Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
J Drug Target ; 31(3): 310-319, 2023 03.
Article in English | MEDLINE | ID: mdl-36440540

ABSTRACT

BACKGROUND: The drug delivery for treatment of glioblastoma multiforme (GBM) has been unsatisfactory mainly due to the drug resistance and low targeting efficiency. The selective targeting of GBM cells and using a cocktail of therapeutic agents to synergistically induce apoptosis may help enhance the drug delivery. METHODS: In this study, mesenchymal stem cells (MSCs) were engineered to produce exosomes, i.e. nanosized natural vesicles presenting anti-EGFRvIII (ab139) antibody on their surface while encapsulating two apoptosis-inducing gene therapy agents, i.e. cytosine deaminase (CDA) and miR-34a. Exosomes were characterised for their size, morphology, protein content and markers using dynamic light scattering and nanoparticle tracking analysis, cryo-TEM, Western blotting, respectively. miR-34a overexpression and Lamp2-ab139 protein expression were analysed using real-time PCR and flow cytometry, respectively. The armed exosomes were delivered to EGFRvIII positive GBM cells (U87EGFRvIII) as well as wild type cell line (U87), which was EGFRvIII negative. Apoptosis was quantified using flow cytometry in both EGFRvIII negative and positive U87 cells, receiving one gene therapy agent (either CDA or miR-34a) or a combination of them (CDAmiR). RESULTS: Spherical shape exosomes with an average diameter of 110 nm and a membrane thickness of 6.5 nm were isolated from MSCs. Lamp2-ab139 was successfully expressed on the surface of transfected cells and their secreted exosomes. Induced apoptosis rates was significantly higher in U87EGFRvIII cells than for U87 cells, indicating selectivity. The cell death rate was 6%, 9% and 12% in U87, 13%, 21% and 40% in U87EGFRvIII cells for CDA, miR-34a and CDAmiR treatment respectively, showing a higher apoptosis rate in the cells receiving both drugs compared to when single therapy was applied. CONCLUSION: The experimental findings clearly show the improved apoptosis rate of GBM cells when treated by engineered exosomes armed with two gene therapy agents and targeted towards EGFRvIII antigen.


Subject(s)
Exosomes , Glioblastoma , MicroRNAs , Humans , Glioblastoma/drug therapy , Exosomes/metabolism , Cell Line, Tumor , Apoptosis , MicroRNAs/genetics , MicroRNAs/metabolism
2.
Mol Pharm ; 20(1): 545-560, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36484477

ABSTRACT

Clinical treatment of glioblastoma (GBM) remains a major challenge because of the blood-brain barrier, chemotherapeutic resistance, and aggressive tumor metastasis. The development of advanced nanoplatforms that can efficiently deliver drugs and gene therapies across the BBB to the brain tumors is urgently needed. The protein "downregulated in renal cell carcinoma" (DRR) is one of the key drivers of GBM invasion. Here, we engineered porous silicon nanoparticles (pSiNPs) with antisense oligonucleotide (AON) for DRR gene knockdown as a targeted gene and drug delivery platform for GBM treatment. These AON-modified pSiNPs (AON@pSiNPs) were selectively internalized by GBM and human cerebral microvascular endothelial cells (hCMEC/D3) cells expressing Class A scavenger receptors (SR-A). AON was released from AON@pSiNPs, knocked down DRR and inhibited GBM cell migration. Additionally, a penetration study in a microfluidic-based BBB model and a biodistribution study in a glioma mice model showed that AON@pSiNPs could specifically cross the BBB and enter the brain. We further demonstrated that AON@pSiNPs could carry a large payload of the chemotherapy drug temozolomide (TMZ, 1.3 mg of TMZ per mg of NPs) and induce a significant cytotoxicity in GBM cells. On the basis of these results, the nanocarrier and its multifunctional strategy provide a strong potential for clinical treatment of GBM and research for targeted drug and gene delivery.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Mice , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Silicon , Porosity , Endothelial Cells , Tissue Distribution , Cell Line, Tumor , Temozolomide/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Drug Resistance, Neoplasm/genetics
3.
Adv Sci (Weinh) ; 9(26): e2201740, 2022 09.
Article in English | MEDLINE | ID: mdl-35851766

ABSTRACT

Central Nervous System (CNS) diseases, such as Alzheimer's diseases (AD), Parkinson's Diseases (PD), brain tumors, Huntington's disease (HD), and stroke, still remain difficult to treat by the conventional molecular drugs. In recent years, various gene therapies have come into the spotlight as versatile therapeutics providing the potential to prevent and treat these diseases. Despite the significant progress that has undoubtedly been achieved in terms of the design and modification of genetic modulators with desired potency and minimized unwanted immune responses, the efficient and safe in vivo delivery of gene therapies still poses major translational challenges. Various non-viral nanomedicines have been recently explored to circumvent this limitation. In this review, an overview of gene therapies for CNS diseases is provided and describes recent advances in the development of nanomedicines, including their unique characteristics, chemical modifications, bioconjugations, and the specific applications that those nanomedicines are harnessed to deliver gene therapies.


Subject(s)
Central Nervous System Diseases , Nanomedicine , Blood-Brain Barrier , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/therapy , Drug Delivery Systems , Genetic Therapy , Humans
4.
J Nanobiotechnology ; 19(1): 60, 2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33637089

ABSTRACT

BACKGROUND: Approximately 80% of brain tumours are gliomas. Despite treatment, patient mortality remains high due to local metastasis and relapse. It has been shown that transferrin-functionalised porous silicon nanoparticles (Tf@pSiNPs) can inhibit the migration of U87 glioma cells. However, the underlying mechanisms and the effect of glioma cell heterogeneity, which is a hallmark of the disease, on the efficacy of Tf@pSiNPs remains to be addressed. RESULTS: Here, we observed that Tf@pSiNPs inhibited heterogeneous patient-derived glioma cells' (WK1) migration across small perforations (3 µm) by approximately 30%. A phenotypical characterisation of the migrated subpopulations revealed that the majority of them were nestin and fibroblast growth factor receptor 1 positive, an indication of their cancer stem cell origin. The treatment did not inhibit cell migration across large perforations (8 µm), nor cytoskeleton formation. This is in agreement with our previous observations that cellular-volume regulation is a mediator of Tf@pSiNPs' cell migration inhibition. Since aquaporin 9 (AQP9) is closely linked to cellular-volume regulation, and is highly expressed in glioma, the effect of AQP9 expression on WK1 migration was investigated. We showed that WK1 migration is correlated to the differential expression patterns of AQP9. However, AQP9-silencing did not affect WK1 cell migration across perforations, nor the efficacy of cell migration inhibition mediated by Tf@pSiNPs, suggesting that AQP9 is not a mediator of the inhibition. CONCLUSION: This in vitro investigation highlights the unique therapeutic potentials of Tf@pSiNPs against glioma cell migration and indicates further optimisations that are required to maximise its therapeutic efficacies.


Subject(s)
Glioma/drug therapy , Nanoparticles/therapeutic use , Porosity , Silicon/pharmacology , Aquaporins/genetics , Brain Neoplasms , Cell Line, Tumor , Cell Movement , Glioblastoma/drug therapy , Humans , Neoplastic Stem Cells , Receptor, Fibroblast Growth Factor, Type 1
5.
ACS Appl Mater Interfaces ; 12(51): 56753-56766, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33226228

ABSTRACT

Here, we have developed and evaluated a microfluidic-based human blood-brain-barrier (µBBB) platform that models and predicts brain tissue uptake of small molecule drugs and nanoparticles (NPs) targeting the central nervous system. By using a photocrosslinkable copolymer that was prepared from monomers containing benzophenone and N-hydroxysuccinimide ester functional groups, we were able to evenly coat and functionalize µBBB chip channels in situ, providing a covalently attached homogenous layer of extracellular matrix proteins. This novel approach allowed the coculture of human endothelial cells, pericytes, and astrocytes and resulted in the formation of a mimic of cerebral endothelium expressing tight junction markers and efflux proteins, resembling the native BBB. The permeability coefficients of a number of compounds, including caffeine, nitrofurantoin, dextran, sucrose, glucose, and alanine, were measured on our µBBB platform and were found to agree with reported values. In addition, we successfully visualized the receptor-mediated uptake and transcytosis of transferrin-functionalized NPs. The BBB-penetrating NPs were able to target glioma cells cultured in 3D in the brain compartment of our µBBB. In conclusion, our µBBB was able to accurately predict the BBB permeability of both small molecule pharmaceuticals and nanovectors and allowed time-resolved visualization of transcytosis. Our versatile chip design accommodates different brain disease models and is expected to be exploited in further BBB studies, aiming at replacing animal experiments.


Subject(s)
Artificial Organs , Blood-Brain Barrier/metabolism , Lab-On-A-Chip Devices , Nanoparticles/chemistry , Organic Chemicals/analysis , Astrocytes/metabolism , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Humans , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Pericytes/metabolism , Transferrin/chemistry
6.
Tissue Eng Part B Rev ; 26(6): 540-554, 2020 12.
Article in English | MEDLINE | ID: mdl-32242476

ABSTRACT

One of the most crucial components of regenerative medicine is the controlled differentiation of embryonic or adult stem cells into the desired cell lineage. Although most of the reported protocols of stem cell differentiation involve the use of soluble growth factors, it is increasingly evident that stem cells also undergo differentiation when cultured in the appropriate microenvironment. When cultured in decellularized tissues, for instance, stem cells can recapitulate the morphogenesis and functional specialization of differentiated cell types with speed and efficiency that often surpass the traditional growth factor-driven protocols. This suggests that the tissue microenvironment (TME) provides stem cells with a holistic "instructive niche" that harbors signals for cellular reprogramming. The translation of this into medical applications requires the decoding of these signals, but this has been hampered by the complexity of TME. This problem is often addressed by a reductionist approach, in which cells are exposed to substrates decorated with simple, empirically designed geometries, textures, and chemical compositions ("bottom-up" approach). Although these studies are invaluable in revealing the basic principles of mechanotransduction mechanisms, their physiological relevance is often uncertain. This review examines the recent progress of an alternative, "top-down" approach, in which the TME is treated as a holistic biological entity. This approach is made possible by recent advances in systems biology and fabrication technologies that enable the isolation, characterization, and reconstitution of TME. It is hoped that these new techniques will elucidate the nature of niche signals so that they can be extracted, replicated, and controlled. This review summarizes these emerging techniques and how the data they generated are changing our view on TME. Impact statement This review summarizes the current state of art of the understanding of instructive niche in the field of tissue microenvironment. Not only did we survey the use of different biochemical preparations as stimuli of stem cell differentiation and summarize the recent effort in dissecting the biochemical composition of these preparations, through the application of extracellular matrix (ECM) arrays and proteomics, but we also introduce the use of open-source, high-content immunohistochemistry projects in contributing to the understanding of tissue-specific composition of ECM. We believe this review would be highly useful for our peer researching in the same field. "Mr. Tulkinghorn is always the same… so oddly out of place and yet so perfectly at home." -Charles Dickens, Bleak House.


Subject(s)
Mechanotransduction, Cellular , Tissue Engineering , Cell Differentiation , Cell Lineage , Extracellular Matrix , Stem Cells
7.
Sci Rep ; 10(1): 2320, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32047170

ABSTRACT

Mortality of glioblastoma multiforme (GBM) has not improved over the last two decades despite medical breakthroughs in the treatment of other types of cancers. Nanoparticles hold tremendous promise to overcome the pharmacokinetic challenges and off-target adverse effects. However, an inhibitory effect of nanoparticles by themselves on metastasis has not been explored. In this study, we developed transferrin-conjugated porous silicon nanoparticles (Tf@pSiNP) and studied their effect on inhibiting GBM migration by means of a microfluidic-based migration chip. This platform, designed to mimic the tight extracellular migration tracts in brain parenchyma, allowed high-content time-resolved imaging of cell migration. Tf@pSiNP were colloidally stable, biocompatible, and their uptake into GBM cells was enhanced by receptor-mediated internalisation. The migration of Tf@pSiNP-exposed cells across the confined microchannels was suppressed, but unconfined migration was unaffected. The pSiNP-induced destabilisation of focal adhesions at the leading front may partially explain the migration inhibition. More corroborating evidence suggests that pSiNP uptake reduced the plasticity of GBM cells in reducing cell volume, an effect that proved crucial in facilitating migration across the tight confined tracts. We believe that the inhibitory effect of Tf@pSiNP on cell migration, together with the drug-delivery capability of pSiNP, could potentially offer a disruptive strategy to treat GBM.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems , Extracellular Space/drug effects , Glioblastoma/drug therapy , Nanoparticles/administration & dosage , Silicon/chemistry , Transferrin/administration & dosage , Apoptosis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Nanoparticles/chemistry , Porosity , Transferrin/chemistry , Tumor Cells, Cultured
8.
Bioconjug Chem ; 30(10): 2539-2543, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31560523

ABSTRACT

The functionalization of proteins with different cargo molecules is highly desirable for a broad range of applications. However, the reproducible production of defined conjugates with multiple functionalities is a significant challenge. Herein, we report the dual site-specific labeling of an antibody fragment, utilizing the orthogonal Sortase A and π-clamp conjugation methods, and demonstrate that binding of the antibody fragment to its target receptor is retained after dual labeling.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/metabolism , Binding Sites , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Ligands , Staining and Labeling
9.
ACS Appl Mater Interfaces ; 11(37): 33637-33649, 2019 Sep 18.
Article in English | MEDLINE | ID: mdl-31433156

ABSTRACT

There is a dire need to develop more effective therapeutics to combat brain cancer such as glioblastoma multiforme (GBM). An ideal treatment is expected to target deliver chemotherapeutics to glioma cells across the blood-brain barrier (BBB). The overexpression of transferrin (Tf) receptor (TfR) on the BBB and the GBM cell surfaces but not on the surrounding cells renders TfR a promising target. While porous silicon nanoparticles (pSiNPs) have been intensely studied as a delivery vehicle due to their high biocompatibility, degradability, and drug-loading capacity, the potential to target deliver drugs with transferrin (Tf)-functionalized pSiNPs remains unaddressed. Here, we developed and systematically evaluated Tf-functionalized pSiNPs (Tf@pSiNPs) as a glioma-targeted drug delivery system. These nanoparticles showed excellent colloidal stability and had a low toxicity profile. As compared with nontargeted pSiNPs, Tf@pSiNPs were selective to BBB-forming cells and GBM cells and were efficiently internalized through clathrin receptor-mediated endocytosis. The anticancer drug doxorubicin (Dox) was effectively loaded (8.8 wt %) and released from Tf@pSiNPs in a pH-responsive manner over 24 h. Furthermore, the results demonstrate that Dox delivered by Tf@pSiNPs induced significantly enhanced cytotoxicity to GBM cells across an in vitro BBB monolayer compared with free Dox. Overall, Tf@pSiNPs offer a potential toolbox for enabling targeted therapy to treat GBM.


Subject(s)
Doxorubicin , Drug Carriers , Glioblastoma/drug therapy , Nanoparticles , Silicon , Transferrin , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Screening Assays, Antitumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Porosity , Silicon/chemistry , Silicon/pharmacokinetics , Silicon/pharmacology , Transferrin/chemistry , Transferrin/pharmacokinetics , Transferrin/pharmacology
10.
Trends Biotechnol ; 37(12): 1295-1314, 2019 12.
Article in English | MEDLINE | ID: mdl-31130308

ABSTRACT

Therapeutic options for neurological disorders currently remain limited. The intrinsic complexity of the brain architecture prevents potential therapeutics from reaching their cerebral target, thus limiting their efficacy. Recent advances in microfluidic technology and organ-on-chip systems have enabled the development of a new generation of in vitro platforms that can recapitulate complex in vivo microenvironments and physiological responses. In this context, microfluidic-based in vitro models of the blood-brain barrier (BBB) are of particular interest as they provide an innovative approach for conducting research related to the brain, including modeling of neurodegenerative diseases and high-throughput drug screening. Here, we present the most recent advances in BBB-on-chip devices and examine validation steps that will strengthen their future applications.


Subject(s)
Brain Diseases , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Models, Cardiovascular , Models, Neurological , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Diseases/metabolism , Brain Diseases/pathology , Humans
11.
ACS Appl Mater Interfaces ; 11(21): 18988-18994, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31051073

ABSTRACT

Thermal scanning probe lithography (t-SPL) is a nanofabrication technique in which an immobilized thermolabile resist, such as polyphthalaldehyde (PPA), is locally vaporized by a heated atomic force microscope tip. Compared with other nanofabrication techniques, such as soft lithography and nanoimprinting lithography, t-SPL is more efficient and convenient as it does not involve time-consuming mask productions or complicated etching procedures, making it a promising candidate technique for the fast prototyping of nanoscale topographies for biological studies. Here, we established the direct use of PPA-coated surfaces as a cell culture substrate. We showed that PPA is biocompatible and that the deposition of allylamine by plasma polymerization on a silicon wafer before PPA coating can stabilize the immobilization of PPA in aqueous solutions. When seeded on PPA-coated surfaces, human mesenchymal stem cells (MSC) adhered, spread, and proliferated in a manner indistinguishable from cells cultured on glass surfaces. This allowed us to subsequently use t-SPL to generate nanotopographies for cell culture experiments. As a proof of concept, we analyzed the surface topography of bovine tendon sections, previously shown to induce morphogenesis and differentiation of MSC, by means of atomic force microscopy, and then "wrote" topographical data on PPA by means of t-SPL. The resulting substrate, matching the native tissue topography on the nanoscale, was directly used for MSC culture. The t-SPL substrate induced similar changes in cell morphology and focal adhesion formation in the MSC compared to native tendon sections, suggesting that t-SPL can rapidly generate cell culture substrates with complex and spatially accurate topographical signals. This technique may greatly accelerate the prototyping of models for the study of cell-matrix interactions.


Subject(s)
Printing , Tissue Engineering/methods , Allylamine/chemistry , Biocompatible Materials/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Plasma Gases/pharmacology , Polymerization , Tendons/drug effects , Tendons/physiology
12.
J Nanobiotechnology ; 16(1): 38, 2018 Apr 13.
Article in English | MEDLINE | ID: mdl-29653579

ABSTRACT

BACKGROUND: Multidrug resistance-associated protein 1 (MRP1) overexpression plays a major role in chemoresistance in glioblastoma multiforme (GBM) contributing to its notorious deadly nature. Although MRP1-siRNA transfection to GBM in vitro has been shown to sensitise the cells to drug, MRP1 silencing in vivo and the phenotypic influence on the tumour and normal tissues upon MRP1 down-regulation have not been established. Here, porous silicon nanoparticles (pSiNPs) that enable high-capacity loading and delivery of siRNA are applied in vitro and in vivo. RESULT: We established pSiNPs with polyethyleneimine (PEI) capping that enables high-capacity loading of siRNA (92 µg of siRNA/mg PEI-pSiNPs), and optimised release profile (70% released between 24 and 48 h). These pSiNPs are biocompatible, and demonstrate cellular uptake and effective knockdown of MRP1 expression in GBM by 30%. Also, siRNA delivery was found to significantly reduce GBM proliferation as an associated effect. This effect is likely mediated by the attenuation of MRP1 transmembrane transport, followed by cell cycle arrest. MRP1 silencing in GBM tumour using MRP1-siRNA loaded pSiNPs was demonstrated in mice (82% reduction at the protein level 48 h post-injection), and it also produced antiproliferative effect in GBM by reducing the population of proliferative cells. These results indicate that in vitro observations are translatable in vivo. No histopathological signs of acute damage were observed in other MRP1-expressing organs despite collateral downregulations. CONCLUSIONS: This study proposes the potential of efficient MRP1-siRNA delivery by using PEI-capped pSiNPs in achieving a dual therapeutic role of directly attenuating the growth of GBM while sensitising residual tumour cells to the effects of chemotherapy post-resection.


Subject(s)
Gene Silencing , Glioblastoma/pathology , Multidrug Resistance-Associated Proteins/metabolism , Nanoparticles/chemistry , Polyethyleneimine/chemistry , RNA, Small Interfering/administration & dosage , Silicon/chemistry , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Knockdown Techniques , Humans , Mice, Nude , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/genetics , Nanoparticles/ultrastructure , Organ Specificity , Phenotype , Porosity , Propionates/pharmacology , Quinolines/pharmacology
13.
ACS Appl Mater Interfaces ; 9(49): 42601-42611, 2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29154535

ABSTRACT

Continuing our research efforts in developing mesoporous silicon nanoparticle-based biomaterials for cancer therapy, we employed here porous silicon nanoparticles as a nanocarrier to deliver contrast agents to diseased cells. Nanoconfinement of small molecule Gd-chelates (L1-Gd) enhanced the T1 contrast dramatically compared to distinct Gd-chelate (L1-Gd) by virtue of its slow tumbling rate, increased number of bound water molecules, and their occupancy time. The newly synthesized Gd-chelate (L1-Gd) was covalently grafted on silicon nanostructures and conjugated to an antibody specific for epidermal growth factor receptor (EGFR) via a hydrazone linkage. The salient feature of this nanosized contrast agent is the capability of EGFR targeted delivery to cancer cells. Mesoporous silicon nanoparticles were chosen as the nanocarrier because of their high porosity, high surface area, and excellent biodegradability. This type of nanosized contrast agent also performs well in high magnetic fields.


Subject(s)
Nanoparticles , Contrast Media , ErbB Receptors , Gadolinium , Magnetic Resonance Imaging , Silicon
14.
Biomaterials ; 74: 217-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26466356

ABSTRACT

Advanced biosensors in future medicine hinge on the evolvement of biomaterials. Porous silicon (pSi), a generally biodegradable and biocompatible material that can be fabricated to include environment-responsive optical characteristics, is an excellent candidate for in vivo biosensors. However, the feasibility of using this material as a subcutaneously implanted optical biosensor has never been demonstrated. Here, we investigated the stability and biocompatibility of a thermally-hydrocarbonised (THC) pSi optical rugate filter, and demonstrated its optical functionality in vitro and in vivo. We first compared pSi films with different surface chemistries and observed that the material was cytotoxic despite the outstanding stability of the THC pSi films. We then showed that the cytotoxicity correlates with reactive oxygen species levels, which could be mitigated by pre-incubation of THC pSi (PITHC pSi). PITHC pSi facilitates normal cellular phenotypes and is biocompatible in vivo. Importantly, the material also possesses optical properties capable of responding to microenvironmental changes that are readable non-invasively in cell culture and subcutaneous settings. Collectively, we demonstrate, for the first time, that PITHC pSi rugate filters are both biocompatible and optically functional for lab-on-a-chip and subcutaneous biosensing scenarios. We believe that this study will deepen our understanding of cell-pSi interactions and foster the development of implantable biosensors.


Subject(s)
Biosensing Techniques , Hydrocarbons/chemistry , Silicon/chemistry , Skin , 3T3 Cells , Animals , Mice , Microscopy, Electron, Scanning , Porosity
15.
BMC Cell Biol ; 15: 10, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24661496

ABSTRACT

BACKGROUND: In vitro experiments on the functional roles of extracellular matrix (ECM) components usually involve the culture of cells on surfaces coated with purified ECM components. These experiments can seldom recuperate the spatial arrangement of ECM found in vivo. In this study, we have overcome this obstacle by using histological sections of bovine Achilles tendon as cell culture substrates. RESULTS: We found that tendon sections can be viewed as a pre-formed block of ECM in which the collagen fibrils exhibited a spatial regularity unraveled in any artificially constructed scaffold. By carving the tendon at different angles relative to its main axis, we created different surfaces with distinct spatial arrangements of collagen fibrils. To assess the cellular responses to these surfaces, human mesenchymal stem cells (MSCs) were directly cultured on these sections, hence exposed to the collagen with different spatial orientations. Cells seeded on longitudinal tendon sections adopted a highly elongated and aligned morphology, and expressed an increased level of tenomodulin, suggesting that the collagen fibrils present in this section provide a microenvironment that facilitates cell morphogenesis and differentiation. However, MSC elongation, alignment and induction of tenomodulin diminished dramatically even as the sectioned angle changed slightly. CONCLUSION: Our results suggest that cell functions are influenced not only by the type or concentration of ECM components, but also by the precise spatial arrangements of these molecules. The method developed in this study offers a simple and robust way for the studying of cell-ECM interactions, and opens many research avenues in the field of matrix biology.


Subject(s)
Cell Differentiation , Extracellular Matrix/chemistry , Mesenchymal Stem Cells/cytology , Animals , Cattle , Cell Adhesion , Cell Culture Techniques , Cell Line , Collagen/chemistry , Extracellular Matrix/metabolism , Humans , Hydrogels/chemistry , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Scanning , Tendons/chemistry , Tendons/pathology
16.
Biofabrication ; 6(1): 015011, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24589941

ABSTRACT

Control of cell migration is important in numerous key biological processes, and is implicated in pathological conditions such as cancer metastasis and inflammatory diseases. Many previous studies indicated that cell migration could be guided by micropatterns fabricated on cell culture surfaces. In this study, we designed a polydimethylsiloxane cell culture substrate with gratings punctuated by corners and ends, and studied its effects on the behavior of MC3T3-E1 osteoblast cells. MC3T3-E1 cells elongated and aligned with the gratings, and the migration paths of the cells appeared to be guided by the grating pattern. Interestingly, more than 88% of the cells cultured on these patterns were observed to reverse their migration directions at least once during the 16 h examination period. Most of the reversal events occurred at the corners and the ends of the pattern, suggesting these localized topographical features induce an abrupt loss in directional persistence. Moreover, the cell speed was observed to increase temporarily right after each directional reversal. Focal adhesion complexes were more well-established in cells on the angular gratings than on flat surfaces, but the formation of filipodia appeared to be imbalanced at the corners and the ends, possibly leading to the loss of directional persistence. This study describes the first engineered cell culture surface that consistently induces changes in the directional persistence of adherent cells. This will provide an experimental model for the study of this phenomenon and a valuable platform to control the cell motility and directionality, which can be used for cell screening and selection.


Subject(s)
Cell Movement , Osteoblasts/cytology , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Animals , Cell Adhesion , Cell Line , Humans , Mice , Surface Properties
17.
Mater Sci Eng C Mater Biol Appl ; 33(3): 1380-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23827585

ABSTRACT

Multi-walled carbon nanotubes (MWNTs) of 0.1 and 0.3 wt.% and hydoxyapatite nanorods (nHAs) of 8-20 wt.% were incorporated into polypropylene (PP) to form biocomposites using melt-compounding and injection molding techniques. The structural, mechanical, thermal and in vitro cell responses of the PP/MWNT-nHA hybrids were investigated. Tensile and impact tests demonstrated that the MWNT additions are beneficial in enhancing the stiffness, tensile strength and impact toughness of the PP/nHA nanocomposites. According to thermal analysis, the nHA and MWNT fillers were found to be very effective to improve dimensional and thermal stability of PP. The results of osteoblast cell cultivation and dimethyl thiazolyl diphenyl tetrazolium (MTT) tests showed that the PP/MWNT-nHA nanocomposites are biocompatible. Such novel PP/MWNT-nHA hybrids are considered to be potential biomaterials for making orthopedic bone implants.


Subject(s)
Biocompatible Materials/pharmacology , Bone and Bones/drug effects , Durapatite/pharmacology , Nanotubes, Carbon/chemistry , Nanotubes/chemistry , Polypropylenes/pharmacology , Tissue Engineering , Calorimetry, Differential Scanning , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Crystallization , Elastic Modulus/drug effects , Humans , Nanocomposites/ultrastructure , Nanotubes, Carbon/ultrastructure , Osteoblasts/cytology , Osteoblasts/ultrastructure , Spectroscopy, Fourier Transform Infrared , Temperature , Tensile Strength/drug effects , Thermogravimetry , X-Ray Diffraction
18.
Biomaterials ; 33(31): 7686-98, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22818988

ABSTRACT

Although many studies have demonstrated that cell phenotype is affected by the surface properties of biomaterials, these materials often fail to mimic the complexity of the native tissue microenvironment (TME). In this study, we have developed a new experimental model that allows the characterisation and functional reconstruction of natural TME. We discovered that mesenchymal stem cells (MSC) cultured on cryostat sections of bovine Achilles tendon adopted an elongated and aligned morphology, and expressed tenocyte marker tenomodulin (TNMD). This suggests that tendon sections contain the signalling cues that guide MSCs to commit to the tenogenic lineage. To reconstruct this instructive niche, we prepared PDMS replica by using tendon sections as template. The resulting bioimprint faithfully copied the physical topography and elasticity of the section. This replica, when coated with collagen 1, supported tenogenesis of MSC without requiring exogenous growth factors. This study illustrates how extracellular biophysical and biochemical features intertwines to form a niche that influences the cell fate and demonstrated that such complex information could be conveniently reconstructed with synthetic materials and purified extracellular matrix proteins.


Subject(s)
Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Cellular Microenvironment/drug effects , Mesenchymal Stem Cells/cytology , Molecular Imprinting/methods , Tendons/cytology , Tendons/drug effects , Animals , Cattle , Cell Adhesion/drug effects , Cell Line , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cryoultramicrotomy , Dimethylpolysiloxanes/chemistry , Dogs , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Mice , Microscopy, Confocal , Models, Biological , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL