Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Elife ; 132024 May 30.
Article in English | MEDLINE | ID: mdl-38813866

ABSTRACT

A new protocol can customize the flavor of lab-grown meat by controlling the level of fat deposited between muscle cells.


Subject(s)
Meat , Taste , Taste/physiology , Animals , In Vitro Meat
2.
Exp Hematol ; 127: 14-27, 2023 11.
Article in English | MEDLINE | ID: mdl-37611730

ABSTRACT

For the past decade, significant advances have been achieved in human hematopoietic stem cell (HSC) transplantation for treating various blood diseases and cancers. However, challenges remain with the quality control, amount, and cost of HSCs and HSC-derived immune cells. The advent of human pluripotent stem cells (hPSCs) may transform HSC transplantation and cancer immunotherapy by providing a cost-effective and scalable cell source for fundamental studies and translational applications. In this review, we discuss the current developments in the field of stem cell engineering for hematopoietic stem and progenitor cell (HSPC) differentiation and further differentiation of HSPCs into functional immune cells. The key advances in stem cell engineering include the generation of HSPCs from hPSCs, genetic modification of hPSCs, and hPSC-derived HSPCs for improved function, further differentiation of HPSCs into functional immune cells, and applications of cell culture platforms for hematopoietic cell manufacturing. Current challenges impeding the translation of hPSC-HSPCs and immune cells as well as further directions to address these challenges are also discussed.


Subject(s)
Hematopoietic Stem Cell Transplantation , Pluripotent Stem Cells , Humans , Hematopoietic Stem Cells , Cell Differentiation/genetics , Cell Culture Techniques
3.
ACS Synth Biol ; 12(8): 2262-2270, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37523468

ABSTRACT

Neutrophils, a key innate immune component, are powerful effector leukocytes for mediating opposing effects on tumor progression and ameliorating pathogen infections. However, their short lifespan and complex purification process have limited neutrophil clinical applications. Here we combined genetic engineering technology with a nanodrug system to construct artificial neutrophils that display functions similar to those of native neutrophils. K562 and HL60 human leukemia cells were engineered to express the human G protein-coupled receptor hM4Di. Compared to the parental cells, engineered hM4Di-K562 and hM4Di-HL60 cells exhibited excellent chemotaxis ability towards clozapine-N-oxide (CNO) and superior bacteria phagocytic behavior, resembling native neutrophils. The antibacterial ability of the hM4Di-K562 cells was further enhanced by loading them with the glycopeptide vancomycin via mesoporous silica nanoparticles (Nano@Van). Our proposed artificial cell engineering platform provides a new avenue to investigate the physiological properties of neutrophils.


Subject(s)
Nanoparticles , Neutrophils , Humans , Neutrophils/chemistry , Receptors, G-Protein-Coupled , Genetic Engineering
4.
Bioact Mater ; 27: 168-180, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37091063

ABSTRACT

Adoptive chimeric antigen receptor (CAR)-engineered natural killer (NK) cells have shown promise in treating various cancers. However, limited immunological memory and access to sufficient numbers of allogenic donor cells have hindered their broader preclinical and clinical applications. Here, we first assess eight different CAR constructs that use an anti-PD-L1 nanobody and/or universal anti-fluorescein (FITC) single-chain variable fragment (scFv) to enhance antigen-specific proliferation and anti-tumor cytotoxicity of NK-92 cells against heterogenous solid tumors. We next genetically engineer human pluripotent stem cells (hPSCs) with optimized CARs and differentiate them into functional dual CAR-NK cells. The tumor microenvironment responsive anti-PD-L1 CAR effectively promoted hPSC-NK cell proliferation and cytotoxicity through antigen-dependent activation of phosphorylated STAT3 (pSTAT3) and pSTAT5 signaling pathways via an intracellular truncated IL-2 receptor ß-chain (ΔIL-2Rß) and STAT3-binding tyrosine-X-X-glutamine (YXXQ) motif. Anti-tumor activities of PD-L1-induced memory-like hPSC-NK cells were further boosted by administering a FITC-folate bi-specific adapter that bridges between a programmable anti-FITC CAR and folate receptor alpha-expressing breast tumor cells. Collectively, our hPSC CAR-NK engineering platform is modular and could constitute a realistic strategy to manufacture off-the-shelf CAR-NK cells with immunological memory-like phenotype for targeted immunotherapy.

5.
Nat Commun ; 14(1): 2266, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37080958

ABSTRACT

Glioblastoma (GBM) is one of the most aggressive and lethal solid tumors in human. While efficacious therapeutics, such as emerging chimeric antigen receptor (CAR)-T cells and chemotherapeutics, have been developed to treat various cancers, their effectiveness in GBM treatment has been hindered largely by the blood-brain barrier and blood-brain-tumor barriers. Human neutrophils effectively cross physiological barriers and display effector immunity against pathogens but the short lifespan and resistance to genome editing of primary neutrophils have limited their broad application in immunotherapy. Here we genetically engineer human pluripotent stem cells with CRISPR/Cas9-mediated gene knock-in to express various anti-GBM CAR constructs with T-specific CD3ζ or neutrophil-specific γ-signaling domains. CAR-neutrophils with the best anti-tumor activity are produced to specifically and noninvasively deliver and release tumor microenvironment-responsive nanodrugs to target GBM without the need to induce additional inflammation at the tumor sites. This combinatory chemo-immunotherapy exhibits superior and specific anti-GBM activities, reduces off-target drug delivery and prolongs lifespan in female tumor-bearing mice. Together, this biomimetic CAR-neutrophil drug delivery system is a safe, potent and versatile platform for treating GBM and possibly other devastating diseases.


Subject(s)
Brain Neoplasms , Glioblastoma , Nanoparticles , Mice , Female , Humans , Animals , Glioblastoma/drug therapy , Glioblastoma/genetics , Immunotherapy, Adoptive , Neutrophils , T-Lymphocytes , Tumor Microenvironment , Brain Neoplasms/drug therapy , Immunotherapy , Nanoparticles/therapeutic use
6.
Cells Tissues Organs ; 212(5): 439-467, 2023.
Article in English | MEDLINE | ID: mdl-36599319

ABSTRACT

The past decade has witnessed significant advances in cancer immunotherapy, particularly through the adoptive transfer of engineered T cells in treating advanced leukemias and lymphomas. Despite these excitements, challenges remain with scale, cost, and ensuring quality control of engineered immune cells, including chimeric antigen receptor T, natural killer cells, and macrophages. The advent of human pluripotent stem cells (hPSCs), including human embryonic stem cells and induced pluripotent stem cells, has transformed immunotherapy by providing a scalable, off-the-shelf source of any desired immune cells for basic research, translational studies, and clinical interventions. The tractability of hPSCs for gene editing could also generate homogenous, universal cellular products with custom functionality for individual or combinatory therapeutic applications. This review will explore various immune cell types whose directed differentiation from hPSCs has been achieved and recently adapted for translational immunotherapy and feature forward-looking bioengineering techniques shaping the future of the stem cell field.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Pluripotent Stem Cells , Humans , Immunotherapy, Adoptive/methods , Killer Cells, Natural/metabolism , T-Lymphocytes/metabolism , Immunotherapy , Neoplasms/metabolism
7.
STAR Protoc ; 4(1): 101953, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36527716

ABSTRACT

Here, we present a protocol to efficiently direct human pluripotent stem cells (hPSCs) into hematopoietic stem and progenitor cells (HSPCs) under a chemically defined, albumin-free system. We describe the induction of aorta-gonad-mesonephros-like hematopoiesis from hPSCs into SOX17+ hemogenic endothelium and then into CD34+CD45+ HSPCs via application of Wnt activator and TGFß inhibitor, respectively. The generated HSPCs, characterized by flow cytometry and colony-forming unit assay, express definitive hematopoiesis markers and exhibit multilineage differentiation potential and the capacity to expand. For complete details on the use and execution of this protocol, please refer to Chang et al. (2022a, 2022b).1,2.


Subject(s)
Hematopoietic Stem Cells , Pluripotent Stem Cells , Humans , Cell Differentiation , Hematopoiesis , Antigens, CD34
8.
Front Bioeng Biotechnol ; 10: 1059243, 2022.
Article in English | MEDLINE | ID: mdl-36466327

ABSTRACT

Heart diseases are leading cause of death around the world. Given their unique capacity to self-renew and differentiate into all types of somatic cells, human pluripotent stem cells (hPSCs) hold great promise for heart disease modeling and cardiotoxic drug screening. hPSC-derived cardiac organoids are emerging biomimetic models for studying heart development and cardiovascular diseases, but it remains challenging to make mature organoids with a native-like structure in vitro. In this study, temporal modulation of Wnt signaling pathway co-differentiated hPSCs into beating cardiomyocytes and cardiac endothelial-like cells in 3D organoids, resulting in cardiac endothelial-bounded chamber formation. These chambered cardiac organoids exhibited more mature membrane potential compared to cardiac organoids composed of only cardiomyocytes. Furthermore, a better response to toxic drugs was observed in chamber-contained cardiac organoids. In summary, spatiotemporal signaling pathway modulation may lead to more mature cardiac organoids for studying cardiovascular development and diseases.

9.
Cell Rep ; 40(3): 111128, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858579

ABSTRACT

Neutrophils, the most abundant white blood cells in circulation, are closely related to cancer development and progression. Healthy primary neutrophils present potent cytotoxicity against various cancer cell lines through direct contact and via generation of reactive oxygen species. However, due to their short half-life and resistance to genetic modification, neutrophils have not yet been engineered with chimeric antigen receptors (CARs) to enhance their antitumor cytotoxicity for targeted immunotherapy. Here, we genetically engineered human pluripotent stem cells with synthetic CARs and differentiated them into functional neutrophils by implementing a chemically defined platform. The resulting CAR neutrophils present superior and specific cytotoxicity against tumor cells both in vitro and in vivo. Collectively, we established a robust platform for massive production of CAR neutrophils, paving the way to myeloid cell-based therapeutic strategies that would boost current cancer-treatment approaches.


Subject(s)
Neoplasms , Pluripotent Stem Cells , Receptors, Chimeric Antigen , Humans , Immunotherapy , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Neutrophils/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Chimeric Antigen/metabolism
10.
ACS Synth Biol ; 11(6): 2001-2008, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35608547

ABSTRACT

Natural killer (NK) cells are one type of innate lymphoid cells, and NK cell-based immunotherapy serves as a potentially curative therapy for cancers. However, the lack of reliable resources for a large amount of NK cells required for clinical infusion has limited the broader application of NK cells in targeted immunotherapy. Substantial effort has thus been made to generate NK-like cells from human pluripotent stem cells (hPSCs), but detailed molecular mechanisms regulating NK cell differentiation remain elusive, preventing us from developing robust strategies for NK cell production. Here, we genetically engineered hPSCs with inducible overexpression of transcription factors NFIL3, ID2, or SPI1 via CRISPR/Cas9-mediated gene knock-in and investigated their temporal roles during NK cell differentiation. Our results demonstrated ID2 overexpression significantly promoted NK cell generation compared with NFIL3 and SPI1 overexpression under a chemically defined, feeder-free culture condition. The resulting ID2 hPSC-derived NK cells exhibited various mature NK-specific markers and displayed effective tumor-killing activities, comparable to NK cells derived from wildtype hPSCs. Our study provides a new platform for efficient NK cell production, serving as a realistic off-the-shelf cell source for targeted cancer immunotherapy.


Subject(s)
Pluripotent Stem Cells , Transcription Factors , Cell Differentiation/genetics , Humans , Immunity, Innate , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Killer Cells, Natural/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism
11.
Annu Rev Chem Biomol Eng ; 13: 255-278, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35320695

ABSTRACT

The emergence of human pluripotent stem cell (hPSC) technology over the past two decades has provided a source of normal and diseased human cells for a wide variety of in vitro and in vivo applications. Notably, hPSC-derived cardiomyocytes (hPSC-CMs) are widely used to model human heart development and disease and are in clinical trials for treating heart disease. The success of hPSC-CMs in these applications requires robust, scalable approaches to manufacture large numbers of safe and potent cells. Although significant advances have been made over the past decade in improving the purity and yield of hPSC-CMs and scaling the differentiation process from 2D to 3D, efforts to induce maturation phenotypes during manufacturing have been slow. Process monitoring and closed-loop manufacturing strategies are just being developed. We discuss recent advances in hPSC-CM manufacturing, including differentiation process development and scaling and downstream processes as well as separation and stabilization.


Subject(s)
Myocytes, Cardiac , Pluripotent Stem Cells , Cell Differentiation , Humans
12.
Adv Healthc Mater ; 10(21): e2100995, 2021 11.
Article in English | MEDLINE | ID: mdl-34459150

ABSTRACT

Spatiotemporally controlled presentation of morphogens and elaborate modulation of signaling pathways elicit pattern formation during development. Though this process is critical for proper organogenesis, unraveling the mechanisms of developmental biology have been restricted by challenges associated with studying human embryos. Human pluripotent stem cells (hPSCs) have been used to model development in vitro, however difficulties in precise spatiotemporal control of the cellular microenvironment have limited the utility of this model in exploring mechanisms of pattern formation. Here, a simple and versatile method is presented to spatially pattern hPSC differentiation in 2-dimensional culture via localized morphogen adsorption on substrates. Morphogens including bone morphogenetic protein 4 (BMP4), activin A, and WNT3a are patterned to induce localized mesendoderm, endoderm, cardiomyocyte (CM), and epicardial cell (EpiC) differentiation from hPSCs and hPSC-derived progenitors. Patterned CM and EpiC co-differentiation allows investigation of intercellular interactions in a spatially controlled manner and demonstrate improved alignment of CMs in proximity to EpiCs. This approach provides a platform for the controlled and systematic study of early pattern formation. Moreover, this study provides a facile approach to generate 2D patterned hPSC-derived tissue structures for modeling disease and drug interactions.


Subject(s)
Pluripotent Stem Cells , Cell Differentiation , Cellular Microenvironment , Humans , Myocytes, Cardiac , Tissue Engineering
13.
Biotechnol J ; 14(8): e1800725, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30927511

ABSTRACT

Cardiomyocytes (CMs) generated from human pluripotent stem cells (hPSCs) are immature in their structure and function, limiting their potential in disease modeling, drug screening, and cardiac cellular therapies. Prior studies have demonstrated that coculture of hPSC-derived CMs with other cardiac cell types, including endothelial cells (ECs), can accelerate CM maturation. To address whether the CM differentiation stage at which ECs are introduced affects CM maturation, the authors coculture hPSC-derived ECs with hPSC-derived cardiac progenitor cells (CPCs) and CMs and analyze the molecular and functional attributes of maturation. ECs have a more significant effect on acceleration of maturation when cocultured with CPCs than with CMs. EC coculture with CPCs increases CM size, expression of sarcomere, and ion channel genes and proteins, the presence of intracellular membranous extensions, and chronotropic response compared to monoculture. Maturation is accelerated with an increasing EC:CPC ratio. This study demonstrates that EC incorporation at the CPC stage of CM differentiation expedites CM maturation, leading to cells that may be better suited for in vitro and in vivo applications of hPSC-derived CMs.


Subject(s)
Coculture Techniques/methods , Endothelial Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/cytology , Adrenergic beta-Agonists/pharmacology , Cell Differentiation , Cell Size , Endothelial Cells/drug effects , Gene Expression Regulation , Humans , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Potassium Channels, Inwardly Rectifying/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Troponin C/metabolism , Troponin I/metabolism
14.
Biotechniques ; 65(5): 289-292, 2018 11.
Article in English | MEDLINE | ID: mdl-30394130

ABSTRACT

Cell culture is a vital component of laboratories throughout the scientific community, yet the absence of standardized protocols and documentation practice challenges laboratory efficiency and scientific reproducibility. We examined the effectiveness of a cloud-based software application, CultureTrax® as a tool for standardizing and transferring a complex cell culture protocol. The software workflow and template were used to electronically format a cardiomyocyte differentiation protocol and share a digitally executable copy with a different lab user. While the protocol was unfamiliar to the recipient, they executed the experiment by solely using CultureTrax and successfully derived cardiomyocytes from human induced pluripotent stem cells. This software tool significantly reduced the time and resources required to effectively transfer and implement a novel protocol.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Software , Cell Differentiation , Humans , Reproducibility of Results , Workflow
15.
Theranostics ; 7(7): 2078-2091, 2017.
Article in English | MEDLINE | ID: mdl-28656061

ABSTRACT

Cardiovascular disease is a leading cause of death worldwide. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) hold immense clinical potential and recent studies have enabled generation of virtually pure hPSC-CMs with high efficiency in chemically defined and xeno-free conditions. Despite these advances, hPSC-CMs exhibit an immature phenotype and are arrhythmogenic in vivo, necessitating development of strategies to mature these cells. hPSC-CMs undergo significant metabolic alterations during differentiation and maturation. A detailed analysis of the metabolic changes accompanying maturation of hPSC-CMs may prove useful in identifying new strategies to expedite hPSC-CM maturation and also may provide biomarkers for testing or validating hPSC-CM maturation. In this study we identified global metabolic changes which take place during long-term culture and maturation of hPSC-CMs derived from three different hPSC lines. We have identified several metabolic pathways, including phospholipid metabolism and pantothenate and Coenzyme A metabolism, which showed significant enrichment upon maturation in addition to fatty acid oxidation and metabolism. We also identified increases in glycerophosphocholine and the glycerophosphocholine:phosphocholine ratio as potential metabolic biomarkers of maturation. These biomarkers were also affected in a similar manner during murine heart development in vivo. These results support that hPSC-CM maturation is associated with extensive metabolic changes in metabolic network utilization and understanding the roles of these metabolic changes has the potential to develop novel approaches to monitor and expedite hPSC-CM maturation.


Subject(s)
Biological Factors/analysis , Cell Differentiation , Metabolomics , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , Animals , Cells, Cultured , Humans , Mice
16.
ACS Appl Mater Interfaces ; 8(34): 22721-31, 2016 Aug 31.
Article in English | MEDLINE | ID: mdl-27513165

ABSTRACT

Guiding newly generated tissues in a gradient pattern, thereby precisely mimicking inherent tissue morphology and subsequently arranging the intimate networks between adjacent tissues, is essential to raise the technical levels of tissue engineering and facilitate its transition into the clinic. In this study, a straightforward electrospinning method (the tubing-electrospinning technique) was developed to create fibrous matrices readily with diverse gradient patterns and to induce patterned cellular responses. Gradient fibrous matrices can be produced simply by installing a series of polymer-containing lengths of tubing into an electrospinning circuit and sequentially processing polymers without a time lag. The loading of polymer samples with different characteristics, including concentration, wettability, and mechanical properties, into the tubing system enabled unique features in fibrous matrices, such as longitudinal gradients in fiber density, surface properties, and mechanical stiffness. The resulting fibrous gradients were shown to arrange cellular migration and residence in a gradient manner, thereby offering efficient cues to mediate patterned tissue formation. The one-step process using tubing-electrospinning apparatus can be used without significant modifications regardless of the type of fibrous gradient. Hence, the tubing-electrospinning system can serve as a platform that can be readily used by a wide-range of users to induce patterned tissue formation in a gradient manner, which will ultimately improve the functionality of tissue engineering scaffolds.

17.
ACS Appl Mater Interfaces ; 8(21): 13320-31, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-27167566

ABSTRACT

Implementing patterned fibrous matrices can offer a highly valuable platform for spatially orchestrating hierarchical cellular constructs, specifically for neural engineering approaches, in which striated alignment or directional growth of axons are key elements for the functional recovery of damaged nervous systems. Thus, understanding the structural parameters of patterned fibrous matrices that can effectively promote neural growth can provide crucial clues for designing state-of-the-art tissue engineering scaffolds. To this end, salt-induced electrospun patterned fiber bundles (SiEP bundles) comprising longitudinally stacked multiple fibers were fabricated, and their capabilities of spatially stimulating the responses of neural cells, including PC12 cells, human neural stem cells (hNSCs), and dorsal root ganglia (DRG), were assessed by comparing them to conventional fibrous matrices having either randomly oriented fibers or individually aligned fibers. The SiEP bundles possessed remarkably distinctive morphological and topographical characteristics: multicomplexed infrastructures with nano- and microscale fibers, rough surfaces, and soft mechanical properties. Importantly, the SiEP bundles resulted in spatial cellular elongations corresponding to the fiber directions and induced highly robust neurite extensions along the patterned fibers. Furthermore, the residence of hNSCs on the topographically rough grooves of the SiEP bundles boosted neuronal differentiation. These findings can provide crucial insights for designing fibrous platforms that can spatially regulate cellular responses and potentially offer powerful strategies for a neural growth system in which directional cellular responses are critical for the functional recovery of damaged neural tissues.


Subject(s)
Salts/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Axons/physiology , Humans , Nerve Tissue/cytology , Neurons/cytology , PC12 Cells , Rats
18.
Angew Chem Int Ed Engl ; 54(26): 7587-91, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-25958994

ABSTRACT

Advanced technologies that can mimic hierarchical architectures found in nature can provide pivotal clues for elucidating numerous biological mechanisms. Herein, a novel technology, spontaneous organization of numerous-layer generation by electrospray (SpONGE), was developed to create self-assembled and multilayered fibrous structures. The simple inclusion of salts in a polymer solution prior to electrospraying was key to mediating the structural versatilities of the fibrous structures. The SpONGE matrix demonstrated great potential as a crucial building block capable of inducing sequential, localized drug delivery or orchestrating cellular distribution in vivo, thereby expanding its scope of use to cover a variety of biomedical applications.

19.
Biomed Microdevices ; 16(6): 793-804, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24972552

ABSTRACT

Electrospun fibrous mats have emerged as powerful tissue engineering scaffolds capable of providing highly effective and versatile physical guidance, mimicking the extracellular environment. However, electrospinning typically produces a sheet-like structure, which is a major limitation associated with current electrospinning technologies. To address this challenge, highly porous, volumetric hydrogel-hybrid fibrous scaffolds were fabricated by one Taylor cone-based side-by-side dual electrospinning of poly (ε-caprolactone) (PCL) and poly (vinyl pyrrolidone) (PVP), which possess distinct properties (i.e., hydrophobic and hydrogel properties, respectively). Immersion of the resulting scaffolds in water induced spatial tortuosity of the hydrogel PVP fibers while maintaining their aligned fibrous structures in parallel with the PCL fibers. The resulting conformational changes in the entire bicomponent fibers upon immersion in water led to volumetric expansion of the fibrous scaffolds. The spatial fiber tortuosity significantly increased the pore volumes of electrospun fibrous mats and dramatically promoted cellular infiltration into the scaffold interior both in vitro and in vivo. Harmonizing the flexible PCL fibers with the soft PVP-hydrogel layers produced highly ductile fibrous structures that could mechanically resist cellular contractile forces upon in vivo implantation. This facile dual electrospinning followed by the spatial fiber tortuosity for fabricating three-dimensional hydrogel-hybrid fibrous scaffolds will extend the use of electrospun fibers toward various tissue engineering applications.


Subject(s)
Biocompatible Materials/chemistry , Hydrogels/chemistry , Nanofibers/chemistry , Polyesters/chemistry , Povidone/chemistry , Tissue Scaffolds/chemistry , Animals , Mice , NIH 3T3 Cells , Porosity , Tissue Engineering
20.
ACS Appl Mater Interfaces ; 6(11): 8288-94, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24827581

ABSTRACT

Controlled release of biosuprastructures, such as viruses, from surfaces has been a challenging task in providing efficient ex vivo gene delivery. Conventional controlled viral release approaches have demonstrated low viral immobilization and burst release, inhibiting delivery efficiency. Here, a highly powerful substrate-mediated viral delivery system was designed by combining two key components that have demonstrated great potential in the fields of gene therapy and surface chemistry, respectively: adeno-associated viral (AAV) vectors and adhesive catecholamine surfaces. The introduction of a nanoscale thin coating of catecholamines, poly(norepinephrine) (pNE) or poly(dopamine) (pDA) to provide AAV adhesion followed by human neural stem cell (hNSC) culture on sticky solid surfaces exhibited unprecedented results: approximately 90% loading vs 25% (AAV_bare surface), no burst release, sustained release at constant rates, approximately 70% infection vs 20% (AAV_bare surface), and rapid internalization. Importantly, the sticky catecholamine-mediated AAV delivery system successfully induced a physiological response from hNSCs, cellular proliferation by a single-shot of AAV encoding fibroblast growth factor-2 (FGF-2), which is typically achieved by multiple treatments with expensive FGF-2 proteins. By combining the adhesive material-independent surface functionalization characters of pNE and pDA, this new sticky "delivering-from" gene delivery platform will make a significant contribution to numerous fields, including tissue engineering, gene therapy, and stem cell therapy.


Subject(s)
Dependovirus/genetics , Dopamine/administration & dosage , Gene Transfer Techniques , Neural Stem Cells/virology , Norepinephrine/administration & dosage , Cell Line , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...