograft bone, or an appropriate structural bone substitute. In this study, we sought to examine a practical alternative for pediatric limb repair, based on decellularized, non-demineralized bone grafts, and to determine whether controlled recellularization prior to implantation has any impact on outcome. Growing New Zealand rabbits (n = 12) with a complete, critical-size defect on the left tibiofibula were equally divided into two groups. One group received a decellularized, non-demineralized leporine tibiofibula graft. The other group received an equivalent graft seeded with mesenchymal stem cells labeled with green fluorescent protein (GFP), at a fixed density. Animals were euthanized at comparable time points 3-8 weeks post-implantation. Statistical analysis was by the Student t-test and Fisher's exact test (P < 0.05). There was no significant difference in the rate of non-union between the two groups, including on 3D micro-CT. Incorporated grafts achieved adequate axial bending rigidity, torsional rigidity, union yield and flexural strength, with no significant differences or unequal variances between the groups. Correspondingly, there were no significant differences in extracellular calcium levels, or alkaline phosphatase activity. Histology confirmed the presence of neobone in both groups, with GFP-positive cells in the recellularized grafts. It was shown that osseous grafts derived from decellularized, non-demineralized bone undergo adequate remodeling in vivo after the repair of critical-size limb defects in a growing leporine model, irrespective of subsequent recellularization. This methodology may become a practical alternative for pediatric limb reconstruction.
Collapse
Affiliation(s)
- Elliot C Pennington
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Farmer DL. Standing on the shoulders of giants: a scientific journey from Singapore to stem cells. J Pediatr Surg 2015; 50:15-22. [PMID: 25598087 DOI: 10.1016/j.jpedsurg.2014.10.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 10/06/2014] [Indexed: 12/18/2022]
Abstract
Cellular therapy was introduced in the early 1980s as adoptive immunotherapy for cancer and has now expanded to stem cell treatment for a wide variety of indications. During the same period, the concept of the fetus as a patient evolved from fantasy to everyday reality. The intersection of these two fields offers great potential for cures in childhood diseases. The fetal treatment of spina bifida is one such disease. Global surgery has also emerged as a cost effective approach to reducing the worldwide burden of childhood disease.
Collapse
Affiliation(s)
- Diana Lee Farmer
- Department of Surgery, UC Davis Children's Hospital, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
26
|
Weiss DJ, Elliott M, Jang Q, Poole B, Birchall M. Tracheal bioengineering: the next steps. Proceeds of an International Society of Cell Therapy Pulmonary Cellular Therapy Signature Series Workshop, Paris, France, April 22, 2014. Cytotherapy 2014; 16:1601-13. [PMID: 25457172 DOI: 10.1016/j.jcyt.2014.10.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 11/15/2022]
Abstract
There has been significant and exciting recent progress in the development of bioengineering approaches for generating tracheal tissue that can be used for congenital and acquired tracheal diseases. This includes a growing clinical experience in both pediatric and adult patients with life-threatening tracheal diseases. However, not all of these attempts have been successful, and there is ongoing discussion and debate about the optimal approaches to be used. These include considerations of optimal materials, particularly use of synthetic versus biologic scaffolds, appropriate cellularization of the scaffolds, optimal surgical approaches and optimal measure of both clinical and biologic outcomes. To address these issues, the International Society of Cell Therapy convened a first-ever meeting of the leading clinicians and tracheal biologists, along with experts in regulatory and ethical affairs, to discuss and debate the issues. A series of recommendations are presented for how to best move the field ahead.
Collapse
Affiliation(s)
- Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Martin Elliott
- Department of Cardiothoracic Surgery, Great Ormond Street Hospital, London, United Kingdom
| | - Queenie Jang
- International Society for Cell Therapy, Vancouver, British Columbia, Canada
| | - Brian Poole
- International Society for Cell Therapy, Vancouver, British Columbia, Canada
| | - Martin Birchall
- Royal National Throat Nose, and Ear Hospital and University College London, London, United Kingdom.
| |
Collapse
|
27
|
Abstract
Regenerative medicine is an alternative solution for organ transplantation. Stem cells and nanoscaffolds are two essential components in regenerative medicine. Mesenchymal stem cells (MSCs) are considered as primary adult stem cells with high proliferation capacity, wide differentiation potential, and immunosuppression properties which make them unique for regenerative medicine and cell therapy. Scaffolds are engineered nanofibers that provide suitable microenvironment for cell signalling which has a great influence on cell proliferation, differentiation, and biology. Recently, application of scaffolds and MSCs is being utilized in obtaining more homogenous population of MSCs with higher cell proliferation rate and greater differentiation potential, which are crucial factors in regenerative medicine. In this review, the definition, biology, source, characterization, and isolation of MSCs and current report of application of nanofibers in regenerative medicine in different lesions are discussed.
Collapse
|
28
|
Tissue engineered scaffolds for an effective healing and regeneration: reviewing orthotopic studies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:398069. [PMID: 25250319 PMCID: PMC4163448 DOI: 10.1155/2014/398069] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/22/2014] [Indexed: 12/20/2022]
Abstract
It is commonly stated that tissue engineering is the most promising approach to treat or replace failing tissues/organs. For this aim, a specific strategy should be planned including proper selection of biomaterials, fabrication techniques, cell lines, and signaling cues. A great effort has been pursued to develop suitable scaffolds for the restoration of a variety of tissues and a huge number of protocols ranging from in vitro to in vivo studies, the latter further differentiating into several procedures depending on the type of implantation (i.e., subcutaneous or orthotopic) and the model adopted (i.e., animal or human), have been developed. All together, the published reports demonstrate that the proposed tissue engineering approaches spread toward multiple directions. The critical review of this scenario might suggest, at the same time, that a limited number of studies gave a real improvement to the field, especially referring to in vivo investigations. In this regard, the present paper aims to review the results of in vivo tissue engineering experimentations, focusing on the role of the scaffold and its specificity with respect to the tissue to be regenerated, in order to verify whether an extracellular matrix-like device, as usually stated, could promote an expected positive outcome.
Collapse
|
29
|
Pennington EC, Rialon KL, Dionigi B, Ahmed A, Zurakowski D, Fauza DO. The impact of gestational age on targeted amniotic cell profiling in experimental neural tube defects. Fetal Diagn Ther 2014; 37:65-9. [PMID: 25171576 DOI: 10.1159/000362811] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/09/2014] [Indexed: 11/19/2022]
Abstract
PURPOSE The proportions of select stem cells in term amniotic fluid have been shown to correlate with the type and size of experimental neural tube defects (NTDs). We sought to determine the impact of gestational age upon this form of targeted amniotic cell profiling. METHODS Sprague-Dawley fetuses with retinoic acid-induced NTDs (n = 110) underwent amniotic fluid procurement at four time points in gestation. Samples were analyzed by flow cytometry for the presence of cells concomitantly expressing Nestin and Sox-2 (neural stem cells, aNSCs) and cells concomitantly expressing CD29 and CD44 (mesenchymal stem cells, aMSCs). Statistical analysis was by nonparametric Kruskal-Wallis ANOVA (p < 0.05). RESULTS There was a statistically significant impact of gestational age on the proportions of both aMSCs (p = 0.01) and aNSCs (p < 0.01) in fetuses with isolated spina bifida. No such impact was noted in normal fetuses (p > 0.10 for both cells), in isolated exencephaly (p > 0.10 for both cells), or in combination defects (p > 0.10 for both cells). Gestational age had no effect on aNSC/aMSC ratios. CONCLUSIONS Targeted quantitative amniotic cell profiling varies with gestational age in experimental isolated spina bifida. This finding should be considered prior to the eventual translation of this diagnostic adjunct into the prenatal evaluation of these anomalies. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Elliot C Pennington
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Mass., USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Significant advances in the field of regenerative medicine have intensified the search for novel sources of stem cells with potential for therapy. Although embryonic and adult tissues can be used for the isolation of pluripotent stem cells, significant limitations including ethical concerns, complexity of isolation/culture and tumorigenicity have hindered translation of laboratory findings to clinical practice.
Collapse
|
31
|
Babic AM, Jang S, Nicolov E, Voicu H, Luckey CJ. Culture of mouse amniotic fluid-derived cells on irradiated STO feeders results in the generation of primitive endoderm cell lines capable of self-renewal in vitro. Cells Tissues Organs 2013; 198:111-26. [PMID: 24060676 DOI: 10.1159/000353942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2013] [Indexed: 12/14/2022] Open
Abstract
The cells present in amniotic fluid (AF) are currently used for prenatal diagnosis of fetal anomalies but are also a potential source of cells for cell therapy. To better characterize putative progenitor cell populations present in AF, we used culture conditions that support self-renewal to determine if these promoted the generation of stable cell lines from AF-derived cells (AFC). Cells isolated from E11.5 mouse were cultured on irradiated STO fibroblast feeder layers in human embryonic germ cell derivation conditions. The cultures grew multicellular epithelial colonies that could be repropagated from single cells. Reverse transcription semiquantitative polymerase chain reaction of established cell lines revealed that they belonged to the extraembryonic endoderm (ExEn) expressing high levels of Gata6, Gata4, Sox17, Foxa2 and Sox7 mRNA. Hierarchical clustering based on the whole transcriptome expression profile of the AFC lines (AFCL) shows significant correlation between transcription profiles of AFCL and blastocyst-derived XEN, an ExEn cell line. In vitro differentiation of AFCL results in the generation of cells expressing albumin and α-fetoprotein (AFP), while intramuscular injection of AFCL into immunodeficient mice produced AFP-positive tumors with primitive endodermal appearance. Hence, E11.5 mouse AF contains cells that efficiently produce XEN lines. These AF-derived XEN lines do not spontaneously differentiate into embryonic-type cells but are phenotypically stable and have the capacity for extensive expansion. The lack of requirement for reprogramming factors to turn AF-derived progenitor cells into stable cell lines capable of massive expansion together with the known ability of ExEn to contribute to embryonic tissue suggests that this cell type may be a candidate for banking for cell therapies.
Collapse
Affiliation(s)
- Aleksandar M Babic
- Department of Pathology and Genomic Medicine, The Methodist Hospital, Houston, Tex., USA
| | | | | | | | | |
Collapse
|
32
|
Di Tomo P, Pipino C, Lanuti P, Morabito C, Pierdomenico L, Sirolli V, Bonomini M, Miscia S, Mariggiò MA, Marchisio M, Barboni B, Pandolfi A. Calcium sensing receptor expression in ovine amniotic fluid mesenchymal stem cells and the potential role of R-568 during osteogenic differentiation. PLoS One 2013; 8:e73816. [PMID: 24040082 PMCID: PMC3767786 DOI: 10.1371/journal.pone.0073816] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 07/24/2013] [Indexed: 02/07/2023] Open
Abstract
Amniotic fluid-derived stem (AFS) cells have been identified as a promising source for cell therapy applications in bone traumatic and degenerative damage. Calcium Sensing Receptor (CaSR), a G protein-coupled receptor able to bind calcium ions, plays a physiological role in regulating bone metabolism. It is expressed in different kinds of cells, as well as in some stem cells. The bone CaSR could potentially be targeted by allosteric modulators, in particular by agonists such as calcimimetic R-568, which may potentially be helpful for the treatment of bone disease. The aim of our study was first to investigate the presence of CaSR in ovine Amniotic Fluid Mesenchymal Stem Cells (oAFMSCs) and then the potential role of calcimimetics in in vitro osteogenesis. oAFMSCs were isolated, characterized and analyzed to examine the possible presence of CaSR by western blotting and flow cytometry analysis. Once we had demonstrated CaSR expression, we worked out that 1 µM R-568 was the optimal and effective concentration by cell viability test (MTT), cell number, Alkaline Phosphatase (ALP) and Alizarin Red S (ARS) assays. Interestingly, we observed that basal diffuse CaSR expression in oAFMSCs increased at the membrane when cells were treated with R-568 (1 µM), potentially resulting in activation of the receptor. This was associated with significantly increased cell mineralization (ALP and ARS staining) and augmented intracellular calcium and Inositol trisphosphate (IP3) levels, thus demonstrating a potential role for calcimimetics during osteogenic differentiation. Calhex-231, a CaSR allosteric inhibitor, totally reversed R-568 induced mineralization. Taken together, our results demonstrate for the first time that CaSR is expressed in oAFMSCs and that calcimimetic R-568, possibly through CaSR activation, can significantly improve the osteogenic process. Hence, our study may provide useful information on the mechanisms regulating osteogenesis in oAFMSCs, perhaps prompting the use of calcimimetics in bone regenerative medicine.
Collapse
Affiliation(s)
- Pamela Di Tomo
- Department of Experimental and Clinical Sciences, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Caterina Pipino
- Department of Experimental and Clinical Sciences, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Paola Lanuti
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Science, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Caterina Morabito
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Department of Neuroscience and Imaging, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Laura Pierdomenico
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Science, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Vittorio Sirolli
- Department of Medicine and Aging Science, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Mario Bonomini
- Department of Medicine and Aging Science, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Sebastiano Miscia
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Science, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Maria Addolorata Mariggiò
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Department of Neuroscience and Imaging, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Marco Marchisio
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Science, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Barbara Barboni
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Department of Comparative Biomedical Science, University of Teramo, Teramo, Italy
| | - Assunta Pandolfi
- Department of Experimental and Clinical Sciences, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “University G. d’Annunzio” Foundation Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- StemTeCh Group Chieti, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- * E-mail:
| |
Collapse
|
33
|
Vrana NE, Lavalle P, Dokmeci MR, Dehghani F, Ghaemmaghami AM, Khademhosseini A. Engineering functional epithelium for regenerative medicine and in vitro organ models: a review. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:529-43. [PMID: 23705900 DOI: 10.1089/ten.teb.2012.0603] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent advances in the fields of microfabrication, biomaterials, and tissue engineering have provided new opportunities for developing biomimetic and functional tissues with potential applications in disease modeling, drug discovery, and replacing damaged tissues. An intact epithelium plays an indispensable role in the functionality of several organs such as the trachea, esophagus, and cornea. Furthermore, the integrity of the epithelial barrier and its degree of differentiation would define the level of success in tissue engineering of other organs such as the bladder and the skin. In this review, we focus on the challenges and requirements associated with engineering of epithelial layers in different tissues. Functional epithelial layers can be achieved by methods such as cell sheets, cell homing, and in situ epithelialization. However, for organs composed of several tissues, other important factors such as (1) in vivo epithelial cell migration, (2) multicell-type differentiation within the epithelium, and (3) epithelial cell interactions with the underlying mesenchymal cells should also be considered. Recent successful clinical trials in tissue engineering of the trachea have highlighted the importance of a functional epithelium for long-term success and survival of tissue replacements. Hence, using the trachea as a model tissue in clinical use, we describe the optimal structure of an artificial epithelium as well as challenges of obtaining a fully functional epithelium in macroscale. One of the possible remedies to address such challenges is the use of bottom-up fabrication methods to obtain a functional epithelium. Modular approaches for the generation of functional epithelial layers are reviewed and other emerging applications of microscale epithelial tissue models for studying epithelial/mesenchymal interactions in healthy and diseased (e.g., cancer) tissues are described. These models can elucidate the epithelial/mesenchymal tissue interactions at the microscale and provide the necessary tools for the next generation of multicellular engineered tissues and organ-on-a-chip systems.
Collapse
Affiliation(s)
- Nihal E Vrana
- 1 Institut National de la Santé et de la Recherche Médicale , INSERM, UMR-S 1121, "Biomatériaux et Bioingénierie," Strasbourg Cedex, France
| | | | | | | | | | | |
Collapse
|
34
|
Liu J, Yang X, Shi W. Overexpression of CXCR4 in tracheal epithelial cells promotes their proliferation and migration to a stromal cell-derived factor-1 gradient. Exp Biol Med (Maywood) 2013; 238:144-50. [PMID: 23576796 DOI: 10.1177/1535370213477598] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Tracheal reconstruction has been an important issue in clinic, but it is limited for the ability of epithelial regeneration. Several reports have shown that stromal cell-derived factor-1 (SDF-1) and chemokine receptor CXCR4 play an important role in cell proliferation and migration of multiple cell types. But there is no report of SDF-1 and CXCR4 in tracheal cells. In this paper, the rat tracheal epithelial cells covered with cilium were isolated and cultured using two enzyme digestions, and CXCR4 lentivirus was constructed and infected to the tracheal cells successfully. The results showed that the expression of CXCR4 which was covered on cellular membrane majorly was low in normal cells, and the cell proliferation was increased accompanied with the increase in SDF-1 concentration. The cell proliferation, migration and intracellular free calcium were increased significantly in CXCR4 lentivirus infected groups in a dose-dependent manner, and these effects could be inhibited after CXCR4 inhibitor AMD3100 treated because the expression of CXCR4 was decreased. Our findings indicate that the activation of CXCR4 may promote tracheal cell proliferation and migration to the sites of airway injury where SDF-1 is regulated.
Collapse
Affiliation(s)
- Jun Liu
- Department of Thoracic Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | | | | |
Collapse
|