28 results on '"Chooi WH"'
Search Results
2. Using magnetic resonance relaxometry to evaluate the safety and quality of induced pluripotent stem cell-derived spinal cord progenitor cells.
- Author
-
Tan J, Chen J, Roxby D, Chooi WH, Nguyen TD, Ng SY, Han J, and Chew SY
- Subjects
- Humans, Neural Stem Cells cytology, Neural Stem Cells metabolism, Spinal Cord Injuries therapy, Iron metabolism, Cells, Cultured, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Cell Differentiation, Spinal Cord cytology
- Abstract
Background: The emergence of induced pluripotent stem cells (iPSCs) offers a promising approach for replacing damaged neurons and glial cells, particularly in spinal cord injuries (SCI). Despite its merits, iPSC differentiation into spinal cord progenitor cells (SCPCs) is variable, necessitating reliable assessment of differentiation and validation of cell quality and safety. Phenotyping is often performed via label-based methods including immunofluorescent staining or flow cytometry analysis. These approaches are often expensive, laborious, time-consuming, destructive, and severely limits their use in large scale cell therapy manufacturing settings. On the other hand, cellular biophysical properties have demonstrated a strong correlation to cell state, quality and functionality and can be measured with ingenious label-free technologies in a rapid and non-destructive manner., Method: In this study, we report the use of Magnetic Resonance Relaxometry (MRR), a rapid and label-free method that indicates iron levels based on its readout (T
2 ). Briefly, we differentiated human iPSCs into SCPCs and compared key iPSC and SCPC cellular markers to their intracellular iron content (Fe3+ ) at different stages of the differentiation process., Results: With MRR, we found that intracellular iron of iPSCs and SCPCs were distinctively different allowing us to accurately reflect varying levels of residual undifferentiated iPSCs (i.e., OCT4+ cells) in any given population of SCPCs. MRR was also able to predict Day 10 SCPC OCT4 levels from Day 1 undifferentiated iPSC T2 values and identified poorly differentiated SCPCs with lower T2 , indicative of lower neural progenitor (SOX1) and stem cell (Nestin) marker expression levels. Lastly, MRR was able to provide predictive indications for the extent of differentiation to Day 28 spinal cord motor neurons (ISL-1/SMI-32) based on the T2 values of Day 10 SCPCs., Conclusion: MRR measurements of iPSCs and SCPCs has clearly indicated its capabilities to identify and quantify key phenotypes of iPSCs and SCPCs for end-point validation of safety and quality parameters. Thus, our technology provides a rapid label-free method to determine critical quality attributes in iPSC-derived progenies and is ideally suited as a quality control tool in cell therapy manufacturing., Competing Interests: Declarations. Ethics approval and consent to participate: This study utilized deidentified iPSC lines and does not involve animals, clinical experiments, or primary patient-derived human tissues. Derivation of the CLEC23 iPSC line had received prior IRB approval (document no.: IRB-2020-05-026, date of approval: 02/06/2020, name of institution: NTU). BJ-iPSCs were derived from the BJ (ATCC CRL-2522) cell line obtained from ATCC ( https://www.atcc.org/products/crl-2522 ) and does not require approvals for derivation as it does not involve the use of patient-derived material. NTU-IRB waived the need for ethical approvals regarding this study in accordance with relevant guidelines and regulations following the Singapore’s Human Tissue Framework under the Human Biomedical Research Act. In accordance to Singapore’s Human Tissue Framework, any form of human biological material which has been substantially manipulated and de-identified are no longer considered human material, and, therefore, ethical approvals or consent to participate does not apply. Consent for publication: Not applicable. Competing interests: J.H., S.Y.C., D.N.R, J.T. filed an intellectual property with a patent that is co-owned by SMART Singapore-MIT Alliance for Research and Technology and NTU. The other authors declared no potential conflicts of interest., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
3. Enterovirus-A71 preferentially infects and replicates in human motor neurons, inducing neurodegeneration by ferroptosis.
- Author
-
Chooi WH, Winanto, Zeng Y, Lee CY, Lim ZQ, Gautam P, Chu JJH, Loh YH, Alonso S, and Ng SY
- Subjects
- Humans, Virus Replication, Mitochondria metabolism, Deferoxamine pharmacology, Viral Load, Iron metabolism, Ferritins metabolism, Ferritins genetics, Ferroptosis drug effects, Enterovirus A, Human physiology, Enterovirus A, Human genetics, Enterovirus A, Human drug effects, Motor Neurons virology, Motor Neurons metabolism, Enterovirus Infections virology, Enterovirus Infections metabolism
- Abstract
Enterovirus A71 (EV-A71) causes Hand, Foot, and Mouth Disease and has been clinically associated with neurological complications. However, there is a lack of relevant models to elucidate the neuropathology of EV-A71 and its mechanism, as the current models mainly utilize animal models or immortalized cell lines. In this study, we established a human motor neuron model for EV-A71 infection. Single cell transcriptomics of a mixed neuronal population reveal higher viral RNA load in motor neurons, suggesting higher infectivity and replication of EV-A71 in motor neurons. The elevated RNA load in motor neurons correlates with the downregulation of ferritin-encoding genes. Subsequent analysis confirms that neurons infected with EV-A71 undergo ferroptosis, as evidenced by increased levels of labile Fe
2+ and peroxidated lipids. Notably, the Fe2+ chelator Deferoxamine improves mitochondrial function and promotes survival of motor neurons by 40% after EV-A71 infection. These findings deepen understanding of the molecular pathogenesis of EV-A71 infection, providing insights which suggest that improving mitochondrial respiration and inhibition of ferroptosis can mitigate the impact of EV-A71 infection in the central nervous system.- Published
- 2024
- Full Text
- View/download PDF
4. Defined hydrogels for spinal cord organoids: challenges and potential applications.
- Author
-
Chooi WH, Wu Y, and Ng SY
- Published
- 2024
- Full Text
- View/download PDF
5. Correction to "Encapsulation of Human Spinal Cord Progenitor Cells in Hyaluronan-Gelatin Hydrogel for Spinal Cord Injury Treatment".
- Author
-
Kwokdinata C, Ramanujam V, Chen J, Nunes de Oliveira P, Nai MH, Chooi WH, Lim CT, Ng SY, David L, and Chew SY
- Published
- 2024
- Full Text
- View/download PDF
6. Label-Free and High-Throughput Removal of Residual Undifferentiated Cells From iPSC-Derived Spinal Cord Progenitor Cells.
- Author
-
Nguyen TD, Chooi WH, Jeon H, Chen J, Tan J, Roxby DN, Lee CY, Ng SY, Chew SY, and Han J
- Subjects
- Humans, Spinal Cord pathology, Cell Differentiation physiology, Induced Pluripotent Stem Cells, Neural Stem Cells, Spinal Cord Injuries therapy, Spinal Cord Injuries pathology
- Abstract
The transplantation of spinal cord progenitor cells (SCPCs) derived from human-induced pluripotent stem cells (iPSCs) has beneficial effects in treating spinal cord injury (SCI). However, the presence of residual undifferentiated iPSCs among their differentiated progeny poses a high risk as these cells can develop teratomas or other types of tumors post-transplantation. Despite the need to remove these residual undifferentiated iPSCs, no specific surface markers can identify them for subsequent removal. By profiling the size of SCPCs after a 10-day differentiation process, we found that the large-sized group contains significantly more cells expressing pluripotent markers. In this study, we used a sized-based, label-free separation using an inertial microfluidic-based device to remove tumor-risk cells. The device can reduce the number of undifferentiated cells from an SCPC population with high throughput (ie, >3 million cells/minute) without affecting cell viability and functions. The sorted cells were verified with immunofluorescence staining, flow cytometry analysis, and colony culture assay. We demonstrated the capabilities of our technology to reduce the percentage of OCT4-positive cells. Our technology has great potential for the "downstream processing" of cell manufacturing workflow, ensuring better quality and safety of transplanted cells., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
7. Encapsulation of Human Spinal Cord Progenitor Cells in Hyaluronan-Gelatin Hydrogel for Spinal Cord Injury Treatment.
- Author
-
Kwokdinata C, Ramanujam V, Chen J, de Oliveira PN, Nai MH, Chooi WH, Lim CT, Ng SY, David L, and Chew SY
- Abstract
Transplanting human induced pluripotent stem cells (iPSCs)-derived spinal cord progenitor cells (SCPCs) is a promising approach to treat spinal cord injuries. However, stem cell therapies face challenges in cell survival, cell localization to the targeted site, and the control of cell differentiation. Here, we encapsulated SCPCs in thiol-modified hyaluronan-gelatin hydrogels and optimized scaffold mechanical properties and cell encapsulation density to promote cell viability and neuronal differentiation in vitro and in vivo . Different compositions of hyaluronan-gelatin hydrogels formulated by varying concentrations of poly(ethylene glycol) diacrylate were mechanically characterized by using atomic force microscopy. In vitro SCPC encapsulation study showed higher cell viability and proliferation with lower substrate Young's modulus (200 Pa vs 580 Pa) and cell density. Moreover, the soft hydrogels facilitated a higher degree of neuronal differentiation with extended filament structures in contrast to clumped cellular morphologies obtained in stiff hydrogels ( p < 0.01). When transplanted in vivo , the optimized SCPC-encapsulated hydrogels resulted in higher cell survival and localization at the transplanted region as compared to cell delivery without hydrogel encapsulation at 2 weeks postimplantation within the rat spinal cord ( p < 0.01). Notably, immunostaining demonstrated that the hydrogel-encapsulated SCPCs differentiated along the neuronal and oligodendroglial lineages in vivo. The lack of pluripotency and proliferation also supported the safety of the SCPC transplantation approach. Overall, the injectable hyaluronan-gelatin hydrogel shows promise in supporting the survival and neural differentiation of human SCPCs after transplantation into the spinal cord.
- Published
- 2023
- Full Text
- View/download PDF
8. Challenges of having a child with thalassemia in Pakistan: A phenomenological study.
- Author
-
Rehman IU, Khan TM, Bukhsh A, Munawar K, Suleiman AK, Ming LC, Chooi WH, Al-Worafi YM, Tahir H, and Choudhry FR
- Subjects
- Female, Humans, Child, Male, Pakistan epidemiology, Parents psychology, Pain, Qualitative Research, Quality of Life, Thalassemia diagnosis, Thalassemia epidemiology, Thalassemia therapy
- Abstract
Background: Thalassemia is a persistent hemolytic disease and has debilitating effects on patients and their parents. Parents of these children experience pain and suffer from additional emotional strain as they provide daily and lifetime care and are mostly concerned about the health and future of their children., Aim: The study aimed to understand the experiences of parents of children with thalassemia related to their family, financial, social, treatment, and psychological issues in Pakistan., Methods: This descriptive phenomenological study recruited 21 parents of children with thalassemia through purposive sampling until data saturation was achieved. Analysis of transcribed interviews was performed through Colaizzi's method and themes and subthemes revolving around diagnosis, challenges, and treatment issues were extracted., Findings: A total of 21 Pakistani parents participated in this study. Most of the participants were females (n = 16, 76.19%), housewives/stay-at-home moms (n = 13 (61.90%), and were uneducated (n = 6, 28.57%). Regarding genetic traits, only three (14.28%) parents declared that they had genetic traits of thalassemia. The findings of our study revealed that thalassemia is enormously influenced by psychosocial and economic problems because of this disease in their families., Conclusion: Our findings indicated that parents of these children face multi-faceted challenges, such as physical, socio-emotional, financial, and familial. These findings may lead to an adequate understanding of their individual needs and efficient utilization of supportive and care programs., Practice Implications: An understanding of such experiences, involving those distinctive to Pakistani culture, is especially vital to inform the care of these children and enhance their quality of life., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. Defined Alginate Hydrogels Support Spinal Cord Organoid Derivation, Maturation, and Modeling of Spinal Cord Diseases.
- Author
-
Chooi WH, Ng CY, Ow V, Harley J, Ng W, Hor JH, Low KE, Malleret B, Xue K, and Ng SY
- Subjects
- Humans, Alginates pharmacology, Reproducibility of Results, Organoids, Hydrogels pharmacology, Hydrogels metabolism, Spinal Cord Diseases metabolism
- Abstract
In the process of generating organoids, basement membrane extracts or Matrigel are often used to encapsulate cells but they are poorly defined and contribute to reproducibility issues. While defined hydrogels are increasingly used for organoid culture, the effects of replacing Matrigel with a defined hydrogel on neural progenitor growth, neural differentiation, and maturation within organoids are not well-explored. In this study, the use of alginate hydrogels as a Matrigel substitute in spinal cord organoid generation is explored. It is found that alginate encapsulation reduces organoid size variability by preventing organoid aggregation. Importantly, alginate supports neurogenesis and gliogenesis of the spinal cord organoids at a similar efficiency to Matrigel, with mature myelinated neurons observed by day 120. Furthermore, using alginate leads to lower expression of non-spinal markers such as FOXA2, suggesting better control over neural fate specification. To demonstrate the feasibility of using alginate-based organoid cultures as disease models, an isogenic pair of induced pluripotent stem cells discordant for the ALS-causing mutation TDP43
G298S is used, where increased TDP43 mislocalization in the mutant organoids is observed. This study shows that alginate is an ideal substitute for Matrigel for spinal cord organoid derivation, especially when a xeno-free and fully defined 3D culture condition is desired., (© 2022 Wiley-VCH GmbH.)- Published
- 2023
- Full Text
- View/download PDF
10. Orthogonally crosslinked alginate conjugate thermogels with potential for cell encapsulation.
- Author
-
Ow V, Chang JJ, Chooi WH, Boo YJ, Tan RPT, Wong JHM, Parikh BH, Su X, Ng SY, Loh XJ, and Xue K
- Subjects
- Hydrogels pharmacology, Cell Encapsulation, Alginates
- Abstract
Hydrogels with more than one mode of crosslinking have gained interest due to improved control over hydrogel properties such as mechanical strength using multiple stimuli. In this work, sodium alginate was covalently conjugated onto thermoresponsive polyurethanes to prepare hybrid polymers (EPC-Alg) that are responsive to both temperature and Ca
2+ , forming orthogonally crosslinked hydrogels which are non-toxic to cells. Notably, the crosslinks are fully reversible, allowing for gel strength to be modulated via selective removal of either stimulus, or complete deconstruction of the hydrogel network by removing both stimuli. Higher alginate fractions increased the hydrophilicity and Ca2+ response of the EPC-Alg hydrogel, enabling tunable modulation of the thermal stability, stiffness and gelation temperatures. The EPC-Alg hydrogel could sustain protein release for a month and encapsulate neural spheroids with high cell viability after 7-day culture, demonstrating feasibility towards 3D cell encapsulation in cell-based biomedical applications such as cell encapsulation and cell therapy., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
11. Modulating neuroinflammation through molecular, cellular and biomaterial-based approaches to treat spinal cord injury.
- Author
-
Lee CY, Chooi WH, Ng SY, and Chew SY
- Abstract
The neuroinflammatory response that is elicited after spinal cord injury contributes to both tissue damage and reparative processes. The complex and dynamic cellular and molecular changes within the spinal cord microenvironment result in a functional imbalance of immune cells and their modulatory factors. To facilitate wound healing and repair, it is necessary to manipulate the immunological pathways during neuroinflammation to achieve successful therapeutic interventions. In this review, recent advancements and fresh perspectives on the consequences of neuroinflammation after SCI and modulation of the inflammatory responses through the use of molecular-, cellular-, and biomaterial-based therapies to promote tissue regeneration and functional recovery will be discussed., Competing Interests: The authors declare no conflict of interest., (© 2022 The Authors. Bioengineering & Translational Medicine published by Wiley Periodicals LLC on behalf of American Institute of Chemical Engineers.)
- Published
- 2022
- Full Text
- View/download PDF
12. Vaccine contamination: Causes and control.
- Author
-
Chooi WH, Ng PW, Hussain Z, Ming LC, Ibrahim B, and Koh D
- Subjects
- Drug Contamination, Vaccines adverse effects
- Abstract
Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2022
- Full Text
- View/download PDF
13. Cell Membrane-Coated Electrospun Fibers Enhance Keratinocyte Growth through Cell-Type Specific Interactions.
- Author
-
Chooi WH, Dong Q, Low JZY, Yuen C, Chin JS, Lin J, Ong W, Liu Q, and Chew SY
- Subjects
- Humans, Materials Testing, Particle Size, Tissue Scaffolds chemistry, Cell Membrane chemistry, Coated Materials, Biocompatible chemistry, Keratinocytes chemistry, Tissue Engineering
- Abstract
Although cell membrane-coated fiber scaffolds can be useful for regenerative medicine by presenting both cell surface antigens and topographical cues, it remains unknown whether changes in cellular behavior on cell membrane-coated scaffolds are due to specific cell-cell interactions. In this work, the effects of scaffold fiber diameters and surface charges on the cell membrane coating efficiency were explored. Furthermore, fibroblast membrane-coated scaffolds improved the growth of human keratinocytes as compared to red blood cell membrane-coated and plain scaffolds. These results suggest the biofunctionality of cell membrane-coated scaffolds and the specific cell-cell interactions that are preserved to modulate cellular response.
- Published
- 2021
- Full Text
- View/download PDF
14. Oriented and sustained protein expression on biomimicking electrospun fibers for evaluating functionality of cells.
- Author
-
Lin J, Chooi WH, Ong W, Zhang N, Bechler ME, Ffrench-Constant C, and Chew SY
- Subjects
- Animals, Cells, Cultured, Rats, Neurites, Neurons
- Abstract
A proper protein orientation is often required in order to achieve specific protein-receptor interaction to elicit a desired biological response. Here, we present a Protein A-based biomimicking platform that is capable of efficiently orienting proteins for evaluating cellular behaviour. By absorbing Protein A onto aligned bio-mimicking polycaprolactone (PCL) fibers, we demonstrate that protein binding could be retained on these fibers for at least 7 days under physiologically relevant conditions. We further show that Protein A served as a molecular orientor to arrange the recombinant proteins in similar orientations. Such protein-orienting scaffolds were further verified to be biologically functional by using sensitive primary rat cortical neurons (CNs) and oligodendrocyte progenitor cells (OPCs), as model neural cells for a stringent proof of concept. Specifically, CNs that were seeded on fibers coated with Protein A and a known enhancer of neurite growth (L1 Cell Adhesion Molecular L1CAM) displayed the longest total neurite length (462.77 ± 100.79 μm, p < 0.001) as compared to the controls. Besides that, OPCs seeded on fibers coated with Protein A and Neuregulin-1 Type III (Nrg1 type III) (myelin enhancer) produced the longest myelin ensheathment length (19.8 ± 11.69 μm). These results demonstrate the efficacy of this platform for protein screening applications., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
15. Scaffold-Based Delivery of CRISPR/Cas9 Ribonucleoproteins for Genome Editing.
- Author
-
Chooi WH, Chin JS, and Chew SY
- Subjects
- Adhesives chemistry, Animals, CRISPR-Associated Protein 9 chemistry, Cell Line, Gene Expression, Gene Transfer Techniques, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Lipids chemistry, Macromolecular Substances chemistry, Macromolecular Substances metabolism, Nanofibers chemistry, RNA, Guide, CRISPR-Cas Systems chemistry, RNA, Guide, CRISPR-Cas Systems genetics, Ribonucleoproteins genetics, Tissue Engineering, CRISPR-Cas Systems, Gene Editing, Ribonucleoproteins metabolism, Tissue Scaffolds chemistry
- Abstract
The simple and versatile CRISPR/Cas9 system is a promising strategy for genome editing in mammalian cells. Generally, the genome editing components, namely Cas9 protein and single-guide RNA (sgRNA), are delivered in the format of plasmids, mRNA, or ribonucleoprotein (RNP) complexes. In particular, non-viral approaches are desirable as they overcome the safety concerns posed by viral vectors. To control cell fate for tissue regeneration, scaffold-based delivery of genome editing components will offer a route for local delivery and provide possible synergistic effects with other factors such as topographical cues that are co-delivered by the same scaffold. In this chapter, we detail a simple method of surface modification to functionalize electrospun nanofibers with CRISPR/Cas9 RNP complexes. The mussel-inspired bio-adhesive coating will be used as it is a simple and effective method to immobilize biomolecules on the surface. Nanofibers will provide a biomimicking microenvironment and topographical cues to seeded cells. For evaluation, a model cell line with single copies of enhanced green fluorescent protein (U2OS.EGFP) will be used to validate the efficiency of gene disruption.
- Published
- 2021
- Full Text
- View/download PDF
16. Localized delivery of CRISPR/dCas9 via layer-by-layer self-assembling peptide coating on nanofibers for neural tissue engineering.
- Author
-
Zhang K, Chooi WH, Liu S, Chin JS, Murray A, Nizetic D, Cheng D, and Chew SY
- Subjects
- Animals, CRISPR-Cas Systems, Peptides, Rats, Tissue Engineering, Clustered Regularly Interspaced Short Palindromic Repeats, Nanofibers
- Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR) systems have a wide variety of applications besides precise genome editing. In particular, the CRISPR/dCas9 system can be used to control specific gene expression by CRISPR activation (CRISPRa) or interference (CRISPRi). However, the safety concerns associated with viral vectors and the possible off-target issues of systemic administration remain huge concerns to be safe delivery methods for CRISPR/Cas9 systems. In this study, a layer-by-layer (LbL) self-assembling peptide (SAP) coating on nanofibers is developed to mediate localized delivery of CRISPR/dCas9 systems. Specifically, an amphiphilic negatively charged SAP
- is first coated onto PCL nanofibers through strong hydrophobic interactions, and the pDNA complexes and positively charged SAP+ -RGD are then absorbed via electrostatic interactions. The SAPcoated scaffolds facilitate efficient loading and sustained release of the pDNA complexes, while enhancing cell adhesion and proliferation. As a proof of concept, the scaffolds are used to activate GDNF expression in mammalian cells, and the secreted GDNF subsequently promotes neurite outgrowth of rat neurons. These promising results suggest that the LbL self-assembling peptide coated nanofibers can be a new route to establish a bioactive interface, which provides a simple and efficient platform for the delivery of CRISPR/dCas9 systems for regenerative medicine., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
17. Correction: Scaffold mediated gene knockdown for neuronal differentiation of human neural progenitor cells.
- Author
-
Chooi WH, Ong W, Murray A, Lin J, Nizetic D, and Chew SY
- Abstract
Correction for 'Scaffold mediated gene knockdown for neuronal differentiation of human neural progenitor cells' by Wai Hon Chooi et al., Biomater. Sci., 2018, 6, 3019-3029.
- Published
- 2019
- Full Text
- View/download PDF
18. Scaffold-mediated non-viral delivery platform for CRISPR/Cas9-based genome editing.
- Author
-
Chin JS, Chooi WH, Wang H, Ong W, Leong KW, and Chew SY
- Subjects
- Cell Line, Tumor, Humans, Indoles chemistry, Indoles pharmacology, Nanofibers chemistry, Polymers chemistry, Polymers pharmacology, CRISPR-Cas Systems, Gene Editing, Gene Transfer Techniques
- Abstract
Genome editing, especially via the simple and versatile type II CRISPR/Cas9 system, offers an effective avenue to precisely control cell fate, an important aspect of tissue regeneration. Unfortunately, most CRISPR/Cas9 non-viral delivery strategies only utilise micro-/nano-particle delivery methods. While these approaches provide reasonable genomic editing efficiencies, their systemic delivery may lead to undesirable off-target effects. For in vivo applications, a more localized and sustained delivery approach may be useful, particularly in the context of tissue regeneration. Here, we developed a scaffold that delivers the CRISPR/Cas9 components (i.e. single guide RNA (sgRNA) and Cas9 protein complexes) in a localized and non-viral manner. Specifically, using mussel-inspired bioadhesive coating, polyDOPA-melanin (pDOPA), we adsorbed Cas9:sgRNA lipofectamine complexes onto bio-mimicking fiber scaffolds. To evaluate the genome-editing efficiency of this platform, U2OS.EGFP cells were used as the model cell type. pDOPA coating was essential in allowing Cas9:sgRNA lipofectamine complexes to adhere onto the scaffolds with a higher loading efficiency, while laminin coating was necessary for maintaining cell viability and proliferation on the pDOPA-coated fibers for effective gene editing (21.5% editing efficiency, p < 0.001). Importantly, U2OS.EGFP cells took up Cas9:sgRNA lipofectamine complexes directly from the scaffolds via reverse transfection. Overall, we demonstrate the efficacy of such fiber scaffolds in providing localized, sustained and non-viral delivery of Cas9:sgRNA complexes. Such genome editing scaffolds may find useful applications in tissue regeneration. STATEMENT OF SIGNIFICANCE: Currently, there is a lack of effective non-viral means to deliver CRISPR/Cas9 components for genome editing. Most existing approaches only utilize micro-/nano-particles by injection or systemic delivery, which may lead to undesirable off-target effects. Here, we report a platform that delivers the CRISPR/Cas9 components (i.e. single guide RNA (sgRNA) and Cas9 protein complexes) in a localized and sustained manner. We used mussel-inspired bioadhesive coating to functionalize the bio-mimicking fiber scaffolds with Cas9:sgRNA lipofectamine complexes, to allow effective gene editing for the cells seeded on the scaffolds. Importantly, the cells took up Cas9:sgRNA lipofectamine complexes directly from the scaffolds. Such genome editing scaffolds may find useful applications in tissue regeneration., (Copyright © 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
19. Modulation of cell-cell interactions for neural tissue engineering: Potential therapeutic applications of cell adhesion molecules in nerve regeneration.
- Author
-
Chooi WH and Chew SY
- Subjects
- Animals, Biocompatible Materials therapeutic use, Cell Adhesion Molecules pharmacology, Cell Adhesion Molecules therapeutic use, Humans, Nerve Regeneration drug effects, Neural Stem Cells metabolism, Oligodendroglia cytology, Oligodendroglia metabolism, Cell Adhesion Molecules metabolism, Cell Communication drug effects, Neural Stem Cells cytology, Neurogenesis drug effects, Tissue Engineering methods
- Abstract
Neural tissue engineering holds great promise in repairing damaged nerve tissues. However, despite the promising results in regenerating the injured nervous system, tissue engineering approaches are still insufficient to result in full functional recovery in severe nerve damages. Majority of these approaches only focus on growth factors and cell-extracellular matrix (ECM) interactions. As another important component in nerve tissues, the potential of modulating cell-cell interactions as a strategy to promote regeneration has been overlooked. Within the central nervous system, there are considerably more cell-cell communications as compared to cell-ECM interactions, since the ECM only contributes 10%-20% of the total tissue volume. Therefore, modulating cell-cell interactions through cell adhesion molecules (CAMs) such as cadherins, neural cell adhesion molecules (NCAM) and L1, may be a potential alternative to improve nerve regeneration. This paper will begin by reviewing the CAMs that play important roles in neurogenic processes. Specifically, we focused on 3 areas, namely the roles of CAMs in neurite outgrowth and regeneration; remyelination; and neuronal differentiation. Following that, we will discuss existing tissue engineering approaches that utilize CAMs and biomaterials to control nerve regeneration. We will also suggest other potential methods that can deliver CAMs efficiently to injured nerve tissues. Overall, we propose that utilizing CAMs with biomaterials may be a promising therapeutic strategy for nerve regeneration., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
20. Scaffold mediated gene knockdown for neuronal differentiation of human neural progenitor cells.
- Author
-
Chooi WH, Ong W, Murray A, Lin J, Nizetic D, and Chew SY
- Subjects
- Cell Adhesion genetics, Cell Differentiation genetics, Cell Movement genetics, Cell Survival genetics, Humans, Neurites metabolism, Gene Knockdown Techniques methods, Induced Pluripotent Stem Cells cytology, Neural Stem Cells cytology, Neurons cytology
- Abstract
The use of human induced pluripotent stem cell-derived neural progenitor cells (hiPSC-NPCs) is an attractive therapeutic option for damaged nerve tissues. To direct neuronal differentiation of stem cells, we have previously developed an electrospun polycaprolactone nanofiber scaffold that was functionalized with siRNA targeting Re-1 silencing transcription factor (REST), by mussel-inspired bioadhesive coating. However, the efficacy of nanofiber-mediated RNA interference on hiPSC-NPCs differentiation remains unknown. Furthermore, interaction between such cell-seeded scaffolds with injured tissues has not been tested. In this study, scaffolds were optimized for REST knockdown in hiPSC-NPCs to enhance neuronal differentiation. Specifically, the effects of two different mussel-inspired bioadhesives and transfection reagents were analyzed. Scaffolds functionalized with RNAiMAX Lipofectamine-siREST complexes enhanced the differentiation of hiPSC-NPCs into TUJ1
+ cells (60% as compared to 22% in controls with scrambled siNEG after 9 days) without inducing high cytotoxicity. When cell-seeded scaffolds were transplanted to transected spinal cord organotypic slices, similar efficiency in neuronal differentiation was observed. The scaffolds also supported the migration of cells and neurite outgrowth from the spinal cord slices. Taken together, the results suggest that this scaffold can be effective in enhancing hiPSC-NPC neuronal commitment by gene-silencing for the treatment of injured spinal cords.- Published
- 2018
- Full Text
- View/download PDF
21. Maturation of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) in 3D collagen matrix: Effects of niche cell supplementation and mechanical stimulation.
- Author
-
Zhang W, Kong CW, Tong MH, Chooi WH, Huang N, Li RA, and Chan BP
- Subjects
- Animals, Coculture Techniques, Gene Expression Regulation drug effects, Human Embryonic Stem Cells drug effects, Human Embryonic Stem Cells metabolism, Humans, Male, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells drug effects, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Rats, Tissue Engineering, Cell Differentiation drug effects, Collagen pharmacology, Human Embryonic Stem Cells cytology, Myocytes, Cardiac cytology, Stem Cell Niche drug effects, Stress, Mechanical
- Abstract
Cardiomyocytes derived from human embryonic stem cells (hESC-CMs) are regarded as a promising source for regenerative medicine, drug testing and disease modeling. Nevertheless, cardiomyocytes are immature in terms of their contractile structure, metabolism and electrophysiological properties. Here, we fabricate cardiac muscle strips by encapsulating hESC-CMs in collagen-based biomaterials. Supplementation of niche cells at 3% to the number of hESC-CMs enhance the maturation of the hESC-CMs in 3D tissue matrix. The benefits of adding mesenchymal stem cells (MSCs) are comparable to that of adding fibroblasts. These two cell types demonstrate similar effects in promoting the compaction and cell spreading, as well as expression of maturation markers at both gene and protein levels. Mechanical loading, particularly cyclic stretch, produces engineered cardiac tissues with higher maturity in terms of twitch force, elastic modulus, sarcomere length and molecular signature, when comparing to static stretch or non-stretched controls. The current study demonstrates that the application of niche cells and mechanical stretch both stimulate the maturation of hESC-CMs in 3D architecture. Our results therefore suggest that this 3D model can be used for in vitro cardiac maturation study., Statement of Significance: Cardiomyocytes derived from human embryonic stem cells (hESC-CMs) are regarded as being a promising source of cells for regenerative medicine, drug testing and disease modeling. Nevertheless, cardiomyocytes are immature in terms of their contractile structure, metabolism and electrophysiological properties. In the current study, we have fabricated cardiac muscle strips by encapsulating hESC-CMs in collagen-based biomaterials and demonstrated that supplementation of mesenchymal niche cells as well as provision of mechanical loading particularly stretching have significantly promoted the maturation of the cardiomyocytes and hence improved the mechanical functional characteristics of the tissue strips. Specifically, with 3% niche cells including both fibroblasts and mesenchymal stem cells, a more mature hESC-CMs derived cardiac strip was resulted, in terms of compaction and spreading of cells, and upregulation of molecular signature in both gene and protein expression of maturation. Mechanical loading, particularly cyclic stretch, produces engineered cardiac tissues with higher maturity in terms of molecular signature markers and functional parameters including twitch force, elastic modulus and sarcomere length, when comparing with static stretch or non-stretched controls. The current study demonstrates that the application of niche cells and mechanical stretch both stimulate the maturation of hESC-CMs in 3D architecture, resulting in more mature cardiac strips. Our results contribute to bioengineering of functional heart tissue strips for drug screening and disease modeling., (Copyright © 2016 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
22. Correction: Loading-Induced Heat-Shock Response in Bovine Intervertebral Disc Organ Culture.
- Author
-
Chooi WH, Chan SC, Gantenbein B, and Chan BP
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0161615.].
- Published
- 2016
- Full Text
- View/download PDF
23. Loading-Induced Heat-Shock Response in Bovine Intervertebral Disc Organ Culture.
- Author
-
Chooi WH, Chan SC, Gantenbein B, and Chan BP
- Subjects
- Animals, Cattle, Weight-Bearing, Extracellular Matrix metabolism, Gene Expression Regulation, HSP70 Heat-Shock Proteins biosynthesis, Heat-Shock Response, Intervertebral Disc metabolism, Stress, Mechanical
- Abstract
Mechanical loading has been shown to affect cell viability and matrix maintenance in the intervertebral disc (IVD) but there is no investigation on how cells survive mechanical stress and whether the IVD cells perceive mechanical loading as stress and respond by expression of heat shock proteins. This study investigates the stress response in the IVD in response to compressive loading. Bovine caudal disc organ culture was used to study the effect of physiological range static loading and dynamic loading. Cell activity, gene expression and immunofluorescence staining were used to analyze the cell response. Cell activity and cytoskeleton of the cells did not change significantly after loading. In gene expression analysis, significant up-regulation of heat shock protein-70 (HSP70) was observed in nucleus pulposus after two hours of loading. However, the expression of the matrix remodeling genes did not change significantly after loading. Similarly, expressions of stress response and matrix remodeling genes changed with application and removal of the dynamic loading. The results suggest that stress response was induced by physiological range loading without significantly changing cell activity and upregulating matrix remodeling. This study provides direct evidence on loading induced stress response in IVD cells and contributes to our understanding in the mechanoregulation of intervertebral disc cells., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
24. Compression loading-induced stress responses in intervertebral disc cells encapsulated in 3D collagen constructs.
- Author
-
Chooi WH and Chan BP
- Subjects
- Animals, Cattle, Cell Survival, Cells, Cultured, Gene Expression Regulation, HSP27 Heat-Shock Proteins genetics, HSP27 Heat-Shock Proteins metabolism, HSP70 Heat-Shock Proteins genetics, HSP70 Heat-Shock Proteins metabolism, Models, Biological, Nucleus Pulposus metabolism, Pressure, Collagen metabolism, Heat-Shock Response, Nucleus Pulposus cytology, Stress, Mechanical, Unfolded Protein Response
- Abstract
Cells protect themselves from stresses through a cellular stress response. In the interverebral disc, such response was also demonstrated to be induced by various environmental stresses. However, whether compression loading will cause cellular stress response in the nucleus pulposus cells (NPCs) is not well studied. By using an in vitro collagen microencapsulation model, we investigated the effect of compression loading on the stress response of NPCs. Cell viability tests, and gene and protein expression experiments were conducted, with primers for the heat shock response (HSR: HSP70, HSF1, HSP27 and HSP90), and unfolded protein response (UPR: GRP78, GRP94, ATF4 and CHOP) genes and an antibody to HSP72. Different gene expression patterns occurred due to loading type throughout experiments. Increasing the loading strain for a short duration did not increase the stress response genes significantly, but over longer durations, HSP70 and HSP27 were upregulated. Longer loading durations also resulted in a continuous upregulation of HSR genes and downregulation of UPR genes, even after load removal. The rate of apoptosis did not increase significantly after loading, suggesting that stress response genes might play a role in cell survival following mechanical stress. These results demonstrate how mechanical stress might induce and control the expression of HSR and UPR genes in NPCs.
- Published
- 2016
- Full Text
- View/download PDF
25. Bioengineering a multicomponent spinal motion segment construct--a 3D model for complex tissue engineering.
- Author
-
Chik TK, Chooi WH, Li YY, Ho FC, Cheng HW, Choy TH, Sze KY, Luk KK, Cheung KM, and Chan BP
- Subjects
- Animals, Cell Adhesion, Cell Survival, Intervertebral Disc Degeneration surgery, Rabbits, Biocompatible Materials chemistry, Collagen chemistry, Implants, Experimental, Mesenchymal Stem Cells metabolism, Spine, Tissue Engineering methods
- Abstract
Intervertebral disc degeneration is an important clinical problem but existing treatments have significant drawbacks. The ability to bioengineer the entire spinal motion segment (SMS) offers hope for better motion preservation strategies but is extremely challenging. Here, fabrication of a multicomponent SMS construct with complex hierarchical organization from mesenchymal stem cells and collagen-based biomaterials, using a module-based integrative approach, is reported. The construct consists of two osteochondral subunits, a nucleus pulposus (NP-)-like core and a multi-lamellae annulus fibrosus (AF-)-like component. Chondrogenic medium is crucial for stabilizing the osteochondral subunits, which are shown to allow passive nutrient diffusion, while cyclic compression is necessary for better fiber matrix organization. Cells adhere, survive, and interact with the NP-like core. Cyclic torsional loading stimulates cell alignment in the AF-like lamellae and the number of lamellae affects the mechanical properties of the construct. This work represents an important milestone in SMS tissue engineering and provides a 3D model for studying tissue maturation and functional remodeling., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
26. Development of a novel biodegradable drug-eluting ventilation tube for chronic otitis media with effusion.
- Author
-
Gan CW, Chooi WH, Ng HC, Wong YS, Venkatraman SS, and Lim LH
- Subjects
- Animals, Coated Materials, Biocompatible, Guinea Pigs, Lactic Acid, Otitis Media with Effusion microbiology, Otoscopy, Polyglycolic Acid, Polylactic Acid-Polyglycolic Acid Copolymer, Pseudomonas Infections microbiology, Random Allocation, Middle Ear Ventilation instrumentation, Ofloxacin pharmacology, Otitis Media with Effusion drug therapy, Otitis Media with Effusion surgery, Pseudomonas Infections drug therapy, Pseudomonas Infections surgery, Pseudomonas aeruginosa drug effects
- Abstract
Objectives/hypothesis: To develop a novel drug-eluting biodegradable ventilation tube (VT), to evaluate in vitro sustained release and antibacterial adherence of ofloxacin-loaded biodegradable VT on Pseudomonas aeruginosa, and to evaluate in vivo biodegradation of VT in guinea pig ears., Study Design: A randomized animal study., Methods: In vitro drug release and degradation of ofloxacin-loaded VT were studied in water for 3 months. Bacterial adherence was evaluated by inoculating the VT with P aeruginosa suspension for 6 days. Scanning electron microscopy (SEM) was used for morphologic analysis. Guinea pigs were assigned to three groups: commercial Mini Shah VT, biodegradable unloaded VT, and biodegradable ofloxacin-loaded VT. Myringotomy and VT insertion were performed. SEM of VTs and histology were performed at 2, 4, 10, and 18 weeks., Results: A total of 81.7% of ofloxacin in VT was eluted over 3 months. Biodegradable VTs had smoother surfaces and less bacteria adherence compared to Mini Shah VTs. VTs with ofloxacin had the least bacteria adherence. VTs resulted in neither inflammation nor otorrhea 18 weeks postinsertion in guinea pigs. Histology showed the new VTs were biocompatible. The VTs were still functioning and patent after 18 weeks postinsertion but had started degrading., Conclusions: The first novel biodegradable ofloxacin-loaded VT with sustainable drug release technology and antibacterial adherence property was studied. Patency beyond 4.5 months allowed an adequate period of ventilation. The complete degradation of the VT warrants further studies to evaluate the duration of VT resorption in situ and healing of the ear drum., (Copyright © 2012 The American Laryngological, Rhinological, and Otological Society, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
27. Determination and validation of reference gene stability for qPCR analysis in polysaccharide hydrogel-based 3D chondrocytes and mesenchymal stem cell cultural models.
- Author
-
Chooi WH, Zhou R, Yeo SS, Zhang F, and Wang DA
- Subjects
- Alginates metabolism, Animals, Chondrocytes cytology, Chondrocytes metabolism, Gene Expression, Glucuronic Acid metabolism, Hexuronic Acids metabolism, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Rabbits, Reference Standards, Swine, Cell Culture Techniques methods, Chondrocytes physiology, Hydrogel, Polyethylene Glycol Dimethacrylate chemistry, Mesenchymal Stem Cells physiology, Polysaccharides metabolism, Reverse Transcriptase Polymerase Chain Reaction methods
- Abstract
Gene expression study is widely used to obtain information of the cell activities and phenotypes. To quantify gene expression, measurement of the mRNA copy number is commonly done by quantitative RT-PCR (RT-qPCR). However, proper reference gene is needed for different tissues to normalize the expression level of different genes accurately. In this study, reference gene determination was done for three-dimensional (3D) artificial tissue constructs in hydrogel. Porcine synovium-derived mesenchymal stem cells (SMSCs) and rabbit chondrocytes were cultured in both alginate and agarose hydrogels to set up four different 3D culture systems to form the artificial tissue constructs. The gene expression levels of candidate genes were determined by RT-qPCR and then analyzed by geNorm, Bestkeeper, and Normfinder. For porcine SMSCs, PPIA, and TBP were selected for tissue in alginate scaffold whereas HPRT and TBP were selected for the agarose scaffold system. On the other hand, HPRT, PPIA, and RPL18 were the stable reference genes for rabbit chondrocytes in alginate scaffold while TBP, RPL5, and RPL18 were selected for rabbit chondrocytes in agarose scaffold. This study has further indicated that suitable reference genes are different for each tissue and study purpose. The reference genes are expressed in different stability when a scaffold of different material is used.
- Published
- 2013
- Full Text
- View/download PDF
28. Chondrogenesis of synovium-derived mesenchymal stem cells in gene-transferred co-culture system.
- Author
-
Varshney RR, Zhou R, Hao J, Yeo SS, Chooi WH, Fan J, and Wang DA
- Subjects
- Animals, Blotting, Western, Cell Survival drug effects, Chondrocytes cytology, Chondrocytes drug effects, Chondrocytes metabolism, Culture Media, Conditioned pharmacology, Enzyme-Linked Immunosorbent Assay, Gene Expression Regulation drug effects, Humans, Immunohistochemistry, Mesenchymal Stem Cells drug effects, Mesenchymal Stem Cells metabolism, Rabbits, Reverse Transcriptase Polymerase Chain Reaction, Transforming Growth Factor beta3 metabolism, Chondrogenesis drug effects, Coculture Techniques methods, Mesenchymal Stem Cells cytology, Synovial Membrane cytology, Transfection methods
- Abstract
A co-culture strategy has been developed in this study wherein rabbit synovial mesenchymal stem cells (SMSCs) are co-cultured with growth factor (GF) transfected articular chondrocytes. Toward this end, both SMSCs and early passage rabbit articular chondrocytes that had been adenovirally transduced with transforming growth factor-beta 3 (TGF-beta3) gene were separately encapsulated in alginate beads and co-cultured in the same pool of chondrogenic medium. The chondrocytes act as transfected companion cells (TCCs) providing GF supply to induce chondrogenic differentiation of SMSCs that play the role of therapeutic progenitor cells (TPCs). Against the same TCC based TGF-beta3 release profile, the co-culture was started at different time points (Day 0, Day 10 and Day 20) but made to last for identical periods of exposure (30 days) so that the exposure conditions could be optimized in terms of initiation and duration. Transfection of TCCs prevents the stem cell based TPCs from undergoing the invasive procedure. It also prevents unpredictable complications in the TPCs caused by long-term constitutive over-expression of a GF. The adenovirally transfected TCCs exhibit a transient GF expression which results in a timely termination of GF supply to the TPCs. The TCC-sourced transgenic TGF-beta3 successfully induced chondrogenesis in the TPCs. Real-time PCR results show enhanced expression of cartilage markers and immuno/histochemical staining for Glycosaminoglycans (GAG) and Collagen II also shows abundant extracellular matrix (ECM) production and chondrogenic morphogenesis in the co-cultured TPCs. These results confirm the efficacy of directing stem cell differentiation towards chondrogenesis and cartilage tissue formation by co-culturing them with GF transfected chondrocytes.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.