Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 79(7): 349, 2022 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-35672585

RESUMO

All living beings continue their life by receiving energy and by excreting waste products. In animals, the arteries are the pathways of these transfers to the cells. Angiogenesis, the formation of the arteries by the development of pre-existed parental blood vessels, is a phenomenon that occurs naturally during puberty due to certain physiological processes such as menstruation, wound healing, or the adaptation of athletes' bodies during exercise. Nonetheless, the same life-giving process also occurs frequently in some patients and, conversely, occurs slowly in some physiological problems, such as cancer and diabetes, so inhibiting angiogenesis has been considered to be one of the important strategies to fight these diseases. Accordingly, in tissue engineering and regenerative medicine, the highly controlled process of angiogenesis is very important in tissue repairing. Excessive angiogenesis can promote tumor progression and lack of enough angiogensis can hinder tissue repair. Thereby, both excessive and deficient angiogenesis can be problematic, this review article introduces and describes the types of factors involved in controlling angiogenesis. Considering all of the existing strategies, we will try to lay out the latest knowledge that deals with stimulating/inhibiting the angiogenesis. At the end of the article, owing to the early-reviewed mechanical aspects that overshadow angiogenesis, the strategies of angiogenesis in tissue engineering will be discussed.


Assuntos
Neovascularização Fisiológica , Engenharia Tecidual , Animais , Humanos , Neovascularização Patológica , Medicina Regenerativa , Cicatrização
2.
Clin Oral Investig ; 27(3): 1215-1225, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36287273

RESUMO

OBJECTIVES: To develop a 3D-printed, microparticulate hydrogel supplemented with dentin matrix molecules (DMM) as a novel regenerative strategy for dental pulp capping. MATERIALS AND METHODS: Gelatin methacryloyl microgels (7% w/v) mixed with varying concentrations of DMM were printed using a digital light projection 3D printer and lyophilized for 2 days. The release profile of the DMM-loaded microgels was measured using a bicinchoninic acid assay. Next, dental pulp exposure defects were created in maxillary first molars of Wistar rats. The exposures were randomly capped with (1) inert material - negative control, (2) microgels, (3) microgels + DMM 500 µg/ml, (4) microgels + DMM 1000 µg/ml, (5) microgels + platelet-derived growth factor (PDGF 10 ng/ml), or (6) MTA (n = 15/group). After 4 weeks, animals were euthanized, and treated molars were harvested and then processed to evaluate hard tissue deposition, pulp tissue organization, and blood vessel density. RESULTS: All the specimens from groups treated with microgel + 500 µg/ml, microgel + 1000 µg/ml, microgel + PDGF, and MTA showed the formation of organized pulp tissue, tertiary dentin, newly formed tubular and atubular dentin, and new blood vessel formation. Dentin bridge formation was greater and pulp necrosis was less in the microgel + DMM groups compared to MTA. CONCLUSIONS: The 3D-printed photocurable microgels doped with DMM exhibited favorable cellular and inflammatory pulp responses, and significantly more tertiary dentin deposition. CLINICAL RELEVANCE: 3D-printed microgel with DMM is a promising biomaterial for dentin and dental pulp regeneration in pulp capping procedures.


Assuntos
Dentina Secundária , Microgéis , Agentes de Capeamento da Polpa Dentária e Pulpectomia , Ratos , Animais , Polpa Dentária , Compostos de Cálcio/uso terapêutico , Capeamento da Polpa Dentária/métodos , Materiais Biocompatíveis , Silicatos/uso terapêutico , Ratos Wistar , Regeneração , Impressão Tridimensional , Combinação de Medicamentos , Óxidos/uso terapêutico
3.
Connect Tissue Res ; 63(5): 514-529, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35132918

