Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
2.
Tissue Eng Part C Methods ; 29(1): 20-29, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36565022

RESUMO

This study presents a novel surgical model developed to provide hematological support for implanted cellularized devices augmenting or replacing liver tissue function. Advances in bioengineering provide tools and materials to create living tissue replacements designed to restore that lost to disease, trauma, or congenital deformity. Such substitutes are often assembled and matured in vitro and need an immediate blood supply upon implantation, necessitating the development of supporting protocols. Animal translational models are required for continued development of engineered structures before clinical implementation, with rodent models often playing an essential early role. Our long-term goal has been generation of living tissue to provide liver function, utilizing advances in additive manufacturing technology to create 3D structures with intrinsic micron to millimeter scale channels modeled on natural vasculature. The surgical protocol developed enables testing various design iterations in vivo by anastomosis to the host rat vasculature. Lobation of rodent liver facilitates partial hepatectomy and repurposing the remaining vasculature to support implanted engineered tissue. Removal of the left lateral lobe exposes the underlying hepatic vasculature and can create space for a device. A shunt is created from the left portal vein to the left hepatic vein by cannulating each with separate silicone tubing. The device is then integrated into the shunt by connecting its inflow and outflow ports to the tubing and reestablishing blood flow. Sustained anticoagulation is maintained with an implanted osmotic pump. In our studies, animals were freely mobile after implantation; devices remained patent while maintaining blood flow through their millifluidic channels. This vascular anastomosis model has been greatly refined during the process of performing over 200 implantation procedures. We anticipate that the model described herein will find utility in developing preclinical translational protocols for evaluation of engineered liver tissue. Impact statement Tissue and organ transplantation are often the best clinically effective treatments for a variety of human ailments. However, the availability of suitable donor organs remains a critical problem. Advances in biotechnology hold potential in alleviating shortages, yet further work is required to surgically integrate large engineered tissues to host vasculature. Improved animal models such as the one described are valuable tools to support continued development and evaluation of novel therapies.


Assuntos
Transplante de Fígado , Roedores , Humanos , Ratos , Animais , Transplante de Fígado/métodos , Fígado/irrigação sanguínea , Hepatectomia/métodos , Engenharia Tecidual
3.
J Pediatr Surg ; 57(1): 9-11, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34716014

RESUMO

This 2020 Presidential Address was given at the American Pediatric Surgical Association 2021 Virtual Annual Meeting, May 20-22, 2021.


Assuntos
Cirurgia Geral , Pediatria , Criança , Congressos como Assunto , Humanos , Sociedades Médicas , Estados Unidos
4.
Pediatr Surg Int ; 36(10): 1123-1133, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32734338

RESUMO

The field of Tissue Engineering and Regenerative Medicine has evolved rapidly over the past thirty years. This review will summarize its history, current status and direction through the lens of clinical need, its progress through science in the laboratory and application back into patients. We can take pride in the fact that much effort and progress began with the surgical problems of children and that many surgeons in the pediatric surgical specialties have become pioneers and investigators in this new field of science, engineering, and medicine. Although the field has yet to fulfill its great promise, there have been several examples where a therapy has progressed from the first idea to human application within a short span of time and, in many cases, it has been applied in the surgical care of children.


Assuntos
Sistemas Automatizados de Assistência Junto ao Leito/organização & administração , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos , Criança , Humanos
5.
J Biomater Appl ; 33(8): 1070-1079, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30651054

RESUMO

Extracellular matrix materials mechanically dissociated into submillimeter particles have a larger surface area than sheet materials and enhanced cellular attachment. Decellularized porcine mesothelial extracellular matrix microparticles were seeded with bone marrow-derived mesenchymal stromal cells and cultured in a rotating bioreactor. The mesenchymal stromal cells attached and grew to confluency on the microparticles. The cell-seeded microparticles were then encapsulated in varying concentrations of fibrin glue, and the cells migrated rapidly off the microparticles. The combination of microparticles and mesenchymal stromal cells was then applied to a splinted full-thickness cutaneous in vivo wound model. There was evidence of increased cell infiltration and collagen deposition in mesenchymal stromal cells-treated wounds. Cell-seeded microparticles have potential as a cell delivery and paracrine therapy in impaired healing environments.


Assuntos
Matriz Extracelular/química , Transplante de Células-Tronco Mesenquimais , Pele/lesões , Alicerces Teciduais/química , Cicatrização , Adulto , Animais , Adesão Celular , Movimento Celular , Células Cultivadas , Matriz Extracelular/ultraestrutura , Fibrina/química , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Pele/ultraestrutura
7.
Cell Prolif ; 51(1)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29110360

RESUMO

OBJECTIVES: Creation of functional, durable vasculature remains an important goal within the field of regenerative medicine. Engineered biological vasculature has the potential to restore or improve human tissue function. We hypothesized that the pleotropic effects of insulin-like growth factor 1 (IGF1) would enhance the engineering of capillary-like vasculature. MATERIALS AND METHODS: The impact of IGF1 upon vasculogenesis was examined in in vitro cultures for a period of up to 40 days and as subcutaneous implants within immunodeficient mice. Co-cultures of human umbilical vein endothelial cells and human bone marrow-derived mesenchymal stem cells in collagen-fibronectin hydrogels were supplemented with either recombinant IGF1 protein or genetically engineered cells to provide sustained IGF1. Morphometric analysis was performed on the vascular networks that formed in four concentrations of IGF1. RESULTS: IGF1 supplementation significantly enhanced de novo vasculogenesis both in vitro and in vivo. Effects were long-term as they lasted the duration of the study period, and included network density, vessel length, and diameter. Bifurcation density was not affected. However, the highest concentrations of IGF1 tested were either ineffective or even deleterious. Sustained IGF1 delivery was required in vivo as the inclusion of recombinant IGF1 protein had minimal impact. CONCLUSION: IGF1 supplementation can be used to produce neovasculature with significantly enhanced network density and durability. Its use is a promising methodology for engineering de novo vasculature to support regeneration of functional tissue.


Assuntos
Colágeno/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Células Cultivadas , Técnicas de Cocultura/métodos , Fibronectinas/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Modelos Animais , Neovascularização Fisiológica/fisiologia , Engenharia Tecidual/métodos
8.
J Tissue Eng Regen Med ; 11(10): 2763-2773, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-27256796

RESUMO

Bone marrow-derived mesenchymal stem cells (BMSCs) can be obtained by minimally invasive means and would be a favourable source for cell-based cartilage regeneration. However, controlling the differentiation of the BMSCs towards the desired chondrogenic pathway has been a challenge hampering their application. The major aim of the present study was to determine if conditioned medium collected from cultured auricular chondrocytes could promote chondrogenic differentiation of BMSCs. Auricular chondrocytes were isolated and grown in BMSC standard culture medium (SM) that was collected and used as chondrocyte-conditioned medium (CCM). The BMSCs were expanded in either CCM or SM for three passages. Cells were seeded onto fibrous collagen scaffolds and precultured for 2 weeks with or without transforming growth factor-beta 3 (TGF-ß3). After preculture, constructs were implanted subcutaneously in nude mice for 6 and 12 weeks and evaluated with real-time polymerase chain reaction, histology, immunohistochemistry and biochemistry. Real-time polymerase chain reaction results showed upregulation of COL2A1 in the constructs cultured in CCM compared with those in SM. After 12 weeks in vivo, abundant neocartilage formation was observed in the implants that had been cultured in CCM, with or without TGF-ß3. In contrast, very little cartilage matrix formation was observed within the SM groups, regardless of the presence of TGF-ß3. Osteogenesis was only observed in the SM group with TGF-ß3. In conclusion, CCM even had a stronger influence on chondrogenesis than the supplementation of the standard culture medium with TGF-ß3, without signs of endochondral ossification. Efficient chondrogenic differentiation of BMSCs could provide a promising alternative cell population for auricular regeneration. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Condrócitos/citologia , Condrogênese/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Pavilhão Auricular/fisiologia , Células-Tronco Mesenquimais/citologia , Medicina Regenerativa/métodos , Animais , Bovinos , Proliferação de Células/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Nus , Ovinos , Alicerces Teciduais/química
9.
J Biomed Mater Res B Appl Biomater ; 105(3): 585-593, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26663848

RESUMO

Decellularized extracellular matrix (ECM) biomaterials are increasingly used in regenerative medicine for abdominal tissue repair. Emerging ECM biomaterials with greater compliance target surgical procedures like breast and craniofacial reconstruction to enhance aesthetic outcome. Clinical studies report improved outcomes with newly designed ECM scaffolds, but their comparative biological characteristics have received less attention. In this study, we investigated scaffolds derived from dermis (AlloDerm Regenerative Tissue Matrix), small intestinal submucosa (Surgisis 4-layer Tissue Graft and OASIS Wound Matrix), and mesothelium (Meso BioMatrix Surgical Mesh and Veritas Collagen Matrix) and evaluated biological properties that modulate cellular responses and recruitment. An assay panel was utilized to assess the ECM scaffold effects upon cells. Results of the material-conditioned media study demonstrated Meso BioMatrix and OASIS best supported cell proliferation. Meso BioMatrix promoted the greatest migration and chemotaxis signaling, followed by Veritas and OASIS; OASIS had superior suppression of cell apoptosis. The direct adhesion assay indicated that AlloDerm, Meso BioMatrix, Surgisis, and Veritas had sidedness that affected cell-material interactions. In the chick chorioallantoic membrane assay, Meso BioMatrix and OASIS best supported cell infiltration. Among tested materials, Meso BioMatrix and OASIS demonstrated characteristics that facilitate scaffold incorporation, making them promising choices for many clinical applications. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 585-593, 2017.


Assuntos
Proliferação de Células , Quimiotaxia , Derme/química , Matriz Extracelular/química , Fibroblastos/metabolismo , Transdução de Sinais , Alicerces Teciduais/química , Animais , Apoptose , Bovinos , Humanos , Camundongos , Células NIH 3T3 , Suínos
10.
Biomicrofluidics ; 10(5): 054116, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27795748

RESUMO

In pre-clinical safety studies, drug-induced vascular injury (DIVI) is defined as an adverse response to a drug characterized by degenerative and hyperplastic changes of endothelial cells and vascular smooth muscle cells. Inflammation may also be seen, along with extravasation of red blood cells into the smooth muscle layer (i.e., hemorrhage). Drugs that cause DIVI are often discontinued from development after considerable cost has occurred. An in vitro vascular model has been developed using endothelial and smooth muscle cells in co-culture across a porous membrane mimicking the internal elastic lamina. Arterial flow rates of perfusion media within the endothelial chamber of the model induce physiologic endothelial cell alignment. Pilot testing with a drug known to cause DIVI induced extravasation of red blood cells into the smooth muscle layer in all devices with no extravasation seen in control devices. This engineered vascular model offers the potential to evaluate candidate drugs for DIVI early in the discovery process. The physiologic flow within the co-culture model also makes it candidate for a wide variety of vascular biology investigations.

11.
Cytotherapy ; 18(6): 729-39, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27173749

RESUMO

BACKGROUND AIMS: The use of bone marrow-derived mesenchymal stromal cells (MSCs) in cell-based therapies is currently being developed for a number of diseases. Thus far, the clinical results have been inconclusive and variable, in part because of the variety of cell isolation procedures and culture conditions used in each study. A new isolation technique that streamlines the method of concentration and demands less time and attention could provide clinical and economic advantages compared with current methodologies. In this study, we evaluated the concentrating capability of an integrated centrifuge-based technology compared with standard Ficoll isolation. METHODS: MSCs were concentrated from bone marrow aspirate using the new device and the Ficoll method. The isolation capabilities of the device and the growth characteristics, secretome production, and differentiation capacity of the derived cells were determined. RESULTS: The new MSC isolation device concentrated the bone marrow in 90 seconds and resulted in a mononuclear cell yield 10-fold higher and with a twofold increase in cell retention compared with Ficoll. The cells isolated using the device were shown to exhibit similar morphology and functional activity as assessed by growth curves and secretome production compared to the Ficoll-isolated cells. The surface marker and trilineage differentiation profile of the device-isolated cells was consistent with the known profile of MSCs. DISCUSSION: The faster time to isolation and greater cell yield of the integrated centrifuge-based technology may make this an improved approach for MSC isolation from bone marrow aspirates.


Assuntos
Células da Medula Óssea/citologia , Separação Celular/métodos , Centrifugação/métodos , Células-Tronco Mesenquimais/citologia , Medula Óssea , Diferenciação Celular/fisiologia , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ficoll , Humanos , Osteoblastos/citologia
12.
J Biomed Mater Res A ; 104(7): 1728-35, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26946064

RESUMO

Extracellular matrix (ECM) materials from animal and human sources have become important materials for soft tissue repair. Microparticles of ECM materials have increased surface area and exposed binding sites compared to sheet materials. Decellularized porcine peritoneum was mechanically dissociated into 200 µm microparticles, seeded with fibroblasts and cultured in a low gravity rotating bioreactor. The cells avidly attached and maintained excellent viability on the microparticles. When the seeded microparticles were placed in a collagen gel, the cells quickly migrated off the microparticles and through the gel. Cells from seeded microparticles migrated to and across an in vitro anastomosis model, increasing the tensile strength of the model. Cell seeded microparticles of ECM material have potential for paracrine and cellular delivery therapies when delivered in a gel carrier. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1728-1735, 2016.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Matriz Extracelular/metabolismo , Microesferas , Modelos Biológicos , Cicatrização , Anastomose Cirúrgica , Animais , Movimento Celular , Colágeno , Fibroblastos/citologia , Humanos , Sus scrofa , Resistência à Tração
13.
Plast Reconstr Surg ; 137(3): 854-863, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26910665

RESUMO

BACKGROUND: Adequate biomaterials for tissue engineering bone and replacement of bone in clinical settings are still being developed. Previously, the combination of mesenchymal stem cells in hydrogels and calcium-based biomaterials in both in vitro and in vivo experiments has shown promising results. However, results may be optimized by careful selection of the material combination. METHODS: ß-Tricalcium phosphate scaffolds were three-dimensionally printed with five different hydrogels: collagen I, gelatin, fibrin glue, alginate, and Pluronic F-127. The scaffolds had eight channels, running throughout the entire scaffold, and macropores. Mesenchymal stem cells (2 × 10) were mixed with each hydrogel, and cell/hydrogel mixes were dispersed onto the corresponding ß-tricalcium phosphate/hydrogel scaffold and cultured under dynamic-oscillating conditions for 6 weeks. Specimens were harvested at 1, 2, 4, and 6 weeks and evaluated histologically, radiologically, biomechanically and, at 6 weeks, for expression of bone-specific proteins by reverse-transcriptase polymerase chain reaction. Statistical correlation analysis was performed between radiologic densities in Hounsfield units and biomechanical stiffness. RESULTS: Collagen I samples had superior bone formation at 6 weeks as demonstrated by volume computed tomographic scanning, with densities of 300 HU, similar to native bone, and the highest compression values. Bone specificity of new tissue was confirmed histologically and by the expression of alkaline phosphatase, osteonectin, osteopontin, and osteocalcin. The bone density correlated closely with histologic and biomechanical testing results. CONCLUSION: Bone formation is supported best by ß-tricalcium phosphate/collagen I hydrogel and mesenchymal stem cells in collagen I hydrogel. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Assuntos
Materiais Biocompatíveis/química , Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Fenômenos Biomecânicos , Fosfatos de Cálcio/química , Força Compressiva , Meios de Cultivo Condicionados , Humanos , Hidrogéis/química , Técnicas In Vitro , Células-Tronco Mesenquimais/citologia , Sensibilidade e Especificidade , Tomografia Computadorizada por Raios X/métodos
14.
Tissue Eng Part A ; 22(3-4): 197-207, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26529401

RESUMO

Advancement of engineered ear in clinical practice is limited by several challenges. The complex, largely unsupported, three-dimensional auricular neocartilage structure is difficult to maintain. Neocartilage formation is challenging in an immunocompetent host due to active inflammatory and immunological responses. The large number of autologous chondrogenic cells required for engineering an adult human-sized ear presents an additional challenge because primary chondrocytes rapidly dedifferentiate during in vitro culture. The objective of this study was to engineer a stable, human ear-shaped cartilage in an immunocompetent animal model using expanded chondrocytes. The impact of basic fibroblast growth factor (bFGF) supplementation on achieving clinically relevant expansion of primary sheep chondrocytes by in vitro culture was determined. Chondrocytes expanded in standard medium were either combined with cryopreserved, primary passage 0 chondrocytes at the time of scaffold seeding or used alone as control. Disk and human ear-shaped scaffolds were made from porous collagen; ear scaffolds had an embedded, supporting titanium wire framework. Autologous chondrocyte-seeded scaffolds were implanted subcutaneously in sheep after 2 weeks of in vitro incubation. The quality of the resulting neocartilage and its stability and retention of the original ear size and shape were evaluated at 6, 12, and 20 weeks postimplantation. Neocartilage produced from chondrocytes that were expanded in the presence of bFGF was superior, and its quality improved with increased implantation time. In addition to characteristic morphological cartilage features, its glycosaminoglycan content was high and marked elastin fiber formation was present. The overall shape of engineered ears was preserved at 20 weeks postimplantation, and the dimensional changes did not exceed 10%. The wire frame within the engineered ear was able to withstand mechanical forces during wound healing and neocartilage maturation and prevented shrinkage and distortion. This is the first demonstration of a stable, ear-shaped elastic cartilage engineered from auricular chondrocytes that underwent clinical-scale expansion in an immunocompetent animal over an extended period of time.


Assuntos
Condrócitos , Cartilagem da Orelha , Orelha , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Humanos , Ovinos
15.
Tissue Eng Part A ; 21(15-16): 2147-55, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26020102

RESUMO

Chronic lower respiratory disease is highly prevalent in the United States, and there remains a need for alternatives to lung transplant for patients who progress to end-stage lung disease. Portable or implantable gas oxygenators based on microfluidic technologies can address this need, provided they operate both efficiently and biocompatibly. Incorporating biomimetic materials into such devices can help replicate native gas exchange function and additionally support cellular components. In this work, we have developed microfluidic devices that enable blood gas exchange across ultra-thin collagen membranes (as thin as 2 µm). Endothelial, stromal, and parenchymal cells readily adhere to these membranes, and long-term culture with cellular components results in remodeling, reflected by reduced membrane thickness. Functionally, acellular collagen-membrane lung devices can mediate effective gas exchange up to ∼288 mL/min/m(2) of oxygen and ∼685 mL/min/m(2) of carbon dioxide, approaching the gas exchange efficiency noted in the native lung. Testing several configurations of lung devices to explore various physical parameters of the device design, we concluded that thinner membranes and longer gas exchange distances result in improved hemoglobin saturation and increases in pO2. However, in the design space tested, these effects are relatively small compared to the improvement in overall oxygen and carbon dioxide transfer by increasing the blood flow rate. Finally, devices cultured with endothelial and parenchymal cells achieved similar gas exchange rates compared with acellular devices. Biomimetic blood oxygenator design opens the possibility of creating portable or implantable microfluidic devices that achieve efficient gas transfer while also maintaining physiologic conditions.


Assuntos
Colágeno/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Dispositivos Lab-On-A-Chip , Membranas Artificiais , Oxigênio/metabolismo , Adesão Celular , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos
16.
J Craniomaxillofac Surg ; 43(3): 382-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25600627

RESUMO

BACKGROUND: Carved autologous costal cartilage and porous polyethylene implants (Medpor) are the most common approaches for total ear reconstruction, but these approaches may have inconsistent cosmetic outcomes, a high risk of extrusion, or other surgical complications. Engineering ear cartilage to emulate native auricular tissue is an appealing approach, but often the cell-seeded scaffolds are susceptible to shrinkage and architectural changes when placed in vivo. The aim of this study was to assess the most favorable conditions for in vitro pre-culture of cell-seeded type I collagen scaffolds prior to in vivo implantation. METHODS: Sheep auricular chondrocytes were seeded into this type I collagen scaffold. The cell-seeded constructs were cultured in either static or dynamic conditions for two days or two weeks and then implanted into nude mice for another six weeks. The harvested constructs were evaluated histologically, immunohistochemically, and biochemically. RESULTS: Robust neo-cartilage formation was found in these collagen scaffolds seeded with auricular chondrocytes, which was comparable to native cartilage morphologically, histologically, and biochemically. Culture under dynamic conditions prior to implantation improved the neo-cartilage formation histologically and biochemically. CONCLUSION: Dynamic culture of this cell-seeded fibrous collagen material could permit predictable engineered auricular cartilage and a promising approach for external ear reconstruction.


Assuntos
Condrócitos/fisiologia , Colágeno Tipo I/química , Cartilagem da Orelha/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Técnicas de Cultura de Células , Separação Celular/métodos , Células Cultivadas , Condrogênese/fisiologia , DNA/análise , Cartilagem da Orelha/anatomia & histologia , Cartilagem da Orelha/química , Elastina/análise , Glicosaminoglicanos/análise , Hidroxiprolina/análise , Camundongos , Camundongos Nus , Ovinos , Tela Subcutânea/cirurgia , Propriedades de Superfície , Fatores de Tempo
17.
Ann Thorac Surg ; 98(5): 1721-9; discussion 1729, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25149047

RESUMO

BACKGROUND: Whole-lung scaffolds can be created by perfusion decellularization of cadaveric donor lungs. The resulting matrices can then be recellularized to regenerate functional organs. This study evaluated the capacity of acellular lung scaffolds to support recellularization with lung progenitors derived from human induced pluripotent stem cells (iPSCs). METHODS: Whole rat and human lungs were decellularized by constant-pressure perfusion with 0.1% sodium dodecyl sulfate solution. Resulting lung scaffolds were cryosectioned into slices or left intact. Human iPSCs were differentiated to definitive endoderm, anteriorized to a foregut fate, and then ventralized to a population expressing NK2 homeobox 1 (Nkx2.1). Cells were seeded onto slices and whole lungs, which were maintained under constant perfusion biomimetic culture. Lineage specification was assessed by quantitative polymerase chain reaction and immunofluorescent staining. Regenerated left lungs were transplanted in an orthotopic position. RESULTS: Activin-A treatment, followed by transforming growth factor-ß inhibition, induced differentiation of human iPSCs to anterior foregut endoderm as confirmed by forkhead box protein A2 (FOXA2), SRY (Sex Determining Region Y)-Box 17 (SOX17), and SOX2 expression. Cells cultured on decellularized lung slices demonstrated proliferation and lineage commitment after 5 days. Cells expressing Nkx2.1 were identified at 40% to 60% efficiency. Within whole-lung scaffolds and under perfusion culture, cells further upregulated Nkx2.1 expression. After orthotopic transplantation, grafts were perfused and ventilated by host vasculature and airways. CONCLUSIONS: Decellularized lung matrix supports the culture and lineage commitment of human iPSC-derived lung progenitor cells. Whole-organ scaffolds and biomimetic culture enable coseeding of iPSC-derived endothelial and epithelial progenitors and enhance early lung fate. Orthotopic transplantation may enable further in vivo graft maturation.


Assuntos
Órgãos Bioartificiais , Células Epiteliais/citologia , Matriz Extracelular/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Transplante de Pulmão/métodos , Pulmão/citologia , Alicerces Teciduais , Animais , Cadáver , Diferenciação Celular , Células Cultivadas , Sobrevivência de Enxerto , Humanos , Ratos , Ratos Sprague-Dawley
18.
Semin Pediatr Surg ; 23(3): 150-5, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24994529

RESUMO

Liver transplantation remains the only definitive treatment for liver failure and is available to only a tiny fraction of patients with end-stage liver diseases. Major limitations for the procedure include donor organ shortage, high cost, high level of required expertise, and long-term consequences of immune suppression. Alternative cell-based liver therapies could potentially greatly expand the number of patients provided with effective treatment. Investigative research into augmenting or replacing liver function extends into three general strategies. Bioartificial livers (BALs) are extracorporeal devices that utilize cartridges of primary hepatocytes or cell lines to process patient plasma. Injection of liver cell suspensions aims to foster organ regeneration or provide a missing metabolic function arising from a genetic defect. Tissue engineering recreates the organ in vitro for subsequent implantation to augment or replace patient liver function. Translational models and clinical trials have highlighted both the immense challenges involved and some striking examples of success.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Doença Hepática Terminal/cirurgia , Regeneração Tecidual Guiada/métodos , Transplante de Fígado , Fígado Artificial , Engenharia Tecidual/métodos , Células-Tronco Embrionárias/transplante , Hepatócitos/transplante , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Pluripotentes/transplante , Alicerces Teciduais
19.
Front Biosci (Landmark Ed) ; 19(8): 1227-39, 2014 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-24896347

RESUMO

Lung tissue engineering is an emerging field focused on the development of lung replacement devices and tissue to treat patients with end stage lung disease. Microfluidic based lung assist devices have been developed that have biomimetically designed vascular networks that achieve physiologic blood flow. Gas exchange in these devices occurs across a thin respiratory membrane. Designed for intrathoracic implantation as a bridge to transplant or destination therapy, these lung assist devices will allow ambulation and hospital discharge for patients with end stage lung disease. Decellularized lungs subsequently recellularized with epithelial and endothelial cells have been implanted in small animal models with demonstration of initial gas exchange. Further development of these tissues and scaling to large animal models will validate this approach and may be an organ source for lung transplantation. Initial clinical success has been achieved with decellularized tracheal implants using autologous stem cells. Development of microfluidic lung models using similar architecture to the lung assist device technology allows study of lung biology and diseases with manipulation of lung cells and respiratory membrane strain.


Assuntos
Órgãos Bioartificiais , Pulmão/citologia , Engenharia Tecidual/métodos , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/fisiologia , Técnicas Analíticas Microfluídicas/métodos , Modelos Biológicos , Troca Gasosa Pulmonar , Alicerces Teciduais
20.
Ann Otol Rhinol Laryngol ; 123(2): 135-40, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24574469

RESUMO

OBJECTIVES: We developed a large animal model for auricular reconstruction with engineered cartilage frameworks and evaluated the performance of porous polyethylene auricular implants in this model. METHODS: Eighteen high-density porous polyethylene auricular frameworks were implanted subcutaneously in the infra-auricular areas of 9 sheep. The implants were harvested 17 weeks later for gross and histologic examination. The perioperative and postoperative courses were carefully documented. RESULTS: Five implants became exposed, and 2 implants needed to be removed at 7 weeks. Additionally, 1 infected implant was removed at 2 weeks. Seromas developed in 2 implants because of drain failures and were drained successfully during the first postoperative week. There were no other surgical site complications. The remaining 10 implants had an acceptable cosmetic appearance at 17 weeks. CONCLUSIONS: The perioperative complication rate in the ovine porous polyethylene auricular implant model was higher than that reported for auricular reconstructions in humans. The implant exposures were likely caused by ischemia and excessive stress on the thin overlying skin, because vascularized flap coverage was not used. The histologic findings were comparable to the results reported for other animal models. This large animal model is appropriate for auricular reconstruction experiments, including engineered constructs.


Assuntos
Pavilhão Auricular/cirurgia , Cartilagem da Orelha/cirurgia , Modelos Animais , Polietileno , Engenharia Tecidual , Alicerces Teciduais , Animais , Feminino , Masculino , Porosidade , Procedimentos de Cirurgia Plástica , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA