Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 270(Pt 2): 132409, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38768918

ABSTRACT

Suture pull-through is a clinical problem in meniscus repair surgery due to the sharp leading edge of sutures. Several tissue adhesives have been developed as an alternative to traditional suturing; however, there is still no suitable tissue adhesive specific for meniscus repair treatment due to unsatisfactory biosafety, biodegradable, sterilizable, and tissue-bonding characteristics. In this study, we used a tissue adhesive composed of chitosan hydrochloride reacted with oxidative periodate-oxidized dextran (ChitHCl-DDA) combined with a chitosan-based hydrogel and oxidative dextran to attach to the meniscus. We conducted viscoelastic tests, viscosity tests, lap shear stress tests, Fourier transform infrared (FTIR) spectroscopy, swelling ratio tests, and degradation behavior tests to characterize these materials. An MTT assay, alcian blue staining, migration assay, cell behavior observations, and protein expression tests were used to understand cell viability and responses. Moreover, ex vivo and in vivo tests were used to analyze tissue regeneration and biocompatibility of the ChitHCl-DDA tissue adhesive. Our results revealed that the ChitHCl-DDA tissue adhesive provided excellent tissue adhesive strength, cell viability, and cell responses. This tissue adhesive has great potential for torn meniscus tissue repair and regeneration.


Subject(s)
Biocompatible Materials , Chitosan , Regeneration , Tissue Adhesives , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Animals , Regeneration/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Materials Testing , Meniscus/drug effects , Dextrans/chemistry , Cell Survival/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Rabbits , Tibial Meniscus Injuries/surgery , Humans , Injections
2.
bioRxiv ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38712035

ABSTRACT

Formation of chondromimetic human mesenchymal stem cells (hMSCs) condensations typically required in vitro culture in defined environments. In addition, extended in vitro culture in differentiation media over several weeks is usually necessary prior to implantation, which is costly, time consuming and delays clinical treatment. Here, this study reports on immediately implantable core/shell microgels with a high-density hMSC-laden core and rapidly degradable hydrogel shell. The hMSCs in the core formed cell condensates within 12 hours and the oxidized and methacrylated alginate (OMA) hydrogel shells were completely degraded within 3 days, enabling spontaneous and precipitous fusion of adjacent condensed aggregates. By delivering transforming growth factor-ß1 (TGF-ß1) within the core, the fused condensates were chondrogenically differentiated and formed cartilage microtissues. Importantly, these hMSC-laden core/shell microgels, fabricated without any in vitro culture, were subcutaneously implanted into mice and shown to form cartilage tissue via cellular condensations in the core after 3 weeks. This innovative approach to form cell condensations in situ without in vitro culture that can fuse together with each other and with host tissue and be matured into new tissue with incorporated bioactive signals, allows for immediate implantation and may be a platform strategy for cartilage regeneration and other tissue engineering applications.

3.
Research (Wash D C) ; 7: 0365, 2024.
Article in English | MEDLINE | ID: mdl-38654733

ABSTRACT

Neutrophils are primed for neutrophil extracellular trap (NET) formation during diabetes, and excessive NET formation from primed neutrophils compromises wound healing in patients with diabetes. Here, we demonstrate that trained immunity mediates diabetes-induced NET priming in neutrophils. Under diabetic conditions, neutrophils exhibit robust metabolic reprogramming comprising enhanced glycolysis via the pentose phosphate pathway and fatty acid oxidation, which result in the accumulation of acetyl-coenzyme A. Adenosine 5'-triphosphate-citrate lyase-mediated accumulation of acetyl-coenzyme A and histone acetyltransferases further induce the acetylation of lysine residues on histone 3 (AcH3K9, AcH3K14, and AcH3K27) and histone 4 (AcH4K8). The pharmacological inhibition of adenosine 5'-triphosphate-citrate lyase and histone acetyltransferases completely inhibited high-glucose-induced NET priming. The trained immunity of neutrophils was further confirmed in neutrophils isolated from patients with diabetes. Our findings suggest that trained immunity mediates functional changes in neutrophils in diabetic environments, and targeting neutrophil-trained immunity may be a potential therapeutic target for controlling inflammatory complications of diabetes.

4.
Tissue Eng Regen Med ; 20(7): 1173-1190, 2023 12.
Article in English | MEDLINE | ID: mdl-37843784

ABSTRACT

BACKGROUND: The emergence of various infectious diseases and the toxic effects of hyperinflammation by biotherapeutics have highlighted the need for in vitro preclinical models mimicking the human immune system. In vitro models studying the relationship between hyperinflammation and acute renal injury mainly rely on 2D culture systems, which have shown limitations in recapitulating kidney function. Herein, we developed an in vitro kidney toxicity model by co-culturing 3D engineered kidney proximal tubules cells (RPTEC/TERT1) with human peripheral blood mononuclear cells (PBMC). METHODS: RPTEC/TERT1 were sandwich cultured to form 3D renal tubules for 16 days. The tubules were then co-cultured with PBMC using transwell (0.4 µm pores) for 24 h. Hyperinflammation of PBMC was induced during co-culture using polyinosinic-polycytidylic acid (polyI:C) and lipopolysaccharide (LPS) to investigate the effects of the induced hyperinflammation on the renal tubules. RESULTS: Encapsulated RPTEC/TERT1 cells in Matrigel exhibited elevated renal function markers compared to 2D culture. The coexistence of PBMC and polyI:C induced a strong inflammatory response in the kidney cells. This hyperinflammation significantly reduced primary cilia formation and upregulated kidney injury markers along the 3D tubules. Similarly, treating co-cultured PBMC with LPS to induce hyperinflammation resulted in comparable inflammatory responses and potential kidney injury. CONCLUSION: The model demonstrated similar changes in kidney injury markers following polyI:C and LPS treatment, indicating its suitability for detecting immune-associated kidney damage resulting from infections and biopharmaceutical applications.


Subject(s)
Leukocytes, Mononuclear , Lipopolysaccharides , Humans , Coculture Techniques , Cell Line , Inflammation
5.
Tissue Eng Part C Methods ; 29(10): 447-458, 2023 10.
Article in English | MEDLINE | ID: mdl-37440328

ABSTRACT

Muscle tissue engineering has been the focus of extensive research due to its potential for numerous medical applications, including ex vivo actuator development and clinical treatments. In this study, we developed a method for harvesting muscle fiber in a floatable and translocatable manner utilizing thermally expandable hydrogels with a chemically patterned polydopamine (PD) layer generated by microcontact printing (µCP). The µCP of PD on the hydrogel facilitated the formation of stripe patterns with varying widths of printed/nonprinted area (50/50, 100/100, and 200/200 µm). The spatially controlled adhesion of C2C12 myoblasts on the PD patterns produced clearly distinguishable muscle fibers, and translocated muscle fibers exhibited preserved extracellular matrix and junction proteins. Furthermore, the development of anisotropic arrangements and mature myotubes within the fibers suggests the potential for functional control of engineered muscle tissues. Overall, the muscle fiber harvesting method developed herein is suitable for both translocation and floating and is a promising technique for muscle tissue engineering as it mimics the structure-function relationship of natural tissue.


Subject(s)
Hydrogels , Muscle Fibers, Skeletal , Tissue Engineering/methods , Myoblasts , Extracellular Matrix , Tissue Scaffolds
6.
ACS Omega ; 7(22): 18471-18480, 2022 Jun 07.
Article in English | MEDLINE | ID: mdl-35694497

ABSTRACT

Cell culture technology has evolved into three-dimensional (3D) artificial tissue models for better reproduction of human native tissues. However, there are some unresolved limitations that arise due to the adhesive properties of cells. In this study, we developed a hexanoyl glycol chitosan (HGC) as a non-cell adhesive polymer for scaffold-based and -free 3D culture. The uniform cell distribution in a porous scaffold was well maintained during the long culutre period on the HGC-coated substrate by preventing ectopic adhesion and migration of cells on the substrate. In addition, when culturing many spheroids in one dish, supplementation of the culture medium with HGC prevented the aggregation of spheroids and maintained the shape and size of spheroids for a long culture duration. Collectively, the use of HGC in 3D culture systems is expected to contribute greatly to creating excellent regenerative therapeutics and screening models of bioproducts.

7.
Tissue Eng Regen Med ; 19(5): 1089-1098, 2022 10.
Article in English | MEDLINE | ID: mdl-35551635

ABSTRACT

BACKGROUND: Tissue engineering approaches to treat damaged bone include various tissue transplants such as autologous, allogeneic, and xenografts. Artificial materials have been widely introduced to meet the demand for graft materials, but insufficiency in supply is still not resolved. In this study, human adipose tissue, easily obtained from the human body, was harvested, and the tissue was decellularized to fabricate a decellularized human adipose tissue matrix (DM) as an alternative graft material. METHODS: Human adipose tissue was obtained via liposuction. The obtained fresh adipose tissue sample was cut into pieces then put into decellularization solution (1% antibiotic-antimycotic solution and 1% phenylmethanesulphonyl fluoride). Lipids were further removed via treatment in isopropanol. The sample was then subjected to another enzymatic digestion and lipid removal processes. The obtained decellularized adipose tissue matrix was lyophilized to form a graft material in disc shape. RESULTS: Decellularization was confirmed by nuclear staining methods and detection of RNA and DNA via PCR. Bone morphogenetic protein 2 (BMP2)-loaded DM showed the ability to form new bone tissue when implanted in subcutaneous tissue. In recovery of a mouse calvarial defect model, BMP2-loaded DM exhibited similar levels of bone tissue regeneration efficiency compared with a well-defined commercial product, BMP2-loaded CollaCote®. CONCLUSION: The DM developed in this study is expected to address the problem of insufficient supply of graft materials and contribute to the treatment of bone defects of critical size as an alternative bone graft material with preserved extracellular matrix components.


Subject(s)
Bone Morphogenetic Protein 2 , Tissue Scaffolds , 2-Propanol/metabolism , Adipose Tissue , Animals , Anti-Bacterial Agents , Bone Morphogenetic Protein 2/metabolism , Bone Regeneration , DNA/metabolism , Extracellular Matrix/metabolism , Fluorides/metabolism , Humans , Lipids , Mice , RNA/metabolism
8.
Bioact Mater ; 15: 185-193, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35386348

ABSTRACT

Recently, 3D bioprinting has been explored as a promising technology for biomedical applications with the potential to create complex structures with precise features. Cell encapsulated hydrogels composed of materials such as gelatin, collagen, hyaluronic acid, alginate and polyethylene glycol have been widely used as bioinks for 3D bioprinting. However, since most hydrogel-based bioinks may not allow rapid stabilization immediately after 3D bioprinting, achieving high resolution and fidelity to the intended architecture is a common challenge in 3D bioprinting of hydrogels. In this study, we have utilized shear-thinning and self-healing ionically crosslinked oxidized and methacrylated alginates (OMAs) as a bioink, which can be rapidly gelled by its self-healing property after bioprinting and further stabilized via secondary crosslinking. It was successfully demonstrated that stem cell-laden calcium-crosslinked OMA hydrogels can be bioprinted into complicated 3D tissue structures with both high resolution and fidelity. Additional photocrosslinking enables long-term culture of 3D bioprinted constructs for formation of functional tissue by differentiation of encapsulated human mesenchymal stem cells.

9.
Mater Sci Eng C Mater Biol Appl ; 128: 112309, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34474860

ABSTRACT

Recently, black phosphorus (BP) has garnered great attention as one of newly emerging two-dimensional nanomaterials. Especially, the degraded platelets of BP in the physiological environment were shown to be nontoxic phosphate anions, which are a component of bone tissue and can be used for mineralization. Here, our study presents the potential of BP as biofunctional and biocompatible nanomaterials for the application to bone tissue engineering and regeneration. An ultrathin layer of BP nanodots (BPNDs) was created on a glass substrate by using a flow-enabled self-assembly process, which yielded a highly uniform deposition of BPNDs in a unique confined geometry. The BPND-coated substrates represented unprecedented favorable topographical microenvironments and supportive matrices suitable for the growth and survival of MC3T3-E1 preosteoblasts. The prepared substrates promoted the spontaneous osteodifferentiation of preosteoblasts, which had been confirmed by determining alkaline phosphatase activity and extracellular calcium deposition as early- and late-stage markers of osteogenic differentiation, respectively. Furthermore, the BPND-coated substrates upregulated the expression of some specific genes (i.e., RUNX2, OCN, OPN, and Vinculin) and proteins, which are closely related to osteogenesis. Conclusively, our BPND-coating strategy suggests that a biologically inert surface can be readily activated as a cell-favorable nanoplatform enabled with excellent biocompatibility and osteogenic ability.


Subject(s)
Osteoblasts , Osteogenesis , Cell Differentiation , Phosphorus , Tissue Engineering
10.
Adv Funct Mater ; 31(24)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34335134

ABSTRACT

Developing and healing tissues begin as a cellular condensation. Spatiotemporal changes in tissue geometry, transformations in the spatial distribution of the cells and extracellular matrix, are essential for its evolution into a functional tissue. 4D materials, 3D materials capable of geometric changes, may have the potential to recreate the aforementioned biological phenomenon. However, most reported 4D materials are non-degradable and/or not biocompatible, which limits their application in regenerative medicine, and to date there are no systems controlling the geometry of high density cellular condensations and differentiation. Here, we describe 4D high cell density tissues based on shape-changing hydrogels. By sequential photocrosslinking of oxidized and methacrylated alginate (OMA) and methacrylated gelatin (GelMA), bi-layered hydrogels presenting controllable geometric changes without any external stimuli were fabricated. Fibroblasts and human adipose-derived stem cells (ASCs) were incorporated at concentrations up to 1.0 × 108 cells/mL to the 4D constructs, and controllable shape changes were achieved in concert with ASCs differentiated down chondrogenic and osteogenic lineages. Bioprinting of the high density cell-laden OMA and GelMA permitted the formation of more complex constructs with defined 4D geometric changes, which may further expand the promise of this approach in regenerative medicine applications.

11.
Immune Netw ; 21(2): e16, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33996172

ABSTRACT

Patients with severe coronavirus disease 2019 (COVID-19) demonstrate dysregulated immune responses including exacerbated neutrophil functions. Massive neutrophil infiltrations accompanying neutrophil extracellular trap (NET) formations are also observed in patients with severe COVID-19. However, the mechanism underlying severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced NET formation has not yet been elucidated. Here we show that 2 viral proteins encoded by SARS-CoV-2, the nucleocapsid protein and the whole spike protein, induce NET formation from neutrophils. NET formation was ROS-independent and was completely inhibited by the spleen tyrosine kinase inhibition. The inhibition of p38 MAPK, protein kinase C, and JNK signaling pathways also inhibited viral protein-induced NET formation. Our findings demonstrate one method by which SARS-CoV-2 evades innate immunity and provide a potential target for therapeutics to treat patients with severe COVID-19.

12.
Adv Sci (Weinh) ; 8(9): 2004616, 2021 05.
Article in English | MEDLINE | ID: mdl-33977070

ABSTRACT

Shape-morphing hydrogels bear promising prospects as soft actuators and for robotics. However, they are mostly restricted to applications in the abiotic domain due to the harsh physicochemical conditions typically necessary to induce shape morphing. Here, multilayer hydrogel actuator systems are developed using biocompatible and photocrosslinkable oxidized, methacrylated alginate and methacrylated gelatin that permit encapsulation and maintenance of living cells within the hydrogel actuators and implement programmed and controlled actuations with multiple shape changes. The hydrogel actuators encapsulating cells enable defined self-folding and/or user-regulated, on-demand-folding into specific 3D architectures under physiological conditions, with the capability to partially bioemulate complex developmental processes such as branching morphogenesis. The hydrogel actuator systems can be utilized as novel platforms for investigating the effect of programmed multiple-step and reversible shape morphing on cellular behaviors in 3D extracellular matrix and the role of recapitulating developmental and healing morphogenic processes on promoting new complex tissue formation.


Subject(s)
Alginates/chemistry , Biocompatible Materials/chemistry , Biomimetics/methods , Hydrogels/chemistry , Morphogenesis/physiology
13.
Sci Rep ; 11(1): 8289, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859336

ABSTRACT

Extracellular vesicles (EVs) are membrane-derived heterogeneous vesicles that mediate intercellular communications. They have recently been considered as ideal vehicles for drug-delivery systems, and immune cells are suggested as a potential source for drug-loaded EVs. In this study, we investigated the possibility of neutrophils as a source for drug-loaded EVs. Neutrophil-like differentiated human promyelocytic leukemia cells (dHL-60) produced massive amounts of EVs within 1 h. The dHL-60 cells are also easily loaded with various cargoes such as antibiotics (penicillin), anticancer drug (paclitaxel), chemoattractant (MCP-1), miRNA, and Cas9. The EVs derived from the dHL-60 cells showed efficient incorporation of these cargoes and significant effector functions, such as bactericidal activity, monocyte chemotaxis, and macrophage polarization. Our results suggest that neutrophils or neutrophil-like promyelocytic cells could be an attractive source for drug-delivery EVs.


Subject(s)
Drug Delivery Systems , Extracellular Vesicles , Granulocyte Precursor Cells , Anti-Bacterial Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Communication , Cell Differentiation , Cells, Cultured , Chemokine CCL2/administration & dosage , Granulocyte Precursor Cells/cytology , Humans , Neutrophils/cytology , Paclitaxel/administration & dosage , Penicillins/administration & dosage
14.
Acta Biomater ; 126: 154-169, 2021 05.
Article in English | MEDLINE | ID: mdl-33705989

ABSTRACT

For 3D bioprinted tissues to be scaled-up to clinically relevant sizes, effective prevascularisation strategies are required to provide the necessary nutrients for normal metabolism and to remove associated waste by-products. The aim of this study was to develop a bioprinting strategy to engineer prevascularised tissues in vitro and to investigate the capacity of such constructs to enhance the vascularisation and regeneration of large bone defects in vivo. From a screen of different bioinks, a fibrin-based hydrogel was found to best support human umbilical vein endothelial cell (HUVEC) sprouting and the establishment of a microvessel network. When this bioink was combined with HUVECs and supporting human bone marrow stem/stromal cells (hBMSCs), these microvessel networks persisted in vitro. Furthermore, only bioprinted tissues containing both HUVECs and hBMSCs, that were first allowed to mature in vitro, supported robust blood vessel development in vivo. To assess the therapeutic utility of this bioprinting strategy, these bioinks were used to prevascularise 3D printed polycaprolactone (PCL) scaffolds, which were subsequently implanted into critically-sized femoral bone defects in rats. Micro-computed tomography (µCT) angiography revealed increased levels of vascularisation in vivo, which correlated with higher levels of new bone formation. Such prevascularised constructs could be used to enhance the vascularisation of a range of large tissue defects, forming the basis of multiple new bioprinted therapeutics. STATEMENT OF SIGNIFICANCE: This paper demonstrates a versatile 3D bioprinting technique to improve the vascularisation of tissue engineered constructs and further demonstrates how this method can be incorporated into a bone tissue engineering strategy to improve vascularisation in a rat femoral defect model.


Subject(s)
Bioprinting , Animals , Printing, Three-Dimensional , Rats , Tissue Engineering , Tissue Scaffolds , X-Ray Microtomography
15.
Theranostics ; 11(6): 2770-2787, 2021.
Article in English | MEDLINE | ID: mdl-33456572

ABSTRACT

Aims: Extracellular vesicles (EVs) are membrane-derived vesicles that mediate intercellular communications. Neutrophils produce different subtypes of EVs during inflammatory responses. Neutrophil-derived trails (NDTRs) are generated by neutrophils migrating toward inflammatory foci, whereas neutrophil-derived microvesicles (NDMVs) are thought to be generated by neutrophils that have arrived at the inflammatory foci. However, the physical and functional characteristics of neutrophil-derived EVs are incompletely understood. In this study, we aimed to investigate the differences between NDTRs and NDMVs. Methods: The generation of neutrophil-derived EVs were visualized by live-cell fluorescence images and the physical characteristics were further analyzed using nanotracking analysis assay, scanning electron microscopic analysis, and marker expressions. Functional characteristics of neutrophil-derived EVs were analyzed using assays for bactericidal activity, monocyte chemotaxis, phenotype polarization of macrophages, and miRNA sequencing. Finally, the effects of neutrophil-derived EVs on the acute and chronic inflammation were examined in vivo. Results: Both EVs share similar characteristics including stimulators, surface marker expression, bactericidal activity, and chemoattractive effect on monocytes via MCP-1. However, the integrin-mediated physical interaction was required for generation of NDTRs whereas NDMV generation was dependent on PI3K pathway. Interestingly, NDTRs contained proinflammatory miRNAs such as miR-1260, miR-1285, miR-4454, and miR-7975, while NDMVs contained anti-inflammatory miRNAs such as miR-126, miR-150, and miR-451a. Although both EVs were easily uptaken by monocytes, NDTRs enhanced proinflammatory macrophage polarization whereas NDMVs induced anti-inflammatory macrophage polarization. Moreover, NDTRs showed protective effects against lethality in a murine sepsis model and pathological changes in a murine chronic colitis model. Conclusion: These results suggest that NDTR is a proinflammatory subtype of neutrophil-derived EVs distinguished from NDMV.


Subject(s)
Extracellular Vesicles/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Animals , Biomarkers/metabolism , Cell Communication/physiology , Cells, Cultured , Chemotaxis/physiology , Colitis/metabolism , Disease Models, Animal , Humans , Macrophage Activation/physiology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Monocytes/metabolism , Sepsis/metabolism , THP-1 Cells/metabolism
16.
Sci Adv ; 6(21): eaaz5913, 2020 05.
Article in English | MEDLINE | ID: mdl-32494742

ABSTRACT

Despite great progress in biomaterial design strategies for replacing damaged articular cartilage, prevention of stem cell-derived chondrocyte hypertrophy and resulting inferior tissue formation is still a critical challenge. Here, by using engineered biomaterials and a high-throughput system for screening of combinatorial cues in cartilage microenvironments, we demonstrate that biomaterial cross-linking density that regulates matrix degradation and stiffness-together with defined presentation of growth factors, mechanical stimulation, and arginine-glycine-aspartic acid (RGD) peptides-can guide human mesenchymal stem cell (hMSC) differentiation into articular or hypertrophic cartilage phenotypes. Faster-degrading, soft matrices promoted articular cartilage tissue formation of hMSCs by inducing their proliferation and maturation, while slower-degrading, stiff matrices promoted cells to differentiate into hypertrophic chondrocytes through Yes-associated protein (YAP)-dependent mechanotransduction. in vitro and in vivo chondrogenesis studies also suggest that down-regulation of the Wingless and INT-1 (WNT) signaling pathway is required for better quality articular cartilage-like tissue production.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cells , Biocompatible Materials/metabolism , Cartilage, Articular/metabolism , Cell Differentiation , Mechanotransduction, Cellular/physiology , Mesenchymal Stem Cells/metabolism , Phenotype , Stem Cells , Tissue Engineering/methods
17.
Biomaterials ; 245: 119973, 2020 07.
Article in English | MEDLINE | ID: mdl-32244091

ABSTRACT

The native extracellular matrix (ECM) contains a host of matricellular proteins and bioactive factors that regulate cell behavior, and many ECM components have been leveraged to guide cell fate. However, the large size and chemical characteristics of these constituents complicate their incorporation into biomaterials without interfering with material properties, motivating the need for alternative approaches to regulate cellular responses. Mesenchymal stromal cells (MSCs) can promote osseous regeneration in vivo directly or indirectly through multiple means including (1) secretion of proangiogenic and mitogenic factors to initiate formation of a vascular template and recruit host cells into the tissue site or (2) direct differentiation into osteoblasts. As MSC behavior is influenced by the properties of engineered hydrogels, we hypothesized that the biochemical and biophysical properties of alginate could be manipulated to promote the dual contributions of encapsulated MSCs toward bone formation. We functionalized alginate with QK peptide to enhance proangiogenic factor secretion and RGD to promote adhesion, while biomechanical-mediated osteogenic cues were controlled by modulating viscoelastic properties of the alginate substrate. A 1:1 ratio of QK:RGD resulted in the highest levels of both proangiogenic factor secretion and mineralization in vitro. Viscoelastic alginate outperformed purely elastic gels in both categories, and this effect was enhanced by stiffness up to 20 kPa. Furthermore, viscoelastic constructs promoted vessel infiltration and bone regeneration in a rat calvarial defect over 12 weeks. These data suggest that modulating viscoelastic properties of biomaterials, in conjunction with dual peptide functionalization, can simultaneously enhance multiple aspects of MSC regenerative potential and improve neovascularization of engineered tissues.


Subject(s)
Hydrogels , Mesenchymal Stem Cells , Animals , Cell Differentiation , Osteogenesis , Peptides , Rats , Stromal Cells
18.
Biomater Res ; 23: 23, 2019.
Article in English | MEDLINE | ID: mdl-31798945

ABSTRACT

BACKGROUND: Two-dimensional black phosphorus nanosheets (BPNSs) have recently emerged as a successive novel nanomaterial owing to their uniqueness in optical and electrical properties. Although BPNSs have found a wide range of biomedical applications, their biosafety is still a major concern to be addressed. METHODS: In this study, we have prepared layered BPNSs using liquid exfoliation procedure, and evaluated their physicochemical properties using Fourier Transform-infrared (FTIR) spectroscopy, Raman spectroscopy, atomic force microscopy, and Zetasizer analyses. We have investigated potential cytotoxicity of BPNSs against three different types of fibroblast cells, i.e. mouse embryonic fibroblast (NIH3T3), primary cultured normal human dermal fibroblast (nHDF), and fibrosarcoma (HT1080). Cell counting kit-8 (CCK-8) assay was carried out to assess cellular metabolic activity in cells whereas lactate dehydrogenase (LDH) activity assay was helpful to study plasma membrane integrity. RESULTS: Our salient research findings showed that BPNSs were polydispersed in solution due to aggregation. Toxic response of BPNSs against fibroblast cells was in the order, HT1080>nHDF>NIH3T3. The nanosheets reduced the number of cancerous cells with significant difference to normal cells. CONCLUSIONS: We suggest that BPNSs can be considered for cancer treatment as they destroy cancerous cells effectively. However, a comprehensive study is required to elucidate other biological effects of BPNSs.

19.
Acta Biomater ; 100: 158-172, 2019 12.
Article in English | MEDLINE | ID: mdl-31542503

ABSTRACT

Cell spheroids as building blocks for engineering micro-tissue should be able to mimic the complex structure of natural tissue. However, control of the distribution of multiple cell populations within cell spheroids is difficult to achieve with current spheroid-harvest methods such as hanging-drop and with the use of microwell plates. In this study, we report the fabrication of core-shell spheroids with the ultimate goal to form 3D complex micro-tissue. We used endothelial cells and two types of stem cells (human turbinate mesenchymal stem cells (hTMSCs)/adipose-derived stem cells (ADSCs)). The stem cells and endothelial cells formed layered micro-sized cell sheets (µCSs) on polydopamine micro-patterned temperature-responsive hydrogel surfaces by a sequential seeding method, and these layered µCSs self-assembled to form core-shell spheroids by expansion of the hydrogels. The co-cultured spheroids formed a core-shell structure irrespective of stem cell type. In addition, the size of the core-shell spheroids was controlled from 90 ± 1 to 144 ± 3 µm by changing pattern sizes (200, 300, and 400 µm). The shell thickness gradually increased from 12 ± 3 to 30 ± 6 µm by adjusting the endothelial cell seeding density. Finally, we fabricated the micro-tissue by fusion of the co-cultured spheroids, and the spheroids with the core-shell structure rapidly induced in vitro vessel-like network in 3 days. Thus, the position of endothelial cells in co-cultured spheroids may be an important factor for the modulation of the vascularization process, which can be useful for the production of 3D complex micro-tissues using spheroids as building blocks. STATEMENT OF SIGNIFICANCE: This manuscript describes our work on the fabrication of core-shell spheroids as building blocks to form 3D complex vascularized micro-tissue. Stem cells (human turbinate mesenchymal stem cells (hTMSCs) or adipose-derived stem cells (ADSCs)) and endothelial cells formed layered micro-sized cell sheets (µCSs) on micro-patterned temperature-responsive hydrogel surfaces by a sequential seeding method, and these layered µCSs self-assembled to form core-shell spheroids (core - stem cells, shell - endothelial cells), irrespective of stem cell type. In addition, the size and shell thickness of the core-shell spheroids were controlled by modifying pattern size and endothelial cell seeding density. We fabricated the vascularized micro-tissue by fusion of the spheroids and demonstrated that the spheroids with a core-shell structure rapidly induced vessel-like network.


Subject(s)
Microtechnology , Neovascularization, Physiologic , Spheroids, Cellular/cytology , Tissue Engineering/methods , Fluorescence , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Spheroids, Cellular/ultrastructure , Temperature
20.
Nanomaterials (Basel) ; 9(9)2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31466309

ABSTRACT

The zero (0-D) and one-dimensional (1-D) carbon nanomaterials have gained attention among researchers because they exhibit a larger surface area to volume ratio, and a smaller size. Furthermore, carbon is ubiquitously present in all living organisms. However, toxicity is a major concern while utilizing carbon nanomaterials for biomedical applications such as drug delivery, biosensing, and tissue regeneration. In the present review, we have summarized some of the recent findings of cellular and animal level toxicity studies of 0-D (carbon quantum dot, graphene quantum dot, nanodiamond, and carbon black) and 1-D (single-walled and multi-walled carbon nanotubes) carbon nanomaterials. The in vitro toxicity of carbon nanomaterials was exemplified in normal and cancer cell lines including fibroblasts, osteoblasts, macrophages, epithelial and endothelial cells of different sources. Similarly, the in vivo studies were illustrated in several animal species such as rats, mice, zebrafish, planktons and, guinea pigs, at various concentrations, route of administrations and exposure of nanoparticles. In addition, we have described the unique properties and commercial usage, as well as the similarities and differences among the nanoparticles. The aim of the current review is not only to signify the importance of studying the toxicity of 0-D and 1-D carbon nanomaterials, but also to emphasize the perspectives, future challenges and possible directions in the field.

SELECTION OF CITATIONS
SEARCH DETAIL
...