RESUMO

BACKGROUND: Oral mucosa equivalents (OMEs) have been used as in vitro models (eg, for studies of human oral mucosa biology and pathology, toxicological and pharmacological tests of oral care products), and clinically to treat oral defects. However, the human oral mucosa is a highly vascularized tissue and implantation of large OMEs can fail due to a lack of vascularization. To develop equivalents that better resemble the human oral mucosa and increase the success of implantation to repair large-sized defects, efforts have been made to prevascularize these constructs. PURPOSE: The aim of this narrative review is to provide an overview of the human oral mucosa structure, common approaches for its reconstruction, and the development of OMEs, their prevascularization, and in vitro and clinical potential applications. STUDY SELECTION: Articles on non-prevascularized and prevascularized OMEs were included, since the development and applications of non-prevascularized OMEs are a foundation for the design, fabrication, and optimization of prevascularized OMEs. CONCLUSIONS: Several studies have reported the development and in vitro and clinical applications of OMEs and only a few were found on prevascularized OMEs using different approaches of fabrication and incorporation of endothelial cells, indicating a lack of standardized protocols to obtain these equivalents. However, these studies have shown the feasibility of prevascularizing OMEs and their implantation in animal models resulted in enhanced integration and healing. Vascularization in tissue equivalents is still a challenge, and optimization of cell culture conditions, biomaterials, and fabrication techniques along with clinical studies is required.


Assuntos
Mucosa Bucal , Engenharia Tecidual , Animais , Materiais Biocompatíveis , Células Endoteliais , Humanos , Neovascularização Fisiológica , Engenharia Tecidual/métodos
4.
Nanomedicine ; 42: 102550, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35292368

RESUMO

mRNA therapeutics have increased in popularity, largely due to the transient and fast nature of protein expression and the low risk of off-target effects. This has increased drastically with the remarkable success of mRNA-based vaccines for COVID-19. Despite advances in lipid nanoparticle (LNP)-based delivery, the mechanisms that regulate efficient endocytic trafficking and translation of mRNA remain poorly understood. Although it is widely acknowledged that the extracellular matrix (ECM) regulates uptake and expression of exogenous nano-complexed genetic material, its specific effects on mRNA delivery and expression have not yet been examined. Here, we demonstrate a critical role for matrix stiffness in modulating both mRNA transfection and expression and uncover distinct mechano-regulatory mechanisms for endocytosis of mRNA through RhoA mediated mTOR signaling and cytoskeletal dynamics. Our findings have implications for effective delivery of therapeutic mRNA to targeted tissues that may be differentially affected by tissue and matrix stiffness.


Assuntos
COVID-19 , Nanopartículas , COVID-19/terapia , Vacinas contra COVID-19 , Humanos , Hidrogéis , Lipídeos/genética , Lipossomos , RNA Mensageiro/genética
5.
Adv Funct Mater ; 31(6)2021 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35422682

RESUMO

Bone is an active organ that continuously undergoes an orchestrated process of remodeling throughout life. Bone tissue is uniquely capable of adapting to loading, hormonal, and other changes happening in the body, as well as repairing bone that becomes damaged to maintain tissue integrity. On the other hand, diseases such as osteoporosis and metastatic cancers disrupt normal bone homeostasis leading to compromised function. Historically, our ability to investigate processes related to either physiologic or diseased bone tissue has been limited by traditional models that fail to emulate the complexity of native bone. Organ-on-a-chip models are based on technological advances in tissue engineering and microfluidics, enabling the reproduction of key features specific to tissue microenvironments within a microfabricated device. Compared to conventional in-vitro and in-vivo bone models, microfluidic models, and especially organs-on-a-chip platforms, provide more biomimetic tissue culture conditions, with increased predictive power for clinical assays. In this review, we will report microfluidic and organ-on-a-chip technologies designed for understanding the biology of bone as well as bone-related diseases and treatments. Finally, we discuss the limitations of the current models and point toward future directions for microfluidics and organ-on-a-chip technologies in bone research.

6.
J Struct Biol ; 212(3): 107636, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33039511

RESUMO

Bone mineralization is a highly specific and dynamic nanoscale process that has been studied extensively from a structural, chemical, and biological standpoint. Bone tissue, therefore, may be defined by the interplay of its intricately mineralized matrix and the cells that regulate its biological function. However, the far majority of engineered bone model systems and bone replacement materials have been unable to replicate this key characteristic of bone tissue; that is, the ability of cells to be gradually and rapidly embedded in a three-dimensional (3D) heavily calcified matrix material. Here we review the characteristics that define the bone matrix from a nanostructural perspective. We then revisit the benefits and challenges of existing model systems and engineered bone replacement materials, and discuss recent efforts to replicate the biological, cellular, mechanical, and materials characteristics of bone tissue on the nano- to microscale. We pay particular attention to a recently proposed method developed by our group, which seeks to replicate key aspects of the entrapment of bone cells within a mineralized matrix with precisions down to the level of individual nano-crystallites, inclusive of the bone vasculature, and osteogenic differentiation process. In summary, this paper discusses existing and emerging evidence pointing towards future developments bridging the gap between the fields of biomineralization, structural biology, stem cells, and tissue engineering, which we believe will hold the key to engineer truly functional bone-like tissue in the laboratory.


Assuntos
Osso e Ossos/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Hidrogéis/farmacologia , Nanoestruturas/química , Diferenciação Celular/efeitos dos fármacos , Humanos , Osteogênese/efeitos dos fármacos , Engenharia Tecidual/métodos
8.
Connect Tissue Res ; 58(5): 414-423, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27657550

RESUMO

AIMS: The structural organization of collagen from mineralized tissues, such as dentin and bone, has been a topic of debate in the recent literature. Recent reports have presented novel interpretations of the complexity of collagen type I at different hierarchical levels and in different tissues. Here, we investigate the nanostructural organization of demineralized dentin collagen following the digestion of non-collagenous components with a trypsin enzyme. MATERIALS AND METHODS: Dentin specimens were obtained from healthy third-molars, cut into small cubes, and polished down to 1 µm roughness. Samples were then demineralized with 10% citric acid for 2 min. Selected specimens were further treated with a solution containing 1 mg/ml trypsin for 48 hours at 37 °C (pH 7.9-9.0). Both untreated and trypsin digested samples were analyzed using SDS-PAGE, Field Emission Scanning Electron Microscopy (FE-SEM), and nanoindentation, where surface hardness and creep properties were compared before and after treatments. RESULTS: FE-SEM images of demineralized dentin showed the banded morphology of D-periodical collagen type I, which upon enzymatic digestion with trypsin appeared to dissociate longitudinally, consistently unraveling ~20 nm structures (microfibril bundles). Such nanoscale structures, to the best of our knowledge, have not been characterized in dentin previously. Mechanical characterization via nanoindentation showed that the unraveling of such microfibril bundles affected the creep displacement and creep rate of demineralized dentin. CONCLUSION: In summary, our results provide novel evidence of the organization of collagen type I from dentin, which may have important implications for the interaction of dental materials with the organic dentin matrix and the mechanical properties of mineralized tissues.


Assuntos
Colágeno Tipo I/química , Dentina/química , Matriz Extracelular/química , Dente Molar/química , Matriz Extracelular/ultraestrutura , Humanos , Dente Molar/ultraestrutura
9.
Adv Exp Med Biol ; 881: 79-94, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26545745

RESUMO

Survival of functional tissue constructs of clinically relevant size depends on the formation of an organized and uniformly distributed network of blood vessels and capillaries. The lack of such vasculature leads to spatio-temporal gradients in oxygen, nutrients and accumulation of waste products inside engineered tissue constructs resulting in negative biological events at the core of the scaffold. Unavailability of a well-defined vasculature also results in ineffective integration of scaffolds to the host vasculature upon implantation. Arguably, one of the greatest challenges in engineering clinically relevant bone substitutes, therefore, has been the development of vascularized bone scaffolds. Various approaches ranging from peptide and growth factor functionalized biomaterials to hyper-porous scaffolds have been proposed to address this problem with reasonable success. An emerging alternative to address this challenge has been the fabrication of pre-vascularized scaffolds by taking advantage of biomanufacturing techniques, such as soft- and photo-lithography or 3D bioprinting, and cell-based approaches, where functional capillaries are engineered in cell-laden scaffolds prior to implantation. These strategies seek to engineer pre-vascularized tissues in vitro, allowing for improved anastomosis with the host vasculature upon implantation, while also improving cell viability and tissue development in vitro. This book chapter provides an overview of recent methods to engineer pre-vascularized scaffolds for bone regeneration. We first review the development of functional blood capillaries in bony structures and discuss controlled delivery of growth factors, co-culture systems, and on-chip studies to engineer vascularized cell-laden biomaterials. Lastly, we review recent studies using microfabrication techniques and 3D printing to engineer pre-vascularized scaffolds for bone tissue engineering.


Assuntos
Regeneração Óssea/fisiologia , Osso e Ossos/fisiologia , Neovascularização Fisiológica/fisiologia , Engenharia Tecidual/métodos , Alicerces Teciduais , Materiais Biocompatíveis/metabolismo , Osso e Ossos/irrigação sanguínea , Osso e Ossos/citologia , Técnicas de Cocultura/métodos , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/tendências
10.
bioRxiv ; 2024 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-38352428

RESUMO

Tissue development, function, and disease are largely driven by the spatial organization of individual cells and their cell-cell interactions. Precision engineered tissues with single-cell spatial resolution, therefore, have tremendous potential for next generation disease models, drug discovery, and regenerative therapeutics. Despite significant advancements in biofabrication approaches to improve feature resolution, strategies to fabricate tissues with the exact same organization of individual cells in their native cellular microenvironment have remained virtually non-existent to date. Here we report a method to spatially pattern single cells with up to eight cell phenotypes and subcellular spatial precision. As proof-of-concept we first demonstrate the ability to systematically assess the influence of cellular microenvironments on cell behavior by controllably altering the spatial arrangement of cell types in bioprinted precision cell-cell interaction arrays. We then demonstrate, for the first time, the ability to produce high-fidelity replicas of a patient's annotated cancer biopsy with subcellular resolution. The ability to replicate native cellular microenvironments marks a significant advancement for precision biofabricated in-vitro models, where heterogenous tissues can be engineered with single-cell spatial precision to advance our understanding of complex biological systems in a controlled and systematic manner.

11.
Biomater Adv ; 159: 213805, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38457904

RESUMO

Bone defects may occur in different sizes and shapes due to trauma, infections, and cancer resection. Autografts are still considered the primary treatment choice for bone regeneration. However, they are hard to source and often create donor-site morbidity. Injectable microgels have attracted much attention in tissue engineering and regenerative medicine due to their ability to replace inert implants with a minimally invasive delivery. Here, we developed novel cell-laden bioprinted gelatin methacrylate (GelMA) injectable microgels, with controllable shapes and sizes that can be controllably mineralized on the nanoscale, while stimulating the response of cells embedded within the matrix. The injectable microgels were mineralized using a calcium and phosphate-rich medium that resulted in nanoscale crystalline hydroxyapatite deposition and increased stiffness within the crosslinked matrix of bioprinted GelMA microparticles. Next, we studied the effect of mineralization in osteocytes, a key bone homeostasis regulator. Viability stains showed that osteocytes were maintained at 98 % viability after mineralization with elevated expression of sclerostin in mineralized compared to non-mineralized microgels, showing that mineralization can effectively enhances osteocyte maturation. Based on our findings, bioprinted mineralized GelMA microgels appear to be an efficient material to approximate the bone microarchitecture and composition with desirable control of sample injectability and polymerization. These bone-like bioprinted mineralized biomaterials are exciting platforms for potential minimally invasive translational methods in bone regenerative therapies.


Assuntos
Gelatina , Microgéis , Gelatina/farmacologia , Gelatina/química , Materiais Biocompatíveis , Metacrilatos/química
12.
Biomicrofluidics ; 18(2): 021502, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38464668

RESUMO

Head and neck cancers (HNCs) rank as the sixth most common cancer globally and result in over 450 000 deaths annually. Despite considerable advancements in diagnostics and treatment, the 5-year survival rate for most types of HNCs remains below 50%. Poor prognoses are often attributed to tumor heterogeneity, drug resistance, and immunosuppression. These characteristics are difficult to replicate using in vitro or in vivo models, culminating in few effective approaches for early detection and therapeutic drug development. Organs-on-a-chip offer a promising avenue for studying HNCs, serving as microphysiological models that closely recapitulate the complexities of biological tissues within highly controllable microfluidic platforms. Such systems have gained interest as advanced experimental tools to investigate human pathophysiology and assess therapeutic efficacy, providing a deeper understanding of cancer pathophysiology. This review outlines current challenges and opportunities in replicating HNCs within microphysiological systems, focusing on mimicking the soft, glandular, and hard tissues of the head and neck. We further delve into the major applications of organ-on-a-chip models for HNCs, including fundamental research, drug discovery, translational approaches, and personalized medicine. This review emphasizes the integration of organs-on-a-chip into the repertoire of biological model systems available to researchers. This integration enables the exploration of unique aspects of HNCs, thereby accelerating discoveries with the potential to improve outcomes for HNC patients.

13.
Adv Healthc Mater ; 12(11): e2200976, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36808718

RESUMO

Bone autografts remain the gold standard for bone grafting surgeries despite having increased donor site morbidity and limited availability. Bone morphogenetic protein-loaded grafts represent another successful commercial alternative. However, the therapeutic use of recombinant growth factors has been associated with significant adverse clinical outcomes. This highlights the need to develop biomaterials that closely approximate the structure and composition of bone autografts, which are inherently osteoinductive and biologically active with embedded living cells, without the need for added supplements. Here, injectable growth factor-free bone-like tissue constructs are developed, that closely approximate the cellular, structural, and chemical composition of bone autografts. It is demonstrated that these micro-constructs are inherently osteogenic, and demonstrate the ability to stimulate mineralized tissue formation and regenerate bone in critical-sized defects in-vivo. Furthermore, the mechanisms that allow human mesenchymal stem cells (hMSCs) to be highly osteogenic in these constructs, despite the lack of osteoinductive supplements, are assessed, whereby Yes activated protein (YAP) nuclear localization and adenosine signaling appear to regulate osteogenic cell differentiation. The findings represent a step toward a new class of minimally invasive, injectable, and inherently osteoinductive scaffolds, which are regenerative by virtue of their ability to mimic the tissue cellular and extracellular microenvironment, thus showing promise for clinical applications in regenerative engineering.


Assuntos
Microgéis , Humanos , Regeneração Óssea/fisiologia , Osteogênese/fisiologia , Osso e Ossos , Materiais Biocompatíveis/química , Diferenciação Celular/fisiologia , Engenharia Tecidual , Alicerces Teciduais/química
14.
Front Immunol ; 14: 1162905, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37081897

RESUMO

Augmenting adaptive immunity is a critical goal for developing next-generation cancer therapies. T and B cells infiltrating the tumor dramatically influence cancer progression through complex interactions with the local microenvironment. Cancer cells evade and limit these immune responses by hijacking normal immunologic pathways. Current experimental models using conventional primary cells, cell lines, or animals have limitations for studying cancer-immune interactions directly relevant to human biology and clinical translation. Therefore, engineering methods to emulate such interplay at local and systemic levels are crucial to expedite the development of better therapies and diagnostic tools. In this review, we discuss the challenges, recent advances, and future directions toward engineering the tumor-immune microenvironment (TME), including key elements of adaptive immunity. We first offer an overview of the recent research that has advanced our understanding of the role of the adaptive immune system in the tumor microenvironment. Next, we discuss recent developments in 3D in-vitro models and engineering approaches that have been used to study the interaction of cancer and stromal cells with B and T lymphocytes. We summarize recent advancement in 3D bioengineering and discuss the need for 3D tumor models that better incorporate elements of the complex interplay of adaptive immunity and the tumor microenvironment. Finally, we provide a perspective on current challenges and future directions for modeling cancer-immune interactions aimed at identifying new biological targets for diagnostics and therapeutics.


Assuntos
Neoplasias , Animais , Humanos , Neoplasias/patologia , Microambiente Tumoral
15.
Adv Healthc Mater ; 12(22): e2202840, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37219011

RESUMO

Successful integration of cell-laden tissue constructs with host vasculature depends on the presence of functional capillaries to provide oxygen and nutrients to the embedded cells. However, diffusion limitations of cell-laden biomaterials challenge regeneration of large tissue defects that require bulk-delivery of hydrogels and cells. Herein, a strategy to bioprint geometrically controlled, endothelial and stem-cell laden microgels in high-throughput is introduced, allowing these cells to form mature and functional pericyte-supported vascular capillaries in vitro, and then injecting these pre-vascularized constructs minimally invasively in-vivo. It is demonstrated that this approach offers both desired scalability for translational applications as well as unprecedented levels of control over multiple microgel parameters to design spatially-tailored microenvironments for better scaffold functionality and vasculature formation. As a proof-of-concept, the regenerative capacity of the bioprinted pre-vascularized microgels is compared with that of cell-laden monolithic hydrogels of the same cellular and matrix composition in hard-to-heal defects in vivo. The results demonstrate that the bioprinted microgels have faster and higher connective tissue formation, more vessels per area, and widespread presence of functional chimeric (human and murine) vascular capillaries across regenerated sites. The proposed strategy, therefore, addresses a significant issue in regenerative medicine, demonstrating a superior potential to facilitate translational regenerative efforts.


Assuntos
Bioimpressão , Microgéis , Camundongos , Humanos , Animais , Engenharia Tecidual/métodos , Bioimpressão/métodos , Materiais Biocompatíveis , Hidrogéis , Alicerces Teciduais , Impressão Tridimensional
16.
bioRxiv ; 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37873385

RESUMO

Bone defects may occur in different sizes and shapes due to trauma, infections, and cancer resection. Autografts are still considered the primary treatment choice for bone regeneration. However, they are hard to source and often create donor-site morbidity. Injectable microgels have attracted much attention in tissue engineering and regenerative medicine due to their ability to replace inert implants with a minimally invasive delivery. Here, we developed novel cell-laden bioprinted gelatin methacrylate (GelMA) injectable microgels, with controllable shapes and sizes that can be controllably mineralized on the nanoscale, while stimulating the response of cells embedded within the matrix. The injectable microgels were mineralized using a calcium and phosphate-rich medium that resulted in nanoscale crystalline hydroxyapatite deposition and increased stiffness within the crosslinked matrix of bioprinted GelMA microparticles. Next, we studied the effect of mineralization in osteocytes, a key bone homeostasis regulator. Viability stains showed that osteocytes were maintained at 98% viability after mineralization with elevated expression of sclerostin in mineralized compared to non-mineralized microgels, indicating that mineralization effectively enhances osteocyte maturation. Based on our findings, bioprinted mineralized GelMA microgels appear to be an efficient material to approximate the bone microarchitecture and composition with desirable control of sample injectability and polymerization. These bone-like bioprinted mineralized biomaterials are exciting platforms for potential minimally invasive translational methods in bone regenerative therapies.

17.
Adv Mater ; 34(3): e2101321, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35060652

RESUMO

Bioprinting has emerged as one of the most promising strategies for fabrication of functional organs in the lab as an alternative to transplant organs. While progress in the field has mostly been restricted to a few miniaturized tissues with minimal biological functionality until a few years ago, recent progress has advanced the concept of building three-dimensional multicellular organ complexity remarkably. This review discusses a series of milestones that have paved the way for bioprinting of tissue constructs that have advanced levels of biological and architectural functionality. Critical materials, engineering and biological challenges that are key to addressing the desirable function of engineered organs are presented. These are discussed in light of the many difficulties to replicate the heterotypic organization of multicellular solid organs, the nanoscale precision of the extracellular microenvironment in hierarchical tissues, as well as the advantages and limitations of existing bioprinting methods to adequately overcome these barriers. In summary, the advances of the field toward realistic manufacturing of functional organs have never been so extensive, and this manuscript serves as a road map for some of the recent progress and the challenges ahead.


Assuntos
Bioimpressão , Bioimpressão/métodos , Impressão Tridimensional , Engenharia Tecidual/métodos
18.
bioRxiv ; 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35411345

RESUMO

Pericytes stabilize blood vessels and promote vascular barrier function. However, vessels subjected to pro-inflammatory conditions have impaired barrier function, which has been suggested to potentially expose perivascular cells to SARS-CoV-2. To test this hypothesis, we engineered pericyte-supported vascular capillaries on-a-chip, and determined that the extravasation and binding of spike protein (S1) on perivascular cells of inflamed vessels to be significantly higher that in healthy controls, indicating a potential target to understand COVID-19 vascular complications.

19.
J Dent ; 121: 104111, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35460865

RESUMO

OBJECTIVES: Self-assembling peptide P11-4 is amphiphilic and pH-triggered, effective on repairing early enamel carious lesions and dentin remineralization. However, P11-4 effects on dentin biomineralization and repair ability remain unexplored. Thus, cytocompatibility and effectiveness of P11-4 on inducing mineralization and migration of odontoblast-like cells (MDPC-23) were investigated. METHODS: MDPC-23 were seeded in contact with P11-4 (0.5 and 1 µg/ml), Dentin Matrix Protein 1 (DMP1 0.5 and 1 µg/ml) or Calcium hydroxide (Ca(OH)2 100 µg/ml) solutions. Cell viability was verified using MTT (n = 6/group). Mineral deposition was tested using Alizarin Red (n = 4/group). Cell migration was assessed by light microscopy (n = 2/group). MTT and Alizarin Red data were compared using Kruskal-Wallis and Mann-Whitney (α=0.01). RESULTS: P11-4 (0.5 and 1 µg/ml) and DMP1 (0.5 and 1 µg/ml) resulted the highest cell viability; Ca(OH)2 presented the lowest. 1 µg/ml DMP1 and 1 µg/ml P11-4 promoted the highest mineral deposition. Ca(OH)2 presented lower values of mineral deposits than DMP1 1 µg/ml (p < 0.01), but similar to P11-4 1 µg/ml. P11-4 and DMP1 at 0.5 µg/ml induced lesser mineral precipitation than P11-4 and DMP1 at 1 µg/ml (p < 0.01), with no difference to Ca(OH)2. All materials stimulated cell migration, however, lower concentrations of DMP1 and P11-4 demonstrated a higher migration potential. CONCLUSION: P11-4 did not affect cell viability, induces mineral deposition and MDPC-23 migration like DMP1. CLINICAL SIGNIFICANCE: Self-assembling peptide P11-4 does not affect the cell viability and induces mineral deposition comparable to native protein involved in biomineralization. Combined with its ability to bind type I collagen, P11-4 is a promising bioinspired molecule that provides native-tissue conditions and foster further studies on its ability to form dentin bridges in pulp-capping strategies.


Assuntos
Glicosiltransferases , Odontoblastos , Movimento Celular , Esmalte Dentário/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fosfoproteínas/metabolismo
20.
Acta Biomater ; 150: 58-66, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35933103

RESUMO

Dental caries is a biofilm-mediated, diet-modulated, multifactorial and dynamic disease that affects more than 90% of adults in Western countries. The current treatment for decayed tissue is based on using materials to replace the lost enamel or dentin. More than 500 million dental restorations are placed annually worldwide, and materials used for these purposes either directly or indirectly interact with dentin and pulp tissues. The development and understanding of the effects of restorative dental materials are based on different in-vitro and in-vivo tests, which have been evolving with time. In this review, we first discuss the characteristics of the tooth and the dentin-pulp interface that are unique for materials testing. Subsequently, we discuss frequently used in-vitro tests to evaluate the biocompatibility of dental materials commonly used for restorative procedures. Finally, we present our perspective on the future directions for biological research on dental materials using tissue engineering and organs on-a-chip approaches. STATEMENT OF SIGNIFICANCE: Dental caries is still the most prevalent infectious disease globally, requiring more than 500 million restorations to be placed every year. Regrettably, the failure rates of such restorations are still high. Those rates are partially based on the fact that current platforms to test dental materials are somewhat inaccurate in reproducing critical components of the complex oral microenvironment. Thus, there is a collective effort to develop new materials while evolving the platforms to test them. In this context, the present review critically discusses in-vitro models used to evaluate the biocompatibility of restorative dental materials and brings a perspective on future directions for tissue-engineered and organs-on-a-chip platforms for testing new dental materials.


Assuntos
Cárie Dentária , Dentina , Adulto , Resinas Compostas , Materiais Dentários/farmacologia , Restauração Dentária Permanente , Humanos , Dispositivos Lab-On-A-Chip , Teste de Materiais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA