Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
3D Print Addit Manuf ; 10(5): 1015-1035, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37886399

RESUMO

Wounds are skin tissue damage due to trauma. Many factors inhibit the wound healing phase (hemostasis, inflammation, proliferation, and alteration), such as oxygenation, contamination/infection, age, effects of injury, sex hormones, stress, diabetes, obesity, drugs, alcoholism, smoking, nutrition, hemostasis, debridement, and closing time. Cellulose is the most abundant biopolymer in nature which is promising as the main matrix of wound dressings because of its good structure and mechanical stability, moisturizes the area around the wound, absorbs excess exudate, can form elastic gels with the characteristics of bio-responsiveness, biocompatibility, low toxicity, biodegradability, and structural similarity with the extracellular matrix (ECM). The addition of active ingredients as a model drug helps accelerate wound healing through antimicrobial and antioxidant mechanisms. Three-dimensional (3D) bioprinting technology can print cellulose as a bioink to produce wound dressings with complex structures mimicking ECM. The 3D printed cellulose-based wound dressings are a promising application in modern wound care. This article reviews the use of 3D printed cellulose as an ideal wound dressing and their properties, including mechanical properties, permeability aspect, absorption ability, ability to retain and provide moisture, biodegradation, antimicrobial property, and biocompatibility. The applications of 3D printed cellulose in the management of chronic wounds, burns, and painful wounds are also discussed.

2.
Biofabrication ; 16(1)2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-37906964

RESUMO

While the field of tissue engineering has progressed rapidly with the advent of 3D bioprinting and human induced pluripotent stem cells (hiPSCs), impact is limited by a lack of functional, thick tissues. One way around this limitation is to 3D bioprint tissues laden with hiPSCs. In this way, the iPSCs can proliferate to populate the thick tissue mass prior to parenchymal cell specification. Here we design a perfusion bioreactor for an hiPSC-laden, 3D-bioprinted chamber with the goal of proliferating the hiPSCs throughout the structure prior to differentiation to generate a thick tissue model. The bioreactor, fabricated with digital light projection, was optimized to perfuse the interior of the hydrogel chamber without leaks and to provide fluid flow around the exterior as well, maximizing nutrient delivery throughout the chamber wall. After 7 days of culture, we found that intermittent perfusion (15 s every 15 min) at 3 ml min-1provides a 1.9-fold increase in the density of stem cell colonies in the engineered tissue relative to analogous chambers cultured under static conditions. We also observed a more uniform distribution of colonies within the tissue wall of perfused structures relative to static controls, reflecting a homogeneous distribution of nutrients from the culture media. hiPSCs remained pluripotent and proliferative with application of fluid flow, which generated wall shear stresses averaging ∼1.0 dyn cm-2. Overall, these promising outcomes following perfusion of a stem cell-laden hydrogel support the production of multiple tissue types with improved thickness, and therefore increased function and utility.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Humanos , Alicerces Teciduais/química , Engenharia Tecidual , Perfusão , Diferenciação Celular , Hidrogéis , Reatores Biológicos
3.
Adv Healthc Mater ; 12(20): e2300584, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36930747

RESUMO

Extracellular vesicles (EVs) are implicated as promising therapeutics and drug delivery vehicles in various diseases. However, successful clinical translation will depend on the development of scalable biomanufacturing approaches, especially due to the documented low levels of intrinsic EV-associated cargo that may necessitate repeated doses to achieve clinical benefit in certain applications. Thus, here the effects of a 3D-printed scaffold-perfusion bioreactor system are assessed on the production and bioactivity of EVs secreted from bone marrow-derived mesenchymal stem cells (MSCs), a cell type widely implicated in generating EVs with therapeutic potential. The results indicate that perfusion bioreactor culture induces an ≈40-80-fold increase (depending on measurement method) in MSC EV production compared to conventional cell culture. Additionally, MSC EVs generated using the perfusion bioreactor system significantly improve wound healing in a diabetic mouse model, with increased CD31+ staining in wound bed tissue compared to animals treated with flask cell culture-generated MSC EVs. Overall, this study establishes a promising solution to a major EV translational bottleneck, with the capacity for tunability for specific applications and general improvement alongside advancements in 3D-printing technologies.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Animais , Camundongos , Vesículas Extracelulares/metabolismo , Reatores Biológicos , Perfusão , Impressão Tridimensional
4.
Biofabrication ; 15(1)2022 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-36126638

RESUMO

3D printing has rapidly become a critical enabling technology in tissue engineering and regenerative medicine for the fabrication of complex engineered tissues. 3D bioprinting, in particular, has advanced greatly to facilitate the incorporation of a broad spectrum of biomaterials along with cells and biomolecules of interest forin vitrotissue generation. The increasing complexity of novel bioink formulations and application-dependent printing conditions poses a significant challenge for replicating or innovating new bioprinting strategies. As the field continues to grow, it is imperative to establish a cohesive, open-source database that enables users to search through existing 3D printing formulations rapidly and efficiently. Through the efforts of the NIH/NIBIB Center for Engineering Complex Tissues, we have developed, to our knowledge, the first bioink database for extrusion-based 3D printing. The database is publicly available and allows users to search through and easily access information on biomaterials and cells specifically used in 3D printing. In order to enable a community-driven database growth, we have established an open-source portal for researchers to enter their publication information for addition into the database. Although the database has a broad range of capabilities, we demonstrate its utility by performing a comprehensive analysis of the printability domains of two well-established biomaterials in the printing world, namely poly(ϵ-caprolactone) and gelatin methacrylate. The database allowed us to rapidly identify combinations of extrusion pressure, temperature, and speed that have been used to print these biomaterials and more importantly, identify domains within which printing was not possible. The data also enabled correlation analysis between all the printing parameters, including needle size and type, that exhibited compatibility for cell-based 3D printing. Overall, this database is an extremely useful tool for the 3D printing and bioprinting community to advance their research and is an important step towards standardization in the field.


Assuntos
Bioimpressão , Alicerces Teciduais , Impressão Tridimensional , Engenharia Tecidual , Materiais Biocompatíveis
5.
Adv Sci (Weinh) ; 9(21): e2105909, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35436042

RESUMO

Diseases of the knee joint such as osteoarthritis (OA) affect all joint elements. An in vitro human cell-derived microphysiological system capable of simulating intraarticular tissue crosstalk is desirable for studying etiologies/pathogenesis of joint diseases and testing potential therapeutics. Herein, a human mesenchymal stem cell-derived miniature joint system (miniJoint) is generated, in which engineered osteochondral complex, synovial-like fibrous tissue, and adipose tissue are integrated into a microfluidics-enabled bioreactor. This novel design facilitates different tissues communicating while still maintaining their respective phenotypes. The miniJoint exhibits physiologically relevant changes when exposed to interleukin-1ß mediated inflammation, which are similar to observations in joint diseases in humans. The potential of the miniJoint in predicting in vivo efficacy of drug treatment is confirmed by testing the "therapeutic effect" of the nonsteroidal anti-inflammatory drug, naproxen, as well as four other potential disease-modifying OA drugs. The data demonstrate that the miniJoint recapitulates complex tissue interactions, thus providing a robust organ chip model for the study of joint pathology and the development of novel therapeutic interventions.


Assuntos
Células-Tronco Mesenquimais , Osteoartrite , Tecido Adiposo/patologia , Humanos , Articulação do Joelho/patologia , Osteoartrite/tratamento farmacológico
6.
J Biomed Mater Res A ; 110(6): 1190-1198, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35080115

RESUMO

Extracellular vesicles (EVs) represent an emerging class of therapeutics with significant potential and broad applicability. However, a general limitation is their rapid clearance after administration. Thus, methods to enable sustained EV release are of great potential value. Here, we demonstrate that EVs from mesenchymal stem/stromal cells (MSCs) can be incorporated into 3D-printed gelatin methacrylate (GelMA) hydrogel bioink, and that the initial burst release of EVs can be reduced by increasing the concentration of crosslinker during gelation. Further, the data show that MSC EV bioactivity in an endothelial gap closure assay is retained after the 3D printing and photocrosslinking processes. Our group previously showed that MSC EV bioactivity in this assay correlates with pro-angiogenic bioactivity in vivo, thus these results indicate the therapeutic potential of MSC EV-laden GelMA bioinks.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Gelatina , Hidrogéis , Metacrilatos , Impressão Tridimensional
7.
Adv Healthc Mater ; 10(23): e2101249, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34617414

RESUMO

Breast cancer and its most radical treatment, the mastectomy, significantly impose both physical transformations and emotional pain in thousands of women across the globe. Restoring the natural appearance of a nipple-areola complex directly on the reconstructed breast represents an important psychological healing experience for these women and remains an unresolved clinical challenge, as current restorative techniques render a flattened disfigured skin tab within a single year. To provide a long-term solution for nipple reconstruction, this work presents 3D printed hybrid scaffolds composed of complementary biodegradable gelatin methacrylate and synthetic non-degradable poly(ethylene) glycol hydrogels to foster the regeneration of a viable nipple-areola complex. In vitro results showcased the robust structural capacity and long-term shape retention of the nipple projection amidst internal fibroblastic contraction, while in vivo subcutaneous implantation of the 3D printed nipple-areola demonstrated minimal fibrotic encapsulation, neovascularization, and the formation of healthy granulation tissue. Envisioned as subdermal implants, these nipple-areola bioprinted regenerative grafts have the potential to transform the appearance of the newly reconstructed breast, reduce subsequent surgical intervention, and revolutionize breast reconstruction practices.


Assuntos
Neoplasias da Mama , Mamilos , Neoplasias da Mama/cirurgia , Estética , Feminino , Humanos , Mastectomia , Impressão Tridimensional
8.
ACS Appl Mater Interfaces ; 13(43): 50812-50822, 2021 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-34670077

RESUMO

Current approaches in small-diameter vascular grafts for coronary artery bypass surgeries fail to address physiological variations along the graft that contribute to thrombus formation and ultimately graft failure. We present an innovative interlayer drug delivery system that can be utilized for the sustained delivery of heparin through a graft with a high degree of temporal and spatial control. A heparin-loaded gelatin methacrylate (gelMA) interlayer sits within a biohybrid composed of decellularized bovine pericardium (dECM) and poly(propylene fumarate) (PPF), and its UV crosslinking is controlled via three-dimensional (3D) printed shadow masks. The masks can be readily designed to modulate the incident light intensity on the graft, enabling us to control the resultant gelMA crosslinking and properties. A high heparin loading efficiency was obtained in gelMA and was independent of crosslinking. We achieved sustained heparin release over the course of 2 weeks within the biohybrid material using the 3D printed mask patterns. High doses of heparin were observed to have detrimental effects on endothelial cell function. However, when exposed to heparin in a slower, more sustained manner consistent with the masks, endothelial cells behave similarly to untreated cells. Further, slower release profiles cause significantly more release of tissue factor pathway inhibitor, an anticoagulant, than a faster release profile. The heparin-loaded gelMA interlayer we have developed is a useful tool for the temporal and spatial control of heparin release that supports endothelial function and promotes an antithrombotic environment.


Assuntos
Sistemas de Liberação de Medicamentos , Heparina/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Impressão Tridimensional , Engenharia Tecidual , Liberação Controlada de Fármacos , Heparina/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos
9.
Essays Biochem ; 65(3): 519-531, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34342360

RESUMO

Recent advances in 3D printing (3DP) and tissue engineering approaches enable the potential application of these technologies to vaccine research. Reconstituting the native tissue or cellular microenvironment will be vital for successful evaluation of pathogenicity of viral infection and screening of potential vaccines. Therefore, establishing a reliable in vitro model to study the vaccine efficiency or delivery of viral disease is important. Here, this review summarizes two major ways that tissue engineering and 3DP strategies could contribute to vaccine research: (1) 3D human tissue models to study the response to virus can be served as a testbed for new potential therapeutics. Using 3D tissue platform attempts to explore alternative options to pre-clinical animal research for evaluating vaccine candidates. (2) 3DP technologies can be applied to improve the vaccination strategies which could replace existing vaccine delivery. Controlled antigen release using carriers that are generated with biodegradable biomaterials can further enhance the efficient development of immunity as well as combination of multiple-dose vaccines into a single injection. This mini review discusses the up-to-date report of current 3D tissue/organ models for potential vaccine potency and known bioengineered vaccine delivery systems.


Assuntos
Doenças Transmissíveis , Vacinas , Animais , Materiais Biocompatíveis , Impressão Tridimensional , Engenharia Tecidual
10.
Aging Cell ; 20(7): e13388, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34086398

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder with features of accelerated aging. The majority of HGPS cases are caused by a de novo point mutation in the LMNA gene (c.1824C>T; p.G608G) resulting in progerin, a toxic lamin A protein variant. Children with HGPS typically die from coronary artery diseases or strokes at an average age of 14.6 years. Endothelial dysfunction is a known driver of cardiovascular pathogenesis; however, it is currently unknown how progerin antagonizes normal angiogenic function in HGPS. Here, we use human iPSC-derived endothelial cell (iPSC-EC) models to study angiogenesis in HGPS. We cultured normal and HGPS iPSC-ECs under both static and fluidic culture conditions. HGPS iPSC-ECs show reduced endothelial nitric oxide synthase (eNOS) expression and activity compared with normal controls and concomitant decreases in intracellular nitric oxide (NO) level, which result in deficits in capillary-like microvascular network formation. Furthermore, the expression of matrix metalloproteinase 9 (MMP-9) was reduced in HGPS iPSC-ECs, while the expression of tissue inhibitor metalloproteinases 1 and 2 (TIMP1 and TIMP2) was upregulated relative to healthy controls. Finally, we used an adenine base editor (ABE7.10max-VRQR) to correct the pathogenic c.1824C>T allele in HGPS iPSC-ECs. Remarkably, ABE7.10max-VRQR correction of the HGPS mutation significantly reduced progerin expression to a basal level, rescued nuclear blebbing, increased intracellular NO level, normalized the misregulated TIMPs, and restored angiogenic competence in HGPS iPSC-ECs. Together, these results provide molecular insights of endothelial dysfunction in HGPS and suggest that ABE could be a promising therapeutic approach for correcting HGPS-related cardiovascular phenotypes.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Progéria/genética , Senescência Celular , Regulação para Baixo , Humanos , Progéria/patologia
11.
Adv Funct Mater ; 31(51)2021 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-35558090

RESUMO

Biomaterials that replicate patterns of microenvironmental signals from the stem cell niche offer the potential to refine platforms to regulate stem cell behavior. While significant emphasis has been placed on understanding the effects of biophysical and biochemical cues on stem cell fate, vascular-derived or angiocrine cues offer an important alternative signaling axis for biomaterial-based stem cell platforms. Elucidating dose-dependent relationships between angiocrine cues and stem cell fate are largely intractable in animal models and 2D cell cultures. In this study, microfluidic mixing devices are leveraged to generate 3D hydrogels containing lateral gradients in vascular density alongside murine hematopoietic stem cells (HSCs). Regional differences in vascular density can be generated via embossed gradients in cell, matrix, or growth factor density. HSCs co-cultured alongside vascular gradients reveal spatial patterns of HSC phenotype in response to angiocrine signals. Notably, decreased Akt signaling in high vessel density regions led to increased expansion of lineage-positive hematopoietic cells. This approach offers a combinatorial tool to rapidly screen a continuum of microenvironments with varying vascular, biophysical, and biochemical cues to reveal the influence of local angiocrine signals on HSC fate.

12.
Acta Biomater ; 121: 204-213, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33271356

RESUMO

Modular tissue engineering is a promising biofabrication strategy to create engineered bone grafts in a bottom-up manner, in which cell-laden micro-modules are prepared as basic building blocks to assemble macroscopic tissues via different integrating mechanisms. In this study, we prepared collagen microbeads loaded with human bone marrow derived mesenchymal stem cells (BMSCs) using a microfluidic approach. The cell-laden microbeads were characterized for size change, cell activity, osteogenesis, as well as their self-assembly properties to generate centimeter-sized constructs. Moreover, using the cell-laden beads as a supporting medium, induced pluripotent stem cell-derived endothelial cells (iPSC-EC) were patterned inside bead aggregates through extrusion-based 3D printing. This fabrication approach that combines modular tissue engineering and supports 3D printing has the potential to create 3D engineered bone grafts with a pre-existing, customized vasculature.


Assuntos
Células Endoteliais , Células-Tronco Mesenquimais , Humanos , Microesferas , Osteogênese , Engenharia Tecidual , Alicerces Teciduais
13.
Adv Funct Mater ; 30(3)2020 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-33041744

RESUMO

Despite recent advances in clinical procedures, the repair of soft tissue remains a reconstructive challenge. Current technologies such as synthetic implants and dermal flap autografting result in inefficient shape retention and unpredictable aesthetic outcomes. 3D printing, however, can be leveraged to produce superior soft tissue grafts that allow enhanced host integration and volume retention. Here, a novel dual bioink 3D printing strategy is presented that utilizes synthetic and natural materials to create stable, biomimetic soft tissue constructs. A double network ink composed of covalently crosslinked poly(ethylene) glycol and ionically crosslinked alginate acts as a physical support network that promotes cell growth and enables long-tersm graft shape retention. This is coupled with a cell-laden, biodegradable gelatin methacrylate bioink in a hybrid printing technique, and the composite scaffolds are evaluated in their mechanical properties, shape retention, and cytotoxicity. Additionally, a new shape analysis technique utilizing CloudCompare software is developed that expands the available toolbox for assessing scaffold aesthetic properties. With this dynamic 3D bioprinting strategy, complex geometries with robust internal structures can be easily modulated by varying the print ratio of non-degradable to sacrificial strands. The versatility of this hybrid printing fabrication platform can inspire the design of future multi-material regenerative implants.

14.
Tissue Eng Part A ; 26(23-24): 1369-1377, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33054685

RESUMO

Liver disease and the subsequent loss of liver function is an enormous clinical challenge. A severe shortage of donor liver tissue greatly limits patients' options for a timely transplantation. Tissue engineering approaches offer a promising alternative to organ transplantation by engineering artificial implantable tissues. We have established a platform of cell-laden microbeads as basic building blocks to assemble macroscopic tissues via different mechanisms. This modular fabrication strategy possesses great potential for liver tissue engineering in a bottom-up manner. In this study, we encapsulated human hepatocytes into microbeads presenting a favorable microenvironment consisting of collagen and mesenchymal stem cells, and then we perfused the beads in a three-dimensional printed tubular perfusion bioreactor that promoted oxygen and medium diffusion to the impregnated cells. We noted high cell vitality and retention of parenchymal cell functionality for up to 30 days in this culture system. Our engineering-based approach led to the advancement in tissue size and long-term functionality of an artificial liver tissue in vitro.


Assuntos
Hepatócitos/citologia , Transplante de Fígado , Engenharia Tecidual , Reatores Biológicos , Células Cultivadas , Colágeno , Humanos , Fígado , Fígado Artificial , Células-Tronco Mesenquimais , Perfusão , Alicerces Teciduais
15.
Adv Sci (Weinh) ; 7(6): 1902403, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32195081

RESUMO

As the most versatile and promising cell source, stem cells have been studied in regenerative medicine for two decades. Currently available culturing techniques utilize a 2D or 3D microenvironment for supporting the growth and proliferation of stem cells. However, these culture systems fail to fully reflect the supportive biological environment in which stem cells reside in vivo, which contain dynamic biophysical growth cues. Herein, a 4D programmable culture substrate with a self-morphing capability is presented as a means to enhance dynamic cell growth and induce differentiation of stem cells. To function as a model system, a 4D neural culture substrate is fabricated using a combination of printing and imprinting techniques keyed to the different biological features of neural stem cells (NSCs) at different differentiation stages. Results show the 4D culture substrate demonstrates a time-dependent self-morphing process that plays an essential role in regulating NSC behaviors in a spatiotemporal manner and enhances neural differentiation of NSCs along with significant axonal alignment. This study of a customized, dynamic substrate revolutionizes current stem cell therapies, and can further have a far-reaching impact on improving tissue regeneration and mimicking specific disease progression, as well as other impacts on materials and life science research.

16.
APL Bioeng ; 4(1): 010901, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32072121

RESUMO

Additive manufacturing in tissue engineering has significantly advanced in acceptance and use to address complex problems. However, there are still limitations to the technologies used and potential challenges that need to be addressed by the community. In this manuscript, we describe how the field can be advanced not only through the development of new materials and techniques but also through the standardization of characterization, which in turn may impact the translation potential of the field as it matures. Furthermore, we discuss how education and outreach could be modified to ensure end-users have a better grasp on the benefits and limitations of 3D printing to aid in their career development.

17.
Artigo em Inglês | MEDLINE | ID: mdl-34308105

RESUMO

Vascularization is among the top challenges that impede the clinical application of engineered tissues. This challenge has spurred tremendous research endeavor, defined as vascular tissue engineering (VTE) in this article, to establish a pre-existing vascular network inside the tissue engineered graft prior to implantation. Ideally, the engineered vasculature can be integrated into the host vasculature via anastomosis to supply nutrient to all cells instantaneously after surgery. Moreover, sufficient vascularization is of great significance in regenerative medicine from many other perspectives. Due to the critical role of vascularization in successful tissue engineering, we aim to provide an up-to-date overview of the fundamentals and VTE strategies in this article, including angiogenic cells, biomaterial/bio-scaffold design and bio-fabrication approaches, along with the reported utility of vascularized tissue complex in regenerative medicine. We will also share our opinion on the future perspective of this field.

18.
PLoS One ; 14(3): e0212502, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30822334

RESUMO

Hematopoietic stem cells (HSCs) play an important physiological role as regulators of all blood and immune cell populations, and are of clinical importance for bone marrow transplants. Regulating HSC biology in vitro for clinical applications requires improved understanding of biological inducers of HSC lineage specification. A significant challenge for controlled HSC expansion and differentiation is the complex network of molecular crosstalk between multiple bone marrow niche components influencing HSC biology. We describe a biology-driven computational approach to model cell kinetics in vitro to gain new insight regarding culture conditions and intercellular signaling networks. We further investigate the balance between self-renewal and differentiation that drives early and late hematopoietic progenitor populations. We demonstrate that changing the feedback driven by cell-secreted biomolecules alters lineage specification in early progenitor populations. Using a first order deterministic model, we are able to predict the impact of media change frequency on cell kinetics, as well as distinctions between primitive long-term HSCs and differentiated myeloid progenitors. Integrating the computational model and sensitivity analyses we identify critical culture parameters for regulating HSC proliferation and myeloid lineage specification. Our analysis suggests that accurately modeling the kinetics of hematopoietic sub-populations in vitro requires direct contributions from early progenitor differentiation along with the more traditionally considered intermediary oligopotent progenitors. While consistent with recent in vivo results, this work suggests the need to revise our perspective on HSC lineage engineering in vitro for expansion of discrete hematopoietic populations.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/metabolismo , Modelos Biológicos , Animais , Células Cultivadas , Feminino , Células-Tronco Hematopoéticas/citologia , Camundongos
19.
Adv Healthc Mater ; 8(5): e1801471, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30707508

RESUMO

The skin is responsible for several important physiological functions and has enormous clinical significance in wound healing. Tissue engineered substitutes may be used in patients suffering from skin injuries to support regeneration of the epidermis, dermis, or both. Skin substitutes are also gaining traction in the cosmetics and pharmaceutical industries as alternatives to animal models for product testing. Recent biomedical advances, ranging from cellular-level therapies such as mesenchymal stem cell or growth factor delivery, to large-scale biofabrication techniques including 3D printing, have enabled the implementation of unique strategies and novel biomaterials to recapitulate the biological, architectural, and functional complexity of native skin. This progress report highlights some of the latest approaches to skin regeneration and biofabrication using tissue engineering techniques. Current challenges in fabricating multilayered skin are addressed, and perspectives on efforts and strategies to meet those limitations are provided. Commercially available skin substitute technologies are also examined, and strategies to recapitulate native physiology, the role of regulatory agencies in supporting translation, as well as current clinical needs, are reviewed. By considering each of these perspectives while moving from bench to bedside, tissue engineering may be leveraged to create improved skin substitutes for both in vitro testing and clinical applications.


Assuntos
Materiais Biocompatíveis/administração & dosagem , Pele/citologia , Engenharia Tecidual/métodos , Pesquisa Translacional Biomédica/métodos , Animais , Humanos , Impressão Tridimensional , Regeneração/fisiologia , Pele Artificial , Alicerces Teciduais/química , Cicatrização/fisiologia
20.
Biomaterials ; 125: 54-64, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28231508

RESUMO

Hematopoietic stem cells (HSC) reside in unique bone marrow niches and are influenced by signals from surrounding cells, the extracellular matrix (ECM), ECM-bound or diffusible biomolecules. Here we describe the use of a three-dimensional hydrogel to alter the balance of HSC-generated autocrine feedback and paracrine signals generated by co-cultured niche-associated cells. We report shifts in HSC proliferation rate and fate specification in the presence of lineage positive (Lin+) niche cells. Hydrogels promoting autocrine feedback enhanced expansion of early hematopoietic progenitors while paracrine signals from Lin+ cells increased myeloid differentiation. We report thresholds where autocrine vs. paracrine cues alter HSC fate transitions, and were able to selectively abrogate the effects of matrix diffusivity and niche cell co-culture via the use of inhibitory cocktails of autocrine or paracrine signals. Together, these results suggest diffusive biotransport in three-dimensional biomaterials are a critical design element for the development of a synthetic stem cell niche.


Assuntos
Comunicação Autócrina/fisiologia , Matriz Extracelular/química , Células-Tronco Hematopoéticas/fisiologia , Hidrogéis/química , Comunicação Parácrina/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Materiais Biomiméticos/química , Linhagem da Célula/fisiologia , Feminino , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA