1
|
Lei C, Mei S, Zhou C, Xia C. Decellularized tracheal scaffolds in tracheal reconstruction: An evaluation of different techniques. J Appl Biomater Funct Mater 2021; 19:22808000211064948. [PMID: 34903089 DOI: 10.1177/22808000211064948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In humans, the trachea is a conduit for ventilation connecting the throat and lungs. However, certain congenital or acquired diseases may cause long-term tracheal defects that require replacement. Tissue engineering is considered a promising method to reconstruct long-segment tracheal lesions and restore the structure and function of the trachea. Decellularization technology retains the natural structure of the trachea, has good biocompatibility and mechanical properties, and is currently a hotspot in tissue engineering studies. This article lists various recent representative protocols for the generation of decellularized tracheal scaffolds (DTSs), as well as their validity and limitations. Based on the advancements in decellularization methods, we discussed the impact and importance of mechanical properties, revascularization, recellularization, and biocompatibility in the production and implantation of DTS. This review provides a basis for future research on DTS and its application in clinical therapy.
Collapse
Affiliation(s)
- Chenyang Lei
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Sheng Mei
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Chun Zhou
- Department of Geriatrics, The 903 Hospital of the Chinese People's Liberation Army Joint Logistics Support Force, Hangzhou, China
| | - Chen Xia
- Department of Orthopedic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
2
|
Farhat W, Chatelain F, Marret A, Faivre L, Arakelian L, Cattan P, Fuchs A. Trends in 3D bioprinting for esophageal tissue repair and reconstruction. Biomaterials 2020; 267:120465. [PMID: 33129189 DOI: 10.1016/j.biomaterials.2020.120465] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 02/08/2023]
Abstract
In esophageal pathologies, such as esophageal atresia, cancers, caustic burns, or post-operative stenosis, esophageal replacement is performed by using parts of the gastrointestinal tract to restore nutritional autonomy. However, this surgical procedure most often does not lead to complete functional recovery and is instead associated with many complications resulting in a decrease in the quality of life and survival rate. Esophageal tissue engineering (ETE) aims at repairing the defective esophagus and is considered as a promising therapeutic alternative. Noteworthy progress has recently been made in the ETE research area but strong challenges remain to replicate the structural and functional integrity of the esophagus with the approaches currently being developed. Within this context, 3D bioprinting is emerging as a new technology to facilitate the patterning of both cellular and acellular bioinks into well-organized 3D functional structures. Here, we present a comprehensive overview of the recent advances in tissue engineering for esophageal reconstruction with a specific focus on 3D bioprinting approaches in ETE. Current biofabrication techniques and bioink features are highlighted, and these are discussed in view of the complexity of the native esophagus that the designed substitute needs to replace. Finally, perspectives on recent strategies for fabricating other tubular organ substitutes via 3D bioprinting are discussed briefly for their potential in ETE applications.
Collapse
Affiliation(s)
- Wissam Farhat
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - François Chatelain
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - Auriane Marret
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France
| | - Lionel Faivre
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Unité de Thérapie Cellulaire, Hôpital Saint-Louis, Paris, France
| | - Lousineh Arakelian
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Unité de Thérapie Cellulaire, Hôpital Saint-Louis, Paris, France
| | - Pierre Cattan
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; Assistance Publique - Hôpitaux de Paris, Service de Chirurgie Digestive, Hôpital Saint-Louis, Paris, France
| | - Alexandra Fuchs
- Université de Paris, Inserm, U976 HIPI, F-75006, Paris, France; AP-HP, Hôpital Saint-Louis, 1 avenue Vellefaux, F-75010, Paris, France; CEA, IRIG, F-38000, Grenoble, France.
| |
Collapse
|
3
|
Dua KS, Sasikala M. Repairing the human esophagus with tissue engineering. Gastrointest Endosc 2018; 88:579-588. [PMID: 30220298 DOI: 10.1016/j.gie.2018.06.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Kulwinder S Dua
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Mitnala Sasikala
- Institute of Basic Sciences and Translational Research, Asian Healthcare Foundation, Asian Institute of Gastroenterology, Hyderabad, India
| |
Collapse
|
4
|
Ershadi R, Rahim M, Jahany S, Rakei S. Transplantation of the decellularized tracheal allograft in animal model (rabbit). Asian J Surg 2018; 41:328-332. [DOI: 10.1016/j.asjsur.2017.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/15/2017] [Accepted: 02/20/2017] [Indexed: 10/19/2022] Open
|
5
|
Duan H, Ma L, Liu H, Zhang Y, Zhang Z, Yan X, Li X. Tanshinone IIA attenuates epithelial-mesenchymal transition to inhibit the tracheal narrowing. J Surg Res 2016; 206:252-262. [PMID: 27916370 DOI: 10.1016/j.jss.2016.04.066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 04/05/2016] [Accepted: 04/28/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND This study examines the effects of tanshinone IIA (TIIA) on epithelial-mesenchymal transition (EMT) in tracheal transplantation and the ability of TIIA to inhibit tracheal narrowing after tracheal transplantation. Mechanisms that may be involved in this process are also explored. METHODS Human bronchial epithelial cells were treated in vitro with TGF-β1 for 72 h. The cells were pretreated with TIIA (40 μg/mL) or DMSO for 2 h before TGF-β1 stimulation. For the in vivo experiments, tracheas (5-6 rings) from Wistar rats were orthotopically transplanted into Sprague-Dawley rats. The experimental group received multiple infusions of sodium TIIA sulfonate (25 mg/kg, qd, intraperitoneally). The control group received infusions of the same volume of saline. Allografts were harvested at 3, 7, 10, 14, 35, and 90 d after transplantation and were examined for tracheal narrowing. Tracheal tissue samples and human bronchial epithelial cell were then subjected to further tests. RESULTS In the in vitro assay, epithelial cadherin expression was decreased after TGF-β1 stimulation, whereas α-smooth muscle actin and vimentin expression levels were increased. The expression levels of ZEB1 and Snail1 were also increased. These changes in expression were partially reversed by treatment with TIIA. In the in vivo assay, TIIA alleviated tracheal stenosis after tracheal allograft transplantation in rats and mitigated EMT by inhibiting the Smad signaling pathway and the expression of the transcription factors ZEB1 and Snail1. CONCLUSIONS Our research suggests that TIIA reduces tracheal narrowing after tracheal transplantation by suppressing TGF-β1-dependent EMT.
Collapse
Affiliation(s)
- Hongtao Duan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lijie Ma
- Department of Respiratory Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Honggang Liu
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yong Zhang
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhipei Zhang
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
6
|
Poghosyan T, Catry J, Luong-Nguyen M, Bruneval P, Domet T, Arakelian L, Sfeir R, Michaud L, Vanneaux V, Gottrand F, Larghero J, Cattan P. Esophageal tissue engineering: Current status and perspectives. J Visc Surg 2015; 153:21-9. [PMID: 26711880 DOI: 10.1016/j.jviscsurg.2015.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Tissue engineering, which consists of the combination and in vivo implantation of elements required for tissue remodeling toward a specific organ phenotype, could be an alternative for classical techniques of esophageal replacement. The current hybrid approach entails creation of an esophageal substitute composed of an acellular matrix and autologous epithelial and muscle cells provides the most successful results. Current research is based on the use of mesenchymal stem cells, whose potential for differentiation and proangioogenic, immune-modulator and anti-inflammatory properties are important assets. In the near future, esophageal substitutes could be constructed from acellular "intelligent matrices" that contain the molecules necessary for tissue regeneration; this should allow circumvention of the implantation step and still obtain standardized in vivo biological responses. At present, tissue engineering applications to esophageal replacement are limited to enlargement plasties with absorbable, non-cellular matrices. Nevertheless, the application of existing clinical techniques for replacement of other organs by tissue engineering in combination with a multiplication of translational research protocols for esophageal replacement in large animals should soon pave the way for health agencies to authorize clinical trials.
Collapse
Affiliation(s)
- T Poghosyan
- Service de chirurgie digestive, oncologique et métabolique, hôpital Ambroise-Paré, AP-HP, 92100 Boulogne-Billancourt, France; CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - J Catry
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service de chirurgie générale, digestive et endocrinienne, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - M Luong-Nguyen
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service de chirurgie générale, digestive et endocrinienne, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - P Bruneval
- Service d'anatomopathologie, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France
| | - T Domet
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Unité de thérapie cellulaire, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - L Arakelian
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - R Sfeir
- Centre de référence des affections congénitales et malformatives de l'œsophage, CHRU de Lille, 59000 Lille, France
| | - L Michaud
- Centre de référence des affections congénitales et malformatives de l'œsophage, CHRU de Lille, 59000 Lille, France
| | - V Vanneaux
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Unité de thérapie cellulaire, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - F Gottrand
- Centre de référence des affections congénitales et malformatives de l'œsophage, CHRU de Lille, 59000 Lille, France
| | - J Larghero
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Unité de thérapie cellulaire, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - P Cattan
- CIC de biothérapies, Inserm UMR 1160, institut universitaire d'hématologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service de chirurgie générale, digestive et endocrinienne, hôpital Saint-Louis, AP-HP, 75010 Paris, France.
| |
Collapse
|
7
|
Jungebluth P, Haag J, Macchiarini P. Regenerative Medizin. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2015. [DOI: 10.1007/s00398-014-1094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
8
|
|
9
|
Sjöqvist S, Jungebluth P, Lim ML, Haag JC, Gustafsson Y, Lemon G, Baiguera S, Burguillos MA, Del Gaudio C, Rodríguez AB, Sotnichenko A, Kublickiene K, Ullman H, Kielstein H, Damberg P, Bianco A, Heuchel R, Zhao Y, Ribatti D, Ibarra C, Joseph B, Taylor DA, Macchiarini P. Experimental orthotopic transplantation of a tissue-engineered oesophagus in rats. Nat Commun 2014; 5:3562. [PMID: 24736316 PMCID: PMC4354271 DOI: 10.1038/ncomms4562] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 03/05/2014] [Indexed: 12/22/2022] Open
Abstract
A tissue-engineered oesophageal scaffold could be very useful for the treatment of pediatric and adult patients with benign or malignant diseases such as carcinomas, trauma or congenital malformations. Here we decellularize rat oesophagi inside a perfusion bioreactor to create biocompatible biological rat scaffolds that mimic native architecture, resist mechanical stress and induce angiogenesis. Seeded allogeneic mesenchymal stromal cells spontaneously differentiate (proven by gene-, protein and functional evaluations) into epithelial- and muscle-like cells. The reseeded scaffolds are used to orthotopically replace the entire cervical oesophagus in immunocompetent rats. All animals survive the 14-day study period, with patent and functional grafts, and gain significantly more weight than sham-operated animals. Explanted grafts show regeneration of all the major cell and tissue components of the oesophagus including functional epithelium, muscle fibres, nerves and vasculature. We consider the presented tissue-engineered oesophageal scaffolds a significant step towards the clinical application of bioengineered oesophagi.
Collapse
Affiliation(s)
- Sebastian Sjöqvist
- 1] Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden [2] Division of Ear, Nose and Throat, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| | - Philipp Jungebluth
- 1] Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden [2] Division of Ear, Nose and Throat, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| | - Mei Ling Lim
- 1] Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden [2] Division of Ear, Nose and Throat, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| | - Johannes C Haag
- 1] Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden [2] Division of Ear, Nose and Throat, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| | - Ylva Gustafsson
- 1] Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden [2] Division of Ear, Nose and Throat, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| | - Greg Lemon
- Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden
| | - Silvia Baiguera
- Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden
| | | | - Costantino Del Gaudio
- Department of Industrial Engineering, Intrauniversitary Consortium for Material Science and Technology (INSTM) Research Unit Tor Vergata, University of Rome, Rome 00133, Italy
| | - Antonio Beltrán Rodríguez
- 1] Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden [2] Division of Ear, Nose and Throat, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| | - Alexander Sotnichenko
- International Scientific-Research Clinical and Educational Center of Regenerative Medicine, Kuban State Medical University, Krasnodar 350040, Russian Federation
| | - Karolina Kublickiene
- 1] Center for Gender Medicine, Karolinska Institutet, SE-141 86 Stockholm, Sweden [2] Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Henrik Ullman
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Heike Kielstein
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Peter Damberg
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention and Technology (CLINTEC) Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Alessandra Bianco
- Department of Industrial Engineering, Intrauniversitary Consortium for Material Science and Technology (INSTM) Research Unit Tor Vergata, University of Rome, Rome 00133, Italy
| | - Rainer Heuchel
- 1] Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86 Stockholm, Sweden [2] Center of Biosciences, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Ying Zhao
- 1] Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86 Stockholm, Sweden [2] Center of Biosciences, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, National Cancer Institute 'Giovanni Paolo II', Bari 70121, Italy
| | - Cristián Ibarra
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Bertrand Joseph
- Cancer Centrum Karolinska, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Doris A Taylor
- Texas Heart Institute, Center for Regenerative Medicine, Houston, Texas 770-30, USA
| | - Paolo Macchiarini
- 1] Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, SE-141 86, Stockholm, Sweden [2] Division of Ear, Nose and Throat, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| |
Collapse
|
10
|
Uygun I, Otcu S, Ozekinci S, Okur MH, Arslan MS, Aydogdu B. Dilated ureter for esophageal substitution: a preliminary experimental study in the rat. Clin Res Hepatol Gastroenterol 2014; 38:92-8. [PMID: 24011824 DOI: 10.1016/j.clinre.2013.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/21/2013] [Accepted: 07/29/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Esophageal replacement using digestive organs such as the colon, stomach, or jejunum has been used to treat long-gap esophageal atresia and caustic esophageal strictures. Nevertheless, it presents a major challenge. Here, we report a preliminary experimental study that examined the use of a free dilated ureter as an option for esophageal substitution in a transplantation rat model. METHODS Ten 28-week-old male donor rats underwent distal ureteral ligation for 4 weeks, and the total dilated ureters were recovered. In each of the ten recipient 20-week-old male rats, a ureter was transplanted through the mediastinum into the esophageal bed, without vascular anastomosis. All rats received cyclosporine and cotrimoxazole for 10 days. On postoperative day 10, the rats were sacrificed, and the transplanted ureters were evaluated macroscopically and histopathologically. RESULTS All procedures were achieved. In the early postoperative period, three transplanted rats died. Upon macroscopic evaluation, no evidence of complications was observed, and all transplanted ureters exhibited apparently good firm tissue. Histopathological examination showed a viable ureteral structure with good vascularity, low inflammation, and regenerated epithelium in all rats. CONCLUSION As an option for esophageal substitution, heterotopic ureteral transplantation can be performed directly into the mediastinal location of the esophagus, without vascular anastomosis in a rat model. In the future, free dilated ureters might be useful for esophageal grafting or patching in humans; however, this procedure must be validated in additional large animal models before being attempted in humans.
Collapse
Affiliation(s)
- Ibrahim Uygun
- Department of Pediatric Surgery, Medical Faculty of Dicle University, 21280 Diyarbakir, Turkey.
| | - Selcuk Otcu
- Department of Pediatric Surgery, Medical Faculty of Dicle University, 21280 Diyarbakir, Turkey
| | - Selver Ozekinci
- Department of Pathology, Medical Faculty of Dicle University, 21280 Diyarbakir, Turkey
| | - Mehmet Hanifi Okur
- Department of Pediatric Surgery, Medical Faculty of Dicle University, 21280 Diyarbakir, Turkey
| | - Mehmet Serif Arslan
- Department of Pediatric Surgery, Medical Faculty of Dicle University, 21280 Diyarbakir, Turkey
| | - Bahattin Aydogdu
- Department of Pediatric Surgery, Medical Faculty of Dicle University, 21280 Diyarbakir, Turkey
| |
Collapse
|
11
|
Steger V, Hampel M, Trick I, Müller M, Walles T. Clinical tracheal replacement: transplantation, bioprostheses and artificial grafts. Expert Rev Med Devices 2014; 5:605-12. [DOI: 10.1586/17434440.5.5.605] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Poghosyan T, Gaujoux S, Vanneaux V, Bruneval P, Domet T, Lecourt S, Jarraya M, Sfeir R, Larghero J, Cattan P. In vitro development and characterization of a tissue-engineered conduit resembling esophageal wall using human and pig skeletal myoblast, oral epithelial cells, and biologic scaffolds. Tissue Eng Part A 2013; 19:2242-52. [PMID: 23672649 DOI: 10.1089/ten.tea.2012.0565] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
INTRODUCTION Tissue engineering represents a promising approach for esophageal replacement, considering the complexity and drawbacks of conventional techniques. AIM To create the components necessary to reconstruct in vitro or in vivo an esophageal wall, we analyzed the feasibility and the optimal conditions of human and pig skeletal myoblast (HSM and PSM) and porcine oral epithelial cell (OEC) culture on biologic scaffolds. MATERIALS AND METHODS PSM and HSM were isolated from striated muscle and porcine OECs were extracted from oral mucosa biopsies. Myoblasts were seeded on an acellular scaffold issue from porcine small intestinal submucosa (SIS) and OEC on decellularized human amniotic membrane (HAM). Seeding conditions (cell concentrations [0.5×10(6) versus 10(6) cells/cm(2)] and culture periods [7, 14 and 21 days]), were analyzed using the methyl thiazoltetrazolium assay, quantitative PCR, flow cytometry, and immunohistochemistry. RESULTS Phenotypic stability was observed after cellular expansion for PSM and HSM (85% and 97% CD56-positive cells, respectively), and OECs (90% AE1/AE3- positive cells). After PSM and HSM seeding, quantities of viable cells were similar whatever the initial cell concentration used and remained stable at all time points. During cell culture on SIS, a decrease of CD56-positive cells was observed (76% and 76% by D7, 56% and 70% by D14, 28% and 60% by D21, for PSM and HSM, respectively). Multilayered surface of α-actin smooth muscle and Desmine-positive cells organized in bundles was seen as soon as D7, with no evidence of cell within the SIS. Myoblasts fusion was observed at D21. Pax3 and Pax7 expression was downregulated and MyoD expression upregulated, at D14.OEC proliferation was observed on HAM with both cell concentrations from D7 to D21. The cell metabolism activity was more important on matrix seeded by 10(6) cells/cm(2). With 0.5×10(6) OEC/cm(2), a single layer of pancytokeratin-positive cells was seen at D7, which became pluristratified by D14, while when 10(6) OEC/cm(2) were used, a pluristratified epithelial structure was seen as soon as D7. Proliferative cells (Proliferating Cell Nuclear Antigen staining) were mainly located at the basal layer. CONCLUSION In this model, the optimal conditions of cell seeding in terms of cell concentration and culture duration were 0.5×10(6) myoblasts/cm(2) and 10(6) OEC/cm(2), and 7 days.
Collapse
Affiliation(s)
- Tigran Poghosyan
- Cell Therapy Unit and Clinical Investigation Center in Biotherapies (CIC-BT501), Saint-Louis Hospital, AP-HP Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Azevedo-Pereira AED, Saka JA, Oliveira-Braga KAD, Pazetti R, Canzian M, Pêgo-Fernandes PM, Jatene FB. The impact of topically applied preservation solutions on the respiratory epithelium of tracheal grafts submitted to cold ischemia: functional and morphological analysis. Clinics (Sao Paulo) 2013; 68:702-9. [PMID: 23778415 PMCID: PMC3654295 DOI: 10.6061/clinics/2013(05)19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/14/2013] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Advances in graft reepithelialization and revascularization have renewed interest in airway transplantation. This study aims to determine whether topically applied preservation solutions can ameliorate ischemic injury to tracheal grafts. We analyzed 1) the effects of cold ischemia on the mucociliary clearance of tracheal grafts and 2) the impact of topically applied preservation solutions on the effects of cold ischemia on mucociliary clearance. METHOD Tracheal segments (n=217) from 109 male Wistar rats were harvested, submerged in low-potassium-dextran-glucose, histidine-tryptophan-ketoglutarate, or saline solution (saline group), and stored at 4°C for 6, 10, 16, or 24 hours. A control group (not submerged) was analyzed immediately after harvesting. In situ mucociliary transport and ciliary beating frequency were measured using a stroboscope. Epithelial integrity, cellular infiltration, and mucus storage were quantified by light microscopy and image analysis software, along with transmission electron microscopy. RESULTS 1) The effects of cold ischemia: in situ mucociliary transport and ciliary beating frequency were greater in the control group than after cold ischemia. Microscopic analysis results were similar between groups. 2) The effects of preservation solutions: there was no difference between the low-potassium-dextran-glucose, histidine-tryptophan-ketoglutarate, and saline groups in functional or light microscopy analysis. The saline group presented stronger signs of ischemic injury with transmission electron microscopy. CONCLUSIONS Cold ischemia diminished the mucociliary clearance of the tracheal respiratory epithelium. Topically applied preservation solutions did not ameliorate the injury caused by cold ischemia to the tracheal respiratory epithelium.
Collapse
Affiliation(s)
- Artur Eugênio de Azevedo-Pereira
- Instituto do Coração InCor, Faculdade de Medicina da Universidade de São Paulo, Laboratory of Research on Thoracic Surgery, LIM-61, São Paulo/SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
14
|
Farwell DG, Birchall MA, Macchiarini P, Luu QC, de Mattos AM, Gallay BJ, Perez RV, Grow MP, Ramsamooj R, Salgado MD, Brodie HA, Belafsky PC. Laryngotracheal transplantation: technical modifications and functional outcomes. Laryngoscope 2013; 123:2502-8. [PMID: 23483551 DOI: 10.1002/lary.24053] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 12/19/2012] [Accepted: 01/22/2013] [Indexed: 12/15/2022]
Abstract
OBJECTIVES/HYPOTHESIS Laryngeal transplantation offers the potential for patients without a larynx to recover their voice, which is critical in our communication age. We report clinical and functional outcomes from a laryngotracheal transplant. Widespread adoption of this technique has been slowed due to the ethical concerns of life-long immunosuppression after a nonvital organ transplant. Our patient was already on immunosuppressive medication from prior kidney-pancreas transplantation, and therefore was not exposed to added long-term risk. We describe the unique technical advances, clinical course, and rehabilitation of this patient and the implications for future laryngeal transplantation. STUDY DESIGN Case report. METHODS A laryngotracheal transplantation was performed in a 51-year-old prior kidney-pancreas transplant recipient presenting with complete laryngotracheal stenosis. Surgical modifications were made in the previously described technique related to retrieval, vascular supply, and reinnervation. This resulted in a robustly vascularized organ with well-perfused long-segment tracheal transplant and early return of motor reinnervation. RESULTS A multidisciplinary approach resulted in a successful transplant without evidence of rejection to date. Postoperatively, the patient continues to rely on a tracheotomy but has had the return of an oral and nasal airway, vocalization, smell, and taste, all experienced for the first time in 11 years. CONCLUSIONS We have demonstrated that our methods may result in a successful laryngotracheal transplant. We describe the preparation, surgical technique, rehabilitation, and interventions employed in achieving optimal outcomes. This report contributes valuable information on this rarely performed composite transplant.
Collapse
Affiliation(s)
- D Gregory Farwell
- Department of Otolaryngology-Head and Neck Surgery, University of California, Davis, Sacramento, California, U.S.A
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Luo X, Liu Y, Zhang Z, Tao R, Liu Y, He A, Yin Z, Li D, Zhang W, Liu W, Cao Y, Zhou G. Long-term functional reconstruction of segmental tracheal defect by pedicled tissue-engineered trachea in rabbits. Biomaterials 2013; 34:3336-44. [PMID: 23380355 DOI: 10.1016/j.biomaterials.2013.01.060] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 01/11/2013] [Indexed: 12/21/2022]
Abstract
Due to lack of satisfactory tracheal substitutes, reconstruction of long segmental tracheal defects (>6 cm) is always a major challenge in trachea surgery. Tissue-engineered trachea (TET) provides a promising approach to address this challenge, but no breakthrough has been achieved yet in repairing segmental tracheal defect. The longest survival time only reached 60 days. The leading reasons for the failure of segmental tracheal defect reconstruction were mainly related to airway stenosis (caused by the overgrowth of granulation tissue), airway collapse (caused by cartilage softening) and mucous impaction (mainly caused by lack of epithelium). To address these problems, the current study proposed an improved strategy, which involved in vitro pre-culture, in vivo maturation, and pre-vascularization of TET grafts as well as the use of silicone stent. The results demonstrated that the two-step strategy of in vitro pre-culture plus in vivo implantation could successfully regenerate tubular cartilage with a mechanical strength similar to native trachea in immunocompetent animals. The use of silicone stents effectively depressed granulation overgrowth, prevented airway stenosis, and thus dramatically enhanced the survival rate at the early stage post-operation. Most importantly, through intramuscular implantation and transplantation with pedicled muscular flap, the TET grafts established stable blood supply, which guaranteed maintenance of tubular cartilage structure and function, accelerated epithelialization of TET grafts, and thus realized long-term functional reconstruction of segmental tracheal defects. The integration of all these improved strategies finally realized long-term survival of animals: 60% of rabbits survived over 6 months. The current improved strategy provided a promising approach for long-term functional reconstruction of long segmental tracheal defect.
Collapse
Affiliation(s)
- Xusong Luo
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai Stem Cell Institute, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Her GJ, Wu HC, Chen MH, Chen MY, Chang SC, Wang TW. Control of three-dimensional substrate stiffness to manipulate mesenchymal stem cell fate toward neuronal or glial lineages. Acta Biomater 2013; 9:5170-80. [PMID: 23079022 DOI: 10.1016/j.actbio.2012.10.012] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 10/01/2012] [Accepted: 10/06/2012] [Indexed: 12/13/2022]
Abstract
The unlimited self-renewal and multipotency of stem cells provide great potential for applications in tissue engineering and regenerative medicine. The differentiation of stem cells can be induced by multiple factors including physical, chemical and biological cues. The fate of stem cells can be manipulated by deliberately controlling the interaction between stem cells and their microenvironment. The purpose of this study is to investigate the change in matrix stiffness under the influence of neurogenic differentiation of human mesenchymal stem cells (hMSCs). In this study, three-dimensional (3-D) porous scaffolds were synthesized by type I collagen (Col) and hyaluronic acid (HA). The elastic modulus of the 3-D substrates was modified by adjusting the concentration of 1-ethyl-3(3-dimethylaminopropyl) carbodiimide (EDC) as a crosslinking agent. The mechanical properties of Col-HA scaffolds were evaluated and the induction and characterization of hMSC differentiation toward neural lineages on substrates with different stiffnesses were studied. Using EDC of different concentrations for crosslinking, the stiffness of the matrices can be controlled in the range of 1-10 kPa for soft to stiff substrates, respectively. The results showed that MSCs were likely to differentiate into neuronal lineage in substrate at 1 kPa, while they transformed into glial cells in matrix at 10 kPa. The morphology and proliferation behavior of hMSCs responded to the different stiffnesses of substrates. Using this modifiable matrix, we can investigate the relationship between stem cell behavior and substrate mechanical properties in extracellular matrix-based biomimetic 3-D scaffolds. A substrate with controllable stiffness capable of inducing hMSCs specifically toward neuronal differentiation may be very useful as a tissue-engineered construct or substitute for delivering hMSCs into the brain and spinal cord.
Collapse
|
17
|
Partington L, Mordan NJ, Mason C, Knowles JC, Kim HW, Lowdell MW, Birchall MA, Wall IB. Biochemical changes caused by decellularization may compromise mechanical integrity of tracheal scaffolds. Acta Biomater 2013; 9:5251-61. [PMID: 23059415 DOI: 10.1016/j.actbio.2012.10.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 09/29/2012] [Accepted: 10/03/2012] [Indexed: 12/13/2022]
Abstract
Tissue-engineered airways have achieved clinical success, but concerns remain about short-term loss of biomechanical properties, necessitating a stent. This study investigated the effect of chemical-enzymatic decellularization on biochemical properties of trachea important for cell attachment and vascularization (fibronectin and laminin) and cartilage matrix homeostasis (type II collagen and glycosaminoglycans (GAG)), as well as biomechanical status. Native trachea was used as a control, and NDC trachea stored in phosphate buffered saline (PBS) in parallel to decellularization was used as a time-matched control. Decellularization removed most cells, but chondrocytes and DNA remained after 25 cycles. Fibronectin was retained throughout the lamina propria and laminin at basement membranes. DNA accumulation along ECM fibres was seen. A decline in soluble collagen was observed in decellularized tissue. GAG content of cartilage rings was reduced, even in PBS control tissue from 20 cycles onwards (p<0.05), but decellularization caused the greatest loss (p<0.01). Tensile strength declined throughout the process, but was significant only at later time points. The data demonstrate that the substantial reduction in GAG might contribute to loss of mechanical integrity of biotracheas. Overcoming structural changes that cause an imbalance in cartilage matrix equilibrium will be necessary to optimize clinical benefit, enabling widespread use of biotracheas.
Collapse
Affiliation(s)
- L Partington
- Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kuppan P, Sethuraman S, Krishnan UM. Tissue engineering interventions for esophageal disorders--promises and challenges. Biotechnol Adv 2012; 30:1481-92. [PMID: 22484299 DOI: 10.1016/j.biotechadv.2012.03.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/12/2012] [Accepted: 03/15/2012] [Indexed: 01/11/2023]
Abstract
The diseases of the esophagus include congenital defects like atresia, tracheoesophageal fistula as well as others such as gastro-esophageal reflux disease (GERD), Barrett's esophagus, carcinoma and strictures. All esophageal disorders require surgical intervention and reconstruction with appropriate substitutes. Primary anastomosis is used to treat most cases but treatment of long gap atresia still remains a clinical challenge. Autologous graft therapies using tissues from colon, and small and large intestine or gastric transplantations have been attempted but have constraints like leakage, infection and stenosis at the implanted site, which leads to severe morbidity and mortality. An alternative for autologous grafts are allogenic and xenogenic grafts, which have better availability but disease transmission and immunogenicity limit their applications. Use of biodegradable and biocompatible scaffolds to engineer the esophagus promises to be an effective regenerative strategy for treatment of esophageal disorders. Nanotopography of the fibrous scaffolds mimics the natural extracellular matrix (ECM) of the tissue and incorporation of chemical cues and tailoring mechanical properties provide the right microenvironment for co-culture of different cell types. Scaffolds cultured with esophageal cells (epithelial cells, fibroblast and smooth muscle cells) might show enhancement of the biofunctionality in vivo. This review attempts to address the various strategies and challenges involved in successful tissue engineering of the esophagus.
Collapse
Affiliation(s)
- Purushothaman Kuppan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | | |
Collapse
|
19
|
|
20
|
|
21
|
Gaujoux S, Le Balleur Y, Bruneval P, Larghero J, Lecourt S, Domet T, Lambert B, Zohar S, Prat F, Cattan P. Esophageal replacement by allogenic aorta in a porcine model. Surgery 2010; 148:39-47. [DOI: 10.1016/j.surg.2009.12.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Accepted: 12/04/2009] [Indexed: 12/13/2022]
|
22
|
Martinod E, Seguin A, Radu D, Marquette CH, Carpentier A. Avancées en chirurgie trachéale : a-t-on enfin trouvé le substitut idéal à la trachée ? Rev Mal Respir 2010; 27:554-64. [DOI: 10.1016/j.rmr.2010.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 12/21/2009] [Indexed: 12/16/2022]
|
23
|
Both epithelial cells and mesenchymal stem cell–derived chondrocytes contribute to the survival of tissue-engineered airway transplants in pigs. J Thorac Cardiovasc Surg 2010; 139:437-43. [DOI: 10.1016/j.jtcvs.2009.10.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2009] [Revised: 09/14/2009] [Accepted: 10/04/2009] [Indexed: 11/20/2022]
|
24
|
Jungebluth P, Go T, Asnaghi A, Bellini S, Martorell J, Calore C, Urbani L, Ostertag H, Mantero S, Conconi MT, Macchiarini P. Structural and morphologic evaluation of a novel detergent-enzymatic tissue-engineered tracheal tubular matrix. J Thorac Cardiovasc Surg 2009; 138:586-93; discussion 592-3. [PMID: 19698839 DOI: 10.1016/j.jtcvs.2008.09.085] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 09/02/2008] [Accepted: 09/12/2008] [Indexed: 10/20/2022]
Abstract
OBJECTIVE We sought to bioengineer a nonimmunogenic tracheal tubular matrix of 6 cm in length and test its structural, functional, and immunologic properties in vitro and in vivo. METHODS Twelve-centimeter tracheal segments were harvested from Yorkshire boars. Half of each segment was subjected to a detergent-enzymatic method (containing sodium deoxycholate/DNase lavations) of decellularization for as many cycles as needed, and the other half was stored in phosphate-buffered saline at 4 degrees C as a control. Bioengineered and control tracheas were then implanted in major histocompatibility complex-unmatched pigs (allograft) or mice (xenograft) heterotopically for 30 days. Structural and functional analysis and immunostaining were performed after each detergent-enzymatic method cycle and transplantation. RESULTS Compared with control tracheas, bioengineered matrices displayed no major histocompatibility complex class I and II antigens after 17 detergent-enzymatic method cycles, without significant (P > .05) differences in their strain ability (rupture force, 56.1 +/- 3.3 vs 55.5 +/- 2.4 N; tissue deformation at 203% +/- 13% vs 200% +/- 8% or 12.2 +/- 0.8 vs 12 +/- 0.5 cm; and applied maximum force, 173.4 +/- 3.2 vs 171.5 +/- 4.6 N). Thirty days after implantation, significantly (P < .01) smaller inflammatory reactions (392 vs 15 macrophages/mm(2) and 874 vs 167 T lymphocytes/mm(2)) and P-selectin expressions (1/6 vs 6/6) were observed in both the xenograft and allograft models with bioengineered matrices compared with those seen with control tracheas. There was no development of anti-pig leukocyte antigen antibodies or increase in both IgM and IgG content in mice implanted with bioengineered tracheas. CONCLUSIONS Bioengineered tracheal matrices displayed similar structural and mechanical characteristics to native tracheas and excite no immune response to 30 days when implanted as allografts or xenografts. This method holds great promise for the future of tissue-engineered airway replacement.
Collapse
Affiliation(s)
- Philipp Jungebluth
- Department of General Thoracic Surgery, Hospital Clinic, Fundació Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, CIBER Enfermedades Respiratorias, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Juhász A, Szilágyi A, Mikó I, Altorjay I, Kecskés G, Altorjay A. Esophageal replacement using cryopreserved tracheal graft. Dis Esophagus 2008; 21:468-72. [PMID: 19125802 DOI: 10.1111/j.1442-2050.2007.00780.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Esophageal replacement using artificial material is not a new concept. Prior experiences with prostheses, allografts and composite grafts have not proved particularly successful. The aim of this study is to investigate whether cryopreserved animal trachea is suitable for the replacement of the esophagus. In 12 beagle dogs a 6-cm-long segment of the cervical esophagus was removed, and was replaced with cryopreserved trachea, which had been stored for 21 days on -86 degrees C. The proximal and distal ends of the esophagus were joined together with the graft by simple continuous suture (Biosyn 3/0) and covered with the sternohyoid flap. Postoperatively 16 hematological parameters were measured. The dogs were planned to be euthanized at random on days 28, 42 and 56 after the operation. Tests for air leak were performed and the inner diameter of the graft was measured to detect shrinkage. The microscopic structure of the graft was analyzed using haematoxylin and eosin staining. There was no indication of insufficiency. Based on the air leak test the sutures withheld properly. The inner diameter of the graft narrowed from an average 19 mm (+/- 1 mm) to 15.8 mm (+/- 0.6 mm). In length, the graft shortened from an average 60 mm to 47 mm (+/- 3 mm). No feeding difficulty was observed. In two cases wound suppuration was found involving only the cutaneous and subcutaneous layers. Concerning the laboratory parameters, only the fibrinogen level and white blood cell count showed temporary although significant changes. Histology findings on the 56th day showed absolute integration of the trachea with the esophagus, with disintegration of the tracheal cartilages. Cryopreserved trachea seems to be suitable for the replacement of a 5-6-cm-long esophageal segment.
Collapse
Affiliation(s)
- A Juhász
- Department of Surgery, St. George University Teaching Hospital, Székesfehérvár, Hungary
| | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Iglesias M, Jungebluth P, Sibila O, Aldabo I, Matute MP, Petit C, Torres A, Macchiarini P. Experimental safety and efficacy evaluation of an extracorporeal pumpless artificial lung in providing respiratory support through the axillary vessels. J Thorac Cardiovasc Surg 2007; 133:339-45. [PMID: 17258560 DOI: 10.1016/j.jtcvs.2006.09.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Revised: 08/20/2006] [Accepted: 09/05/2006] [Indexed: 12/23/2022]
Abstract
OBJECTIVE We sought to investigate the safety and feasibility of implanting the pumpless interventional lung assist device (Novalung; Novalung GmbH, Hechingen, Germany) to the axillary vessels either by means of direct cannulation or end-to-side graft interposition and the capability of either type of vascular access to provide respiratory support during apneic ventilation in adult pigs. METHODS Ten pigs were ventilated for 4 hours (respiratory rate, 20-25 breaths/min; tidal volume, 10-12 mL/kg; fraction of inspired oxygen, 1.0; positive end-expiratory pressure, 5 cm H2O). Thereafter, the interventional lung assist device was surgically connected to the right axillary artery and vein by using direct cannulation (n = 5) or end-to-side ringed polytetrafluoroethylene graft interposition (n = 5), and ventilatory settings were reduced to achieve near apneic ventilation (respiratory rate, 4 breaths/min; tidal volume, 1-2 mL/kg; fraction of inspired oxygen, 1.0; positive end-expiratory pressure, 20 cm H2O). Hemodynamic and intrathoracic volumes and lung cytokine levels were measured. RESULTS Blood flow through the interventional lung assist device was 1.7 +/- 0.4 L/min or 30% +/- 14% of the cardiac output, and the mean pressure gradient across the interventional lung assist device was 10 +/- 2 mm Hg. The interventional lung assist device allowed an O2 transfer of 225.7 +/- 70 mL/min and a CO2 removal of 261.7 +/- 28.5 mL/min. Although the amount of blood flow perfusing the interventional lung assist device was significantly higher (P < .01) with direct cannulation (2.1 +/- 0.3 L/min) compared with that seen in graft interposition (1.3 +/- 0.3 L/min), the latter allowed similar respiratory support with reduced hemodynamic instability. CONCLUSIONS The axillary vessels are a safe and attractive cannulation site for pumpless partial respiratory support. Compared with direct cannulation, graft interposition was equally able to support the interventional lung assist device-driven gas exchange requirements during apneic ventilation with better hemodynamic stability.
Collapse
Affiliation(s)
- Manuela Iglesias
- General Thoracic Surgical Experimental Laboratory, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Chirurgie de la trachée : vieux problèmes, nouvelles techniques. Rev Mal Respir 2006. [DOI: 10.1016/s0761-8425(06)71670-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
29
|
|
30
|
Walles T, Giere B, Hofmann M, Schanz J, Hofmann F, Mertsching H, Macchiarini P. Experimental generation of a tissue-engineered functional and vascularized trachea. J Thorac Cardiovasc Surg 2004; 128:900-6. [PMID: 15573075 DOI: 10.1016/j.jtcvs.2004.07.036] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We sought to grow in vitro functional smooth muscle cells, chondrocytes, and respiratory epithelium on a biologic, directly vascularized matrix as a scaffold for tracheal tissue engineering. METHODS Ten- to 15-cm-long free jejunal segments with their own vascular pedicle were harvested and acellularized from donor pigs (n = 10) and used as a vascular matrix. Autologous costal chondrocytes, smooth muscle cells, and respiratory epithelium and endothelial progenitor cells were first cultured in vitro and then disseminated on the previously acellularized vascular matrix. Histologic, immunohistologic, molecular imaging, and Western blotting studies were then performed to assess cell viability. RESULTS The endothelial progenitor cells re-endothelialized the matrix to such an extent that endothelial cell viability was uniformly documented through 2-(18F)-fluoro-2'-deoxyglucose positron emission tomography. This vascularized scaffold was seeded with functional (according to Western blot analysis) smooth muscle cells and successfully reseeded with viable ciliated respiratory epithelium. Chondrocyte growth and production of extracellular cartilaginous matrix was observed as soon as 2 weeks after their culture. CONCLUSIONS The fundamental elements for a bioartificial trachea were successfully engineered in vitro in a direct vascularized 10- to 15-cm-long bioartificial matrix. Future experimental work will be directed to give them a 3-dimensional aspect and a biomechanical profile of a functioning trachea.
Collapse
Affiliation(s)
- Thorsten Walles
- General Thoracic Surgery Biological Laboratory, Heidehaus Hospital, Hannover Medical School, Am Leineufer 70, D-30419 Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Martinod E, Seguin A, Pfeuty K, Fornes P, Kambouchner M, Azorin JF, Carpentier AF. Long-term evaluation of the replacement of the trachea with an autologous aortic graft. Ann Thorac Surg 2003; 75:1572-8; discussion 1578. [PMID: 12735581 DOI: 10.1016/s0003-4975(03)00120-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Tracheal reconstruction after extensive resection remains a challenge in thoracic surgery. The goal of this experimental study was to analyze the long-term evolution of tracheal replacement using an autologous aortic graft. METHODS In 21 sheep, a 5-cm segment of the cervical trachea was replaced by a segment of the descending thoracic aorta that was reconstructed to a prosthetic graft. Because of the airway collapse reported in a previous series, a permanent (n = 13) or temporary (n = 8) stent was systematically placed in the lumen of the graft. Clinical, bronchoscopic, and histologic examinations were performed up to 3 years after implantation. RESULTS All animals survived the operation with no paraplegia. In the group with a permanent stent, three complications occurred: one stent displacement, one laryngeal edema, and one infection. Stent removal was tolerated after 6 months in the group with a temporary stent. Histologic examination showed a progressive transformation of the arterial segment into first extensive inflammatory tissue with a squamous epithelium, and after 6 to 36 months well-differentiated tracheal tissue including a continuous mucociliary epithelium and regular rings of newly formed cartilage. CONCLUSIONS An autologous aortic graft used as a substitute for extensive tracheal replacement in sheep remained functional for periods up to 3 years. The progressive transformation of the graft into a structure resembling tracheal tissue seems to be a key factor in long-term patency. The mechanism of this regenerative process and the possibility of using arterial homografts, which would make clinical application easier, remain to be evaluated.
Collapse
Affiliation(s)
- Emmanuel Martinod
- Laboratoire d'Etude des Greffes et Prothèses Cardiaques, Hôpital Broussais, Upres 264, Université Paris 6, France.
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
This review discusses the need for tracheal replacement, distinct from resection with primary anastomosis, the requirements for replacement, and the many efforts over the past century to accomplish this goal experimentally and clinically. Approaches have included use of foreign materials, nonviable tissue, autogenous tissue, tissue engineering, and transplantation. Biological problems in each category are noted.
Collapse
Affiliation(s)
- Hermes C Grillo
- Division of General Thoracic Surgery, Massachusetts General Hospital, Boston 02114, USA.
| |
Collapse
|
34
|
Murakawa T, Nakajima J, Motomura N, Murakami A, Takamoto S. Successful allotransplantation of cryopreserved tracheal grafts with preservation of the pars membranacea in nonhuman primates. J Thorac Cardiovasc Surg 2002; 123:153-60. [PMID: 11782769 DOI: 10.1067/mtc.2002.119056] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study was performed to confirm the feasibility of cryopreserved tracheal allotransplantation in primates, the anatomy and immunology of which are considered to be more closely related to those of humans than those of other animals. METHODS Cryopreserved tracheal allotransplantations were performed in 3 recipient primates. In the control group fresh tracheal allotransplantations were performed in 2 primates (control A), and a tracheal allotransplantation with a simply frozen tracheal graft was performed in 1 primate (control B). Monthly bronchoscopic examinations, histologic examinations, electron microscopic examinations, and immunohistochemical investigations were performed in each of the primates. RESULTS In the cryopreserved tracheal allotransplantation group, 3 recipient monkeys were killed on the 35th, 144th, and 387th postoperative days, respectively. All grafts were incorporated by the recipient trachea without stenosis in the cryopreserved group. In the control group 2 recipient monkeys were killed on the 93rd postoperative day (control A), and one was killed on the 84th postoperative day (control B). Severe stenosis was observed after the transplantation in all of the control monkeys. Immunologic reactions appeared to be attenuated by the cryopreservation, whereas T cell-mediated immunologic rejection (control A) and loss of cartilage viability (control B) were considered to be the causes of graft failure in the control group. CONCLUSION The immunogenicity of the tracheal allografts was reduced by cryopreservation, and cryopreserved tracheal allotransplantation was successful in our primate model. Further investigation of cryopreserved tracheal allotransplantation with regard to proper clinical applications and the limitations of the procedure should be performed.
Collapse
Affiliation(s)
- Tomohiro Murakawa
- Department of Cardiothoracic Surgery, Faculty of Medicine, University of Tokyo, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
35
|
Martinod E, Zegdi R, Zakine G, Aupecle B, Fornes P, D'audiffret A, Chachques JC, Azorin J, Carpentier A. A novel approach to tracheal replacement: the use of an aortic graft. J Thorac Cardiovasc Surg 2001; 122:197-8. [PMID: 11436064 DOI: 10.1067/mtc.2001.114346] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- E Martinod
- Laboratory for the Study of Cardiac Grafts and Prostheses, Broussais Hospital, and the Department of Thoracic and Vascular Surgery, Avicenne Hospital, Bobigny, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Macchiarini P, Candelier JJ, Coullin P, Guerra N, de Montpreville V, Dartevelle P, Duprez-Angioi K, Oriol R. Use of embryonic human trachea grown in nude mice to patch-repair congenital tracheal stenosis. Transplantation 2000; 70:1555-9. [PMID: 11152215 DOI: 10.1097/00007890-200012150-00004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Long congenital tracheal stenosis is a life-threatening condition, and the available surgical treatments do not give satisfactory long-term results. METHODS Human embryonic tracheas were implanted in the abdominal cavities of nude mice until their differentiation was completed. These differentiated tracheas were used to patch-repair surgically induced tracheal stenosis in piglets. The human, mouse, or pig origin, of all the cells in the two successive xenotransplants in the nude mouse and the pig, was determined on tissue sections by in situ hybridization with species-specific DNA probes. RESULTS The transplanted pigs thrived and reached normal adulthood, irrespective of the administration of immunosuppressive treatment. The human tracheal tissue developed in nude mice conserved human structures, with the exception of feeding capillaries, which were of mouse origin. The tracheal patch in the adult healthy pigs comprised only pig cells organized into a fibrous scar, which was covered by normal pig epithelium. CONCLUSIONS Results suggest that human embryonic trachea grown in nude mice can be successfully used as patch tracheoplasty for long congenital tracheal stenosis without conventional immunosuppression.
Collapse
Affiliation(s)
- P Macchiarini
- Department of Thoracic and Vascular Surgery, Heidehaus Hospital Hannover Medical School, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Invited commentary. Ann Thorac Surg 2000. [DOI: 10.1016/s0003-4975(00)02061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Martinod E, Zakine G, Fornes P, Zegdi R, d'Audiffret A, Aupecle B, Goussef N, Azorin J, Chachques JC, Fabiani JN, Carpentier A. [Metaplasia of aortic tissue into tracheal tissue. Surgical perspectives]. COMPTES RENDUS DE L'ACADEMIE DES SCIENCES. SERIE III, SCIENCES DE LA VIE 2000; 323:455-60. [PMID: 10879293 DOI: 10.1016/s0764-4469(00)00150-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tracheal reconstruction after extensive resection remains an unsolved surgical problem. Numerous attempts have been made using tracheal grafts or prosthetic conduits with disappointing results. In this study, we propose a new alternative using an aortic autograft as tracheal substitute. In a first series of experiments, a half circumference of two rings was replaced with an autologous carotid artery patch. In a second series, a complete segment of trachea was replaced with an autologous aortic graft supported by an endoluminal tracheal stent. No dehiscence or stenosis was observed. Microscopic examinations at 3 and 6 months showed the replacement of the aortic tissue by tracheal tissue comprising neoformation of cartilage and mucociliary or non-keratinizing metaplastic polystratified squamous epithelium. Although these results need to be confirmed by a larger series of experiments, they showed that a vascular tissue placed in a different environment with a different function can be submitted to a metaplastic transformation which tends to restore a normal structure adapted to its new function. These remarkable findings offer new perspectives in tracheal reconstruction in human.
Collapse
Affiliation(s)
- E Martinod
- Laboratoire d'étude des greffes et prothèses cardiaques, Hôpital Broussais, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Macchiarini P, Dulmet E, de Montpreville V, Mazmanian GM, Chapelier A, Dartevelle P. Tracheal growth after slide tracheoplasty. J Thorac Cardiovasc Surg 1997; 113:558-66. [PMID: 9081103 DOI: 10.1016/s0022-5223(97)70371-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Our goal was to investigate the effects of slide tracheoplasty on tracheal growth in newborn piglets. METHODS Slide tracheoplasty was performed on normal trachea (n = 6) and a model of tracheal stenosis resembling that seen in infants (n = 6). After division of the trachea at its midportion between the second cartilaginous ring above and the right upper lobe takeoff below (around 23 rings), the proximal and distal segments were incised vertically on opposite anterior and posterior surfaces and reconstructed together. RESULTS The reconstructed tracheas lengthened and their cross-sectional areas enlarged linearly at a rate of 0.94 cm per month and 1.55 mm2/kg, respectively, as the piglets grew over a 6-month period from 4.7 +/- 0.6 to 64.4 +/- 5.7 kg (+/- standard deviation). Growth was not different between the two studied groups. There was no narrowing or late restenosis. The mean anastomotic cross-sectional area was overall 1.63 +/- 0.28 times larger (range 1.2 to 2.7) than the cross-sectional area of the unreconstructed trachea. When the animals were put to death, all tracheal lumina were completely lined with normal respiratory epithelium and all layers were histologically intact; anastomotic trachealis muscles contracted less (p < 0.001) but relaxed similarly to those muscles lining normal tracheas. Tracheal blood supply was macroscopically and microscopically normal in both groups; however, newborn piglets had an almost twofold increased number of intramural capillary vessels as opposed to adult pigs (p < 0.001). CONCLUSIONS Results suggest that slide tracheoplasty is not limited by the length of stenosis, provides a permanent enlargement of the cross-sectional airway diameter, does not compromise tracheal vascular supply, and does not impair tracheal growth as somatic growth continues.
Collapse
Affiliation(s)
- P Macchiarini
- Department of Thoracic and Vascular Surgery, Hôpital Marie-Lannelongue, (Paris-Sud University), France
| | | | | | | | | | | |
Collapse
|