1
|
Tanneberger AE, Blomberg R, Bilousova G, Ryan AL, Magin CM. Engineered hydrogel biomaterials facilitate lung progenitor cell differentiation from induced pluripotent stem cells. Am J Physiol Lung Cell Mol Physiol 2025; 328:L379-L388. [PMID: 39884665 DOI: 10.1152/ajplung.00419.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/15/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025] Open
Abstract
Lung progenitor (LP) cells identified by the expression of transcription factor NK2 homeobox 1 (NKX2.1) are essential for the development of all lung epithelial cell types and hold tremendous potential for pulmonary research and translational regenerative medicine applications. Here, we present engineered hydrogels as a promising alternative to the naturally derived materials that are often used to differentiate human-induced pluripotent stem cells (iPSCs) into LP cells. Poly(ethylene glycol) norbornene (PEGNB) hydrogels with defined composition were used to systematically investigate the role of microenvironmental stiffness, cell origin, and splitting during the differentiation process. Results demonstrated that each factor impacted LP differentiation efficiency and that the soft hydrogels replicating healthy lung stiffness [elastic modulus (E) = 4.00 ± 0.25 kPa] produced the highest proportion of LP cells based on flow cytometric analysis results (54%) relative to the stiff hydrogels (48%) and Matrigel controls (32%) at the end of the nonsplit differentiation protocol. Collectively, these results showed that engineered hydrogels provide a well-defined microenvironment for iPSC-to-LP differentiation and perform as effectively as the current gold standard Matrigel-coated tissue culture plastic. Adopting engineered biomaterials in cell culture protocols may enable greater control over differentiation parameters and has the potential to enhance the clinical translation of iPSC-derived LP cells.NEW & NOTEWORTHY Standard iPSC differentiation protocols rely on Matrigel, a basement membrane extract from mouse sarcoma cells that is poorly defined and exhibits significant batch-to-batch variation. Due to these limitations, Matrigel-derived products have never been approved by the Food and Drug Administration. This study introduces a novel method for differentiating iPSCs into lung progenitor cells using well-defined hydrogel substrates. These biomaterials not only enhance differentiation efficiency but also streamline the regulatory pathway, facilitating their potential therapeutic application.
Collapse
Affiliation(s)
- Alicia E Tanneberger
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, United States
| | - Rachel Blomberg
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, United States
| | - Ganna Bilousova
- Department of Dermatology, Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Amy L Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
2
|
Chu X, Zhou Z, Qian X, Shen H, Cheng H, Zhang J. Functional regeneration strategies of hair follicles: advances and challenges. Stem Cell Res Ther 2025; 16:77. [PMID: 39985119 PMCID: PMC11846195 DOI: 10.1186/s13287-025-04210-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/29/2025] [Indexed: 02/24/2025] Open
Abstract
Hair follicles are essential appendages of human skin that function in protection, sensation, thermoregulation and social interactions. The multicellular components, particularly the dermal papilla, matrix and bulge housing stem cells, enable cyclic hair growth postnatally. However, miniaturization and loss of hair follicles can occur in the context of ageing, trauma and various alopecia-related diseases. Conventional treatments involve the redistribution of existing follicles, which may not be viable in patients lacking follicular resources. Recent progress in the comprehension of morphogenesis and the development of biomaterials has significantly advanced follicle reconstruction, incorporating organ germ assembling, stem cell induction and bioprinting techniques. Despite these advancements, fully restoring hair follicles remains challenging due to the complexities of replicating embryonic signals and sustaining growth cycles. Identifying suitable cell sources for clinical applications also presents a hurdle. Here, we retrospect the progress made in the field of hair follicle regeneration, aiming to offer an exhaustive analysis on the benefits and limitations of these methods, and to foster the development of innovative solutions.
Collapse
Affiliation(s)
- Xi Chu
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Zhentao Zhou
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Xifei Qian
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Haiyan Shen
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Hanxiao Cheng
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China
| | - Jufang Zhang
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
3
|
Tanneberger AE, Blomberg R, Kary AD, Lu A, Riches DW, Magin CM. Biomaterial-based 3D human lung models replicate pathological characteristics of early pulmonary fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637970. [PMID: 40027659 PMCID: PMC11870410 DOI: 10.1101/2025.02.12.637970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and incurable lung disease characterized by tissue scarring that disrupts gas exchange. Epithelial cell dysfunction, fibroblast activation, and excessive extracellular matrix deposition drive this pathology that ultimately leads to respiratory failure. Mechanistic studies have shown that repeated injury to alveolar epithelial cells initiates an aberrant wound-healing response in surrounding fibroblasts through secretion of mediators like transforming growth factor-β, yet the precise biological pathways contributing to disease progression are not fully understood. To better study these interactions there is a critical need for lung models that replicate the cellular heterogeneity, geometry, and biomechanics of the distal lung microenvironment. In this study, induced pluripotent stem cell-derived alveolar epithelial type II (iATII) cells and human pulmonary fibroblasts were arranged to replicate human lung micro-architecture and embedded in soft or stiff poly(ethylene glycol) norbornene (PEG-NB) hydrogels that recapitulated the mechanical properties of healthy and fibrotic lung tissue, respectively. The co-cultured cells were then exposed to pro-fibrotic biochemical cues, including inflammatory cytokines and growth factors. iATIIs and fibroblasts exhibited differentiation pathways and gene expression patterns consistent with trends observed during IPF progression in vivo. A design of experiments statistical analysis identified stiff hydrogels combined with pro-fibrotic biochemical cue exposure as the most effective condition for modeling fibrosis in vitro. Finally, treatment with Nintedanib, one of only two Food and Drug Administration (FDA)-approved drugs for IPF, was assessed. Treatment reduced fibroblast activation, as indicated by downregulation of key activation genes, and upregulated several epithelial genes. These findings demonstrate that human 3D co-culture models hold tremendous potential for advancing our understanding of IPF and identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Alicia E. Tanneberger
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
| | - Rachel Blomberg
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
| | - Anton D. Kary
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
| | - Andrew Lu
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
| | - David W.H. Riches
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO
- Department of Research, Veterans Affairs Eastern Colorado Health Care System, Aurora, CO
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Chelsea M. Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO
- Department of Pediatrics, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
4
|
Zhang Y, Li L, Dong L, Cheng Y, Huang X, Xue B, Jiang C, Cao Y, Yang J. Hydrogel-Based Strategies for Liver Tissue Engineering. CHEM & BIO ENGINEERING 2024; 1:887-915. [PMID: 39975572 PMCID: PMC11835278 DOI: 10.1021/cbe.4c00079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/15/2024] [Accepted: 09/15/2024] [Indexed: 02/21/2025]
Abstract
The liver's role in metabolism, detoxification, and immune regulation underscores the urgency of addressing liver diseases, which claim millions of lives annually. Due to donor shortages in liver transplantation, liver tissue engineering (LTE) offers a promising alternative. Hydrogels, with their biocompatibility and ability to mimic the liver's extracellular matrix (ECM), support cell survival and function in LTE. This review analyzes recent advances in hydrogel-based strategies for LTE, including decellularized liver tissue hydrogels, natural polymer-based hydrogels, and synthetic polymer-based hydrogels. These materials are ideal for in vitro cell culture and obtaining functional hepatocytes. Hydrogels' tunable properties facilitate creating artificial liver models, such as organoids, 3D bioprinting, and liver-on-a-chip technologies. These developments demonstrate hydrogels' versatility in advancing LTE's applications, including hepatotoxicity testing, liver tissue regeneration, and treating acute liver failure. This review highlights the transformative potential of hydrogels in LTE and their implications for future research and clinical practice.
Collapse
Affiliation(s)
- Yu Zhang
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| | - Luofei Li
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Liang Dong
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Yuanqi Cheng
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Xiaoyu Huang
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Bin Xue
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Chunping Jiang
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| | - Yi Cao
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| | - Jiapeng Yang
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| |
Collapse
|
5
|
Zhang X, Zhao G, Ma T, Simmons CA, Santerre JP. A critical review on advances and challenges of bioprinted cardiac patches. Acta Biomater 2024; 189:1-24. [PMID: 39374681 DOI: 10.1016/j.actbio.2024.09.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
Myocardial infarction (MI), which causes irreversible myocardium necrosis, affects 0.25 billion people globally and has become one of the most significant epidemics of our time. Over the past few years, bioprinting has moved beyond a concept of simply incorporating cells into biomaterials, to strategically defining the microenvironment (e.g., architecture, biomolecular signalling, mechanical stimuli, etc.) within which the cells are printed. Among the different bioprinting applications, myocardial repair is a field that has seen some of the most significant advances towards the management of the repaired tissue microenvironment. This review critically assesses the most recent biomedical innovations being carried out in cardiac patch bioprinting, with specific considerations given to the biomaterial design parameters, growth factors/cytokines, biomechanical and bioelectrical conditioning, as well as innovative biomaterial-based "4D" bioprinting (3D scaffold structure + temporal morphology changes) of myocardial tissues, immunomodulation and sustained delivery systems used in myocardium bioprinting. Key challenges include the ability to generate large quantities of cardiac cells, achieve high-density capillary networks, establish biomaterial designs that are comparable to native cardiac extracellular matrix, and manage the sophisticated systems needed for combining cardiac tissue microenvironmental cues while simultaneously establishing bioprinting technologies yielding both high-speed and precision. This must be achieved while considering quality assurance towards enabling reproducibility and clinical translation. Moreover, this manuscript thoroughly discussed the current clinical translational hurdles and regulatory issues associated with the post-bioprinting evaluation, storage, delivery and implantation of the bioprinted myocardial patches. Overall, this paper provides insights into how the clinical feasibility and important regulatory concerns may influence the design of the bioink (biomaterials, cell sources), fabrication and post-fabrication processes associated with bioprinting of the cardiac patches. This paper emphasizes that cardiac patch bioprinting requires extensive collaborations from imaging and 3D modelling technical experts, biomaterial scientists, additive manufacturing experts and healthcare professionals. Further, the work can also guide the field of cardiac patch bioprinting moving forward, by shedding light on the potential use of robotics and automation to increase productivity, reduce financial cost, and enable standardization and true commercialization of bioprinted cardiac patches. STATEMENT OF SIGNIFICANCE: The manuscript provides a critical review of important themes currently pursued for heart patch bioprinting, including critical biomaterial design parameters, physiologically-relevant cardiac tissue stimulations, and newly emerging cardiac tissue bioprinting strategies. This review describes the limited number of studies, to date in the literature, that describe systemic approaches to combine multiple design parameters, including capabilities to yield high-density capillary networks, establish biomaterial composite designs similar to native cardiac extracellular matrix, and incorporate cardiac tissue microenvironmental cues, while simultaneously establishing bioprinting technologies that yield high-speed and precision. New tools such as artificial intelligence may provide the analytical power to consider multiple design parameters and identify an optimized work-flow(s) for enabling the clinical translation of bioprinted cardiac patches.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| | - Guangtao Zhao
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Tianyi Ma
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administrative Region of China
| | - Craig A Simmons
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| | - J Paul Santerre
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, China; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| |
Collapse
|
6
|
Zheng F, Tian R, Lu H, Liang X, Shafiq M, Uchida S, Chen H, Ma M. Droplet Microfluidics Powered Hydrogel Microparticles for Stem Cell-Mediated Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401400. [PMID: 38881184 DOI: 10.1002/smll.202401400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/21/2024] [Indexed: 06/18/2024]
Abstract
Stem cell-related therapeutic technologies have garnered significant attention of the research community for their multi-faceted applications. To promote the therapeutic effects of stem cells, the strategies for cell microencapsulation in hydrogel microparticles have been widely explored, as the hydrogel microparticles have the potential to facilitate oxygen diffusion and nutrient transport alongside their ability to promote crucial cell-cell and cell-matrix interactions. Despite their significant promise, there is an acute shortage of automated, standardized, and reproducible platforms to further stem cell-related research. Microfluidics offers an intriguing platform to produce stem cell-laden hydrogel microparticles (SCHMs) owing to its ability to manipulate the fluids at the micrometer scale as well as precisely control the structure and composition of microparticles. In this review, the typical biomaterials and crosslinking methods for microfluidic encapsulation of stem cells as well as the progress in droplet-based microfluidics for the fabrication of SCHMs are outlined. Moreover, the important biomedical applications of SCHMs are highlighted, including regenerative medicine, tissue engineering, scale-up production of stem cells, and microenvironmental simulation for fundamental cell studies. Overall, microfluidics holds tremendous potential for enabling the production of diverse hydrogel microparticles and is worthy for various stem cell-related biomedical applications.
Collapse
Affiliation(s)
- Fangqiao Zheng
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
| | - Ruizhi Tian
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hongxu Lu
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiao Liang
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
| | - Muhammad Shafiq
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
| | - Hangrong Chen
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ming Ma
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
7
|
Mishra A, Kumar R, Harilal S, Nigam M, Datta D, Singh S. Emerging Landscape of In Vitro Models for Assessing Rheumatoid Arthritis Management. ACS Pharmacol Transl Sci 2024; 7:2280-2305. [PMID: 39144547 PMCID: PMC11320735 DOI: 10.1021/acsptsci.4c00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024]
Abstract
Rheumatoid arthritis (RA) is a complex condition that is influenced by various causes, including immunological, genetic, and environmental factors. Several studies using animal models have documented immune system dysfunction and described the clinical characteristics of the disease. These studies have provided valuable insights into the pathogenesis of inflammatory arthritis and the identification of new targets for treatment. Nevertheless, none of these animal models successfully replicated all the characteristics of RA. Additionally, numerous experimental medications, which were developed based on our enhanced comprehension of the immune system's function in RA, have shown potential in animal research but ultimately proved ineffective during different stages of clinical trials. There have been several novel therapy alternatives, which do not achieve a consistently outstanding therapeutic outcome in all patients. This underscores the importance of employing the progress in in vitro models, particularly 3D models like tissue explants, and diverse multicomponent approaches such as coculture strategies, synovial membrane, articular cartilage, and subchondral bone models that accurately replicate the structural characteristics of RA pathophysiology. These methods are crucial for the advancement of potential therapeutic strategies. This review discusses the latest advancements in in vitro models and their potential to greatly impact research on managing RA.
Collapse
Affiliation(s)
- Abhay
Prakash Mishra
- Department
of Pharmacology, University of Free State, Bloemfontein 9301, South Africa
- Department
of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Rajesh Kumar
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Seetha Harilal
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Manisha Nigam
- Department
of Biochemistry, Hemvati Nandan Bahuguna
Garhwal University, Srinagar
Garhwal, Uttarakhand 246174, India
| | - Deepanjan Datta
- Department
of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sudarshan Singh
- Office of
Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
- Faculty of
Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
8
|
Chehelgerdi M, Behdarvand Dehkordi F, Chehelgerdi M, Kabiri H, Salehian-Dehkordi H, Abdolvand M, Salmanizadeh S, Rashidi M, Niazmand A, Ahmadi S, Feizbakhshan S, Kabiri S, Vatandoost N, Ranjbarnejad T. Exploring the promising potential of induced pluripotent stem cells in cancer research and therapy. Mol Cancer 2023; 22:189. [PMID: 38017433 PMCID: PMC10683363 DOI: 10.1186/s12943-023-01873-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 11/30/2023] Open
Abstract
The advent of iPSCs has brought about a significant transformation in stem cell research, opening up promising avenues for advancing cancer treatment. The formation of cancer is a multifaceted process influenced by genetic, epigenetic, and environmental factors. iPSCs offer a distinctive platform for investigating the origin of cancer, paving the way for novel approaches to cancer treatment, drug testing, and tailored medical interventions. This review article will provide an overview of the science behind iPSCs, the current limitations and challenges in iPSC-based cancer therapy, the ethical and social implications, and the comparative analysis with other stem cell types for cancer treatment. The article will also discuss the applications of iPSCs in tumorigenesis, the future of iPSCs in tumorigenesis research, and highlight successful case studies utilizing iPSCs in tumorigenesis research. The conclusion will summarize the advancements made in iPSC-based tumorigenesis research and the importance of continued investment in iPSC research to unlock the full potential of these cells.
Collapse
Affiliation(s)
- Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fereshteh Behdarvand Dehkordi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Hamidreza Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | - Mohammad Abdolvand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Sharareh Salmanizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar-Jereeb Street, Isfahan, 81746-73441, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saba Ahmadi
- Department of Molecular and Medical Genetics, Tbilisi State Medical University, Tbilisi, Georgia
| | - Sara Feizbakhshan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saber Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Nasimeh Vatandoost
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tayebeh Ranjbarnejad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
9
|
Fadul SM, Arshad A, Mehmood R. CRISPR-based epigenome editing: mechanisms and applications. Epigenomics 2023; 15:1137-1155. [PMID: 37990877 DOI: 10.2217/epi-2023-0281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
Epigenomic anomalies contribute significantly to the development of numerous human disorders. The development of epigenetic research tools is essential for understanding how epigenetic marks contribute to gene expression. A gene-editing technique known as CRISPR (clustered regularly interspaced short palindromic repeats) typically targets a particular DNA sequence using a guide RNA (gRNA). CRISPR/Cas9 technology has been remodeled for epigenome editing by generating a 'dead' Cas9 protein (dCas9) that lacks nuclease activity and juxtaposing it with an epigenetic effector domain. Based on fusion partners of dCas9, a specific epigenetic state can be achieved. CRISPR-based epigenome editing has widespread application in drug screening, cancer treatment and regenerative medicine. This paper discusses the tools developed for CRISPR-based epigenome editing and their applications.
Collapse
Affiliation(s)
- Shaima M Fadul
- Department of Life Sciences, College of Science & General Studies, Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Aleeza Arshad
- Medical Teaching Insitute, Ayub Teaching Hospital, Abbottabad, 22020, Pakistan
| | - Rashid Mehmood
- Department of Life Sciences, College of Science & General Studies, Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| |
Collapse
|
10
|
Chen X, Li K, Chen J, Tan S. Breakthrough in large-scale production of iPSCs-derived exosomes to promote clinical applications. Front Bioeng Biotechnol 2023; 11:1257186. [PMID: 37691905 PMCID: PMC10484304 DOI: 10.3389/fbioe.2023.1257186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
| | | | | | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Park S, Gwon Y, Khan SA, Jang KJ, Kim J. Engineering considerations of iPSC-based personalized medicine. Biomater Res 2023; 27:67. [PMID: 37420273 DOI: 10.1186/s40824-023-00382-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/19/2023] [Indexed: 07/09/2023] Open
Abstract
Personalized medicine aims to provide tailored medical treatment that considers the clinical, genetic, and environmental characteristics of patients. iPSCs have attracted considerable attention in the field of personalized medicine; however, the inherent limitations of iPSCs prevent their widespread use in clinical applications. That is, it would be important to develop notable engineering strategies to overcome the current limitations of iPSCs. Such engineering approaches could lead to significant advances in iPSC-based personalized therapy by offering innovative solutions to existing challenges, from iPSC preparation to clinical applications. In this review, we summarize how engineering strategies have been used to advance iPSC-based personalized medicine by categorizing the development process into three distinctive steps: 1) the production of therapeutic iPSCs; 2) engineering of therapeutic iPSCs; and 3) clinical applications of engineered iPSCs. Specifically, we focus on engineering strategies and their implications for each step in the development of iPSC-based personalized medicine.
Collapse
Affiliation(s)
- Sangbae Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
- Institute of Nano-Stem Cells Therapeutics, NANOBIOSYSTEM Co, Ltd, Gwangju, 61011, Republic of Korea
| | - Yonghyun Gwon
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Shahidul Ahmed Khan
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kyoung-Je Jang
- Department of Bio-Systems Engineering, Institute of Smart Farm, Gyeongsang National University, Jinju, 52828, Republic of Korea.
- Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Jangho Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Institute of Nano-Stem Cells Therapeutics, NANOBIOSYSTEM Co, Ltd, Gwangju, 61011, Republic of Korea.
| |
Collapse
|
12
|
Barrachina L, Arshaghi TE, O'Brien A, Ivanovska A, Barry F. Induced pluripotent stem cells in companion animals: how can we move the field forward? Front Vet Sci 2023; 10:1176772. [PMID: 37180067 PMCID: PMC10168294 DOI: 10.3389/fvets.2023.1176772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023] Open
Abstract
Following a one medicine approach, the development of regenerative therapies for human patients leads to innovative treatments for animals, while pre-clinical studies on animals provide knowledge to advance human medicine. Among many different biological products under investigation, stem cells are among the most prominent. Mesenchymal stromal cells (MSCs) are extensively investigated, but they present challenges such as senescence and limited differentiation ability. Embryonic stem cells (ESCs) are pluripotent cells with a virtually unlimited capacity for self-renewal and differentiation, but the use of embryos carries ethical concerns. Induced pluripotent stem cells (iPSCs) can overcome all of these limitations, as they closely resemble ESCs but are derived from adult cells by reprogramming in the laboratory using pluripotency-associated transcription factors. iPSCs hold great potential for applications in therapy, disease modeling, drug screening, and even species preservation strategies. However, iPSC technology is less developed in veterinary species compared to human. This review attempts to address the specific challenges associated with generating and applying iPSCs from companion animals. Firstly, we discuss strategies for the preparation of iPSCs in veterinary species and secondly, we address the potential for different applications of iPSCs in companion animals. Our aim is to provide an overview on the state of the art of iPSCs in companion animals, focusing on equine, canine, and feline species, as well as to identify which aspects need further optimization and, where possible, to provide guidance on future advancements. Following a "step-by-step" approach, we cover the generation of iPSCs in companion animals from the selection of somatic cells and the reprogramming strategies, to the expansion and characterization of iPSCs. Subsequently, we revise the current applications of iPSCs in companion animals, identify the main hurdles, and propose future paths to move the field forward. Transferring the knowledge gained from human iPSCs can increase our understanding in the biology of pluripotent cells in animals, but it is critical to further investigate the differences among species to develop specific approaches for animal iPSCs. This is key for significantly advancing iPSC application in veterinary medicine, which at the same time will also allow gaining pre-clinical knowledge transferable to human medicine.
Collapse
Affiliation(s)
| | | | | | | | - Frank Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, University of Galway, Galway, Ireland
| |
Collapse
|
13
|
Liang S, Su Y, Yao R. 3D Bioprinting of Induced Pluripotent Stem Cells and Disease Modeling. Handb Exp Pharmacol 2023; 281:29-56. [PMID: 36882603 DOI: 10.1007/164_2023_646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Patient-derived induced pluripotent stem cells (iPSCs), carrying the genetic information of the disease and capable of differentiating into multilineages in vitro, are valuable for disease modeling. 3D bioprinting enables the assembly of the cell-laden hydrogel into hierarchically three-dimensional architectures that recapitulate the natural tissues and organs. Investigation of iPSC-derived physiological and pathological models constructed by 3D bioprinting is a fast-growing field still in its infancy. Distinctly from cell lines and adult stem cells, iPSCs and iPSC-derived cells are more susceptible to external stimuli which can disturb the differentiation, maturation, and organization of iPSCs and their progeny. Here we discuss the fitness of iPSCs and 3D bioprinting from the perspective of bioinks and printing technologies. We provide a timely review of the progress of 3D bioprinting iPSC-derived physiological and pathological models by exemplifying the relatively prosperous cardiac and neurological fields. We also discuss scientific rigors and highlight the remaining issues to offer a guiding framework for bioprinting-assisted personalized medicine.
Collapse
Affiliation(s)
- Shaojun Liang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China
| | - Yijun Su
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China
| | - Rui Yao
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China.
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, China.
| |
Collapse
|
14
|
Xu J, Zhang M, Du W, Zhao J, Ling G, Zhang P. Chitosan-based high-strength supramolecular hydrogels for 3D bioprinting. Int J Biol Macromol 2022; 219:545-557. [PMID: 35907459 DOI: 10.1016/j.ijbiomac.2022.07.206] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 12/11/2022]
Abstract
The loss of tissues and organs is a major challenge for biomedicine, and the emerging 3D bioprinting technology has brought the dawn for the development of tissue engineering and regenerative medicine. Chitosan-based supramolecular hydrogels, as novel biomaterials, are considered as ideal materials for 3D bioprinting due to their unique dynamic reversibility and fantastic biological properties. Although chitosan-based supramolecular hydrogels have wonderful biological properties, the mechanical properties are still under early exploration. This paper aims to provide some inspirations for researchers to further explore. In this review, common 3D bioprinting techniques and the properties required for bioink for 3D bioprinting are firstly described. Then, several strategies to enhance the mechanical properties of chitosan hydrogels are introduced from the perspectives of both materials and supramolecular binding motifs. Finally, current challenges and future opportunities in this field are discussed. The combination of chitosan-based supramolecular hydrogels and 3D bioprinting will hold promise for developing novel biomedical implants.
Collapse
Affiliation(s)
- Jiaqi Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Manyue Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wenzhen Du
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Jiuhong Zhao
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Peng Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
15
|
Kahn-Krell A, Pretorius D, Guragain B, Lou X, Wei Y, Zhang J, Qiao A, Nakada Y, Kamp TJ, Ye L, Zhang J. A three-dimensional culture system for generating cardiac spheroids composed of cardiomyocytes, endothelial cells, smooth-muscle cells, and cardiac fibroblasts derived from human induced-pluripotent stem cells. Front Bioeng Biotechnol 2022; 10:908848. [PMID: 35957645 PMCID: PMC9361017 DOI: 10.3389/fbioe.2022.908848] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/04/2022] [Indexed: 01/22/2023] Open
Abstract
Cardiomyocytes (CMs), endothelial cells (ECs), smooth-muscle cells (SMCs), and cardiac fibroblasts (CFs) differentiated from human induced-pluripotent stem cells (hiPSCs) are the fundamental components of cell-based regenerative myocardial therapy and can be used as in-vitro models for mechanistic studies and drug testing. However, newly differentiated hiPSC-CMs tend to more closely resemble fetal CMs than the mature CMs of adult hearts, and current techniques for improving CM maturation can be both complex and labor-intensive. Thus, the production of CMs for commercial and industrial applications will require more elementary methods for promoting CM maturity. CMs tend to develop a more mature phenotype when cultured as spheroids in a three-dimensional (3D) environment, rather than as two-dimensional monolayers, and the activity of ECs, SMCs, and CFs promote both CM maturation and electrical activity. Here, we introduce a simple and reproducible 3D-culture-based process for generating spheroids containing all four cardiac-cell types (i.e., cardiac spheroids) that is compatible with a wide range of applications and research equipment. Subsequent experiments demonstrated that the inclusion of vascular cells and CFs was associated with an increase in spheroid size, a decline in apoptosis, an improvement in sarcomere maturation and a change in CM bioenergetics.
Collapse
Affiliation(s)
- Asher Kahn-Krell
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Danielle Pretorius
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bijay Guragain
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xi Lou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuhua Wei
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianhua Zhang
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Aijun Qiao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuji Nakada
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Timothy J. Kamp
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI, United States,Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Lei Ye
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States,Department of Medicine/Cardiovascular Diseases, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: Jianyi Zhang,
| |
Collapse
|
16
|
Wang J, Huang D, Yu H, Cheng Y, Ren H, Zhao Y. Developing tissue engineering strategies for liver regeneration. ENGINEERED REGENERATION 2022; 3:80-91. [DOI: 10.1016/j.engreg.2022.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
17
|
Li S, Yoshioka M, Li J, Liu L, Ye S, Kamei KI, Chen Y. Nanocasting of fibrous morphology on a substrate for long-term propagation of human induced pluripotent stem cells. Biomed Mater 2022; 17. [PMID: 35114658 DOI: 10.1088/1748-605x/ac51b8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/03/2022] [Indexed: 11/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSCs) can be self-renewed for many generations on nanofibrous substrates. Herein, a casting method is developed to replicate the nanofibrous morphology into a thin layer of polymethylsiloxane (PDMS). The template is obtained by electrospinning and chemical crosslinking of gelatin nanofibers on a glass slide. The replicas of the template are surface-functionalized by gelatin and used for propagation of hiPSCs over tenth generations. The performance of the propagated hiPSCs is checked by immunofluorescence imaging, flowcytometry, and RT-PCR, confirming the utility of the method. The results are also compared with those obtained using electrospun nanofiber substrates. Inherently, the PDMS replicas is of low stiffness and can be reproduced easily. Compared to other patterning techniques, casting is more flexible and cost-effective, suggesting that this method might find applications in cell-based assays that rely on stringent consideration of both substrate stiffness and surface morphology.
Collapse
Affiliation(s)
- Sisi Li
- Chemistry, Ecole Normale Superieure, 24 rue Lhomond, Paris, Île-de-France, 75230, FRANCE
| | - Momoko Yoshioka
- Kyoto University, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Junjun Li
- Institute for Integrated Cell-Material Sciences, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Li Liu
- Kyoto University, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Sixin Ye
- University of Paris, 94276 Le Kremlin Bicêtre, Paris, 75006, FRANCE
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Yong Chen
- Chemistry, Ecole Normale Superieure, 24 rue Lhomond, F-75231 Paris Cedex 05, Paris, Île-de-France, 75230, FRANCE
| |
Collapse
|
18
|
Song Y, Chen W, Gai K, Lin F, Sun W. Culture models produced via biomanufacturing for neural tissue-like constructs based on primary neural and neural stem cells. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
19
|
Hall GN, Tam WL, Andrikopoulos KS, Casas-Fraile L, Voyiatzis GA, Geris L, Luyten FP, Papantoniou I. Patterned, organoid-based cartilaginous implants exhibit zone specific functionality forming osteochondral-like tissues in vivo. Biomaterials 2021; 273:120820. [PMID: 33872857 DOI: 10.1016/j.biomaterials.2021.120820] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/16/2022]
Abstract
Tissue engineered constructs have the potential to respond to the unmet medical need of treating deep osteochondral defects. However, current tissue engineering strategies struggle in the attempt to create patterned constructs with biologically distinct functionality. In this work, a developmentally-inspired modular approach is proposed, whereby distinct cartilaginous organoids are used as living building blocks. First, a hierarchical construct was created, composed of three layers of cartilaginous tissue intermediates derived from human periosteum-derived cells: (i) early (SOX9), (ii) mature (COL2) and (iii) (pre)hypertrophic (IHH, COLX) phenotype. Subcutaneous implantation in nude mice generated a hybrid tissue containing one mineralized and one non-mineralized part. However, the non-mineralized part was represented by a collagen type I positive fibrocartilage-like tissue. To engineer a more stable articular cartilage part, iPSC-derived cartilage microtissues (SOX9, COL2; IHH neg) were generated. Subcutaneous implantation of assembled iPSC-derived cartilage microtissues resulted in a homogenous cartilaginous tissue positive for collagen type II but negative for osteocalcin. Finally, iPSC-derived cartilage microtissues in combination with the pre-hypertrophic cartilage organoids (IHH, COLX) could form dual tissues consisting of i) a cartilaginous safranin O positive and ii) a bony osteocalcin positive region upon subcutaneous implantation, corresponding to the pre-engineered zonal pattern. The assembly of functional building blocks, as presented in this work, opens possibilities for the production of complex tissue engineered implants by embedding zone-specific functionality through the use of pre-programmed living building blocks.
Collapse
Affiliation(s)
- Gabriella Nilsson Hall
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium; Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium
| | - Wai Long Tam
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium
| | - Konstantinos S Andrikopoulos
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas, Stadiou, 26504, Platani, Patras, Greece; Department of Physics, University of Patras, GR-265 00, Rio-Patras, Greece
| | - Leire Casas-Fraile
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, PB 813, Leuven, 3000, Belgium
| | - George A Voyiatzis
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas, Stadiou, 26504, Platani, Patras, Greece
| | - Liesbet Geris
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium; GIGA in Silico Medicine, Université de Liège, Avenue de L'Hôpital 11 - BAT 34, 4000, Liège 1, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C, PB 2419, 3001, Leuven, Belgium
| | - Frank P Luyten
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium; Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium.
| | - Ioannis Papantoniou
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium; Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium; Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas, Stadiou, 26504, Platani, Patras, Greece.
| |
Collapse
|
20
|
Yang H, Sun L, Pang Y, Hu D, Xu H, Mao S, Peng W, Wang Y, Xu Y, Zheng YC, Du S, Zhao H, Chi T, Lu X, Sang X, Zhong S, Wang X, Zhang H, Huang P, Sun W, Mao Y. Three-dimensional bioprinted hepatorganoids prolong survival of mice with liver failure. Gut 2021; 70:567-574. [PMID: 32434830 PMCID: PMC7873413 DOI: 10.1136/gutjnl-2019-319960] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 04/28/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Shortage of organ donors, a critical challenge for treatment of end-stage organ failure, has motivated the development of alternative strategies to generate organs in vitro. Here, we aim to describe the hepatorganoids, which is a liver tissue model generated by three-dimensional (3D) bioprinting of HepaRG cells and investigate its liver functions in vitro and in vivo. DESIGN 3D bioprinted hepatorganoids (3DP-HOs) were constructed using HepaRG cells and bioink, according to specific 3D printing procedures. Liver functions of 3DP-HOs were detected after 7 days of differentiation in vitro, which were later transplanted into Fah-deficient mice. The in vivo liver functions of 3DP-HOs were evaluated by survival time and liver damage of mice, human liver function markers and human-specific debrisoquine metabolite production. RESULTS 3DP-HOs broadly acquired liver functions, such as ALBUMIN secretion, drug metabolism and glycogen storage after 7 days of differentiation. After transplantation into abdominal cavity of Fah-/-Rag2-/- mouse model of liver injury, 3DP-HOs further matured and displayed increased synthesis of liver-specific proteins. Particularly, the mice acquired human-specific drug metabolism activities. Functional vascular systems were also formed in transplanted 3DP-HOs, further enhancing the material transport and liver functions of 3DP-HOs. Most importantly, transplantation of 3DP-HOs significantly improved the survival of mice. CONCLUSIONS Our results demonstrated a comprehensive proof of principle, which indicated that 3DP-HO model of liver tissues possessed in vivo hepatic functions and alleviated liver failure after transplantation, suggesting that 3D bioprinting could be used to generate human liver tissues as the alternative transplantation donors for treatment of liver diseases.
Collapse
Affiliation(s)
- Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Lejia Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, China,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, China,Overseas Expertise Introduction Center for Discipline Innovation, Tsinghua University, Beijing, China
| | - Dandan Hu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China,Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Shuangshuang Mao
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, China,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, China,Overseas Expertise Introduction Center for Discipline Innovation, Tsinghua University, Beijing, China
| | - Wenbo Peng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yanan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yiyao Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Yong-Chang Zheng
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Tianyi Chi
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Shouxian Zhong
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Wang
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, Inner Mongolia, China,Hepatoscience Section, Cell Lab Tech Inc, Sunnyvale, California, USA,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Pengyu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China .,Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, China .,Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, China.,Overseas Expertise Introduction Center for Discipline Innovation, Tsinghua University, Beijing, China.,Department of Mechanical Engineering, Drexel University, Philadelphia, Pennsylvania, USA
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Induced Pluripotency: A Powerful Tool for In Vitro Modeling. Int J Mol Sci 2020; 21:ijms21238910. [PMID: 33255453 PMCID: PMC7727808 DOI: 10.3390/ijms21238910] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
One of the greatest breakthroughs of regenerative medicine in this century was the discovery of induced pluripotent stem cell (iPSC) technology in 2006 by Shinya Yamanaka. iPSCs originate from terminally differentiated somatic cells that have newly acquired the developmental capacity of self-renewal and differentiation into any cells of three germ layers. Before iPSCs can be used routinely in clinical practice, their efficacy and safety need to be rigorously tested; however, iPSCs have already become effective and fully-fledged tools for application under in vitro conditions. They are currently routinely used for disease modeling, preparation of difficult-to-access cell lines, monitoring of cellular mechanisms in micro- or macroscopic scales, drug testing and screening, genetic engineering, and many other applications. This review is a brief summary of the reprogramming process and subsequent differentiation and culture of reprogrammed cells into neural precursor cells (NPCs) in two-dimensional (2D) and three-dimensional (3D) conditions. NPCs can be used as biomedical models for neurodegenerative diseases (NDs), which are currently considered to be one of the major health problems in the human population.
Collapse
|
22
|
Ghaemi RV, Siang LC, Yadav VG. Improving the Rate of Translation of Tissue Engineering Products. Adv Healthc Mater 2019; 8:e1900538. [PMID: 31386306 DOI: 10.1002/adhm.201900538] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/04/2019] [Indexed: 12/18/2022]
Abstract
Over 100 000 research articles and 9000 patents have been published on tissue engineering (TE) in the past 20 years. Yet, very few TE products have made their way to the market during the same period. Experts have proposed a variety of strategies to address the lack of translation of TE products. However, since these proposals are guided by qualitative insights, they are limited in scope and impact. Machine learning is utilized in the current study to analyze the entire body of patents that have been published over the past twenty years and understand patenting trends, topics, areas of application, and exemplifications. This analysis yields surprising and little-known insights about the differences in research priorities and perceptions of innovativeness of tissue engineers in academia and industry, as well as aids to chart true advances in the field during the past twenty years. It is hoped that this analysis and subsequent proposal to improve translational rates of TE products will spur much needed dialogue about this important pursuit.
Collapse
Affiliation(s)
- Roza Vaez Ghaemi
- Department of Chemical and Biological Engineeringand School of Biomedical EngineeringThe University of British Columbia Vancouver V6T 1Z3 Canada
| | - Lim C. Siang
- Department of Chemical and Biological Engineeringand School of Biomedical EngineeringThe University of British Columbia Vancouver V6T 1Z3 Canada
| | - Vikramaditya G. Yadav
- Department of Chemical and Biological Engineeringand School of Biomedical EngineeringThe University of British Columbia Vancouver V6T 1Z3 Canada
| |
Collapse
|
23
|
Han U, Kim YJ, Kim W, Park JH, Hong J. Construction of nano-scale cellular environments by coating a multilayer nanofilm on the surface of human induced pluripotent stem cells. NANOSCALE 2019; 11:13541-13551. [PMID: 31290516 DOI: 10.1039/c9nr02375e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Interactions with peripheral environments, such as extracellular matrix (ECM) and other cells, and their balance play a crucial role in the maintenance of pluripotency and self-renewal of human pluripotent stem cells. In this study, we focused on a nano-sized artificial cellular environment that is directly attached to the cytoplasmic membrane as a facile method that can effect intercellular interactions at the single-cell level. We designed multilayered nanofilms that are self-assembled on the surface of human induced pluripotent stem cells (iPSCs), by repetitive adsorption of fibronectin and heparin or chondroitin sulfate. However, the surface modification process could also lead to the loss of cell-cell adhesion, which may result in apoptotic cell death. We investigated the proliferation and pluripotency of the iPSCs coated with the nanofilm in order to establish the suitable nanofilm structure and coating conditions. As a result, the cell viability reduced with the increase in the duration of the coating process, but the undifferentiated state and proliferation of the cells were maintained until 2 bilayers were coated. To suppress the dissociation-induced apoptosis, Y-27632, the Rho-associated kinase inhibitor (ROCKi), was added to the coating solution; this allowed the coating of up to 4 bilayers of the nanofilm onto the iPSCs. These results are expected to accelerate the pace of iPSC studies on 3-dimensional cultures and naïve pluripotency, in which the regulation of cellular interactions plays a critical role.
Collapse
Affiliation(s)
- Uiyoung Han
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Yu Jin Kim
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea.
| | - Wijin Kim
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea.
| | - Ju Hyun Park
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea.
| | - Jinkee Hong
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
24
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|
25
|
Intestinal organoids: A new paradigm for engineering intestinal epithelium in vitro. Biomaterials 2019; 194:195-214. [DOI: 10.1016/j.biomaterials.2018.12.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/22/2018] [Accepted: 12/08/2018] [Indexed: 12/11/2022]
|
26
|
Laminin as a Potent Substrate for Large-Scale Expansion of Human Induced Pluripotent Stem Cells in a Closed Cell Expansion System. Stem Cells Int 2019; 2019:9704945. [PMID: 30805013 PMCID: PMC6362483 DOI: 10.1155/2019/9704945] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/28/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022] Open
Abstract
The number of high-quality cells required for engineering an adult human-sized bioartificial organ is greater than one billion. Until the emergence of induced pluripotent stem cells (iPSCs), autologous cell sources of this magnitude and with the required complexity were not available. Growing this number of cells in a traditional 2D cell culture system requires extensive time, resources, and effort and does not always meet clinical requirements. The use of a closed cell culture system is an efficient and clinically applicable method that can be used to expand cells under controlled conditions. We aimed to use the Quantum Cell Expansion System (QES) as an iPSC monolayer-based expansion system. Human iPSCs were expanded (up to 14-fold) using the QES on two different coatings (laminin 521 (LN521) and vitronectin (VN)), and a karyotype analysis was performed. The cells were characterized for spontaneous differentiation and pluripotency by RT-PCR and flow cytometry. Our results demonstrated that the QES provides the necessary environment for exponential iPSC growth, reaching 689.75 × 106 ± 86.88 × 106 in less than 7 days using the LN521 coating with a population doubling level of 3.80 ± 0.19. The same result was not observed when VN was used as a coating. The cells maintained normal karyotype (46-XX), expressed pluripotency markers (OCT4, NANOG, LIN28, SOX2, REX1, DPPA4, NODAL, TDGFb, TERT3, and GDF), and expressed high levels of OCT4, SOX2, NANOG, SSEA4, TRA1-60, and TRA1-81. Spontaneous differentiation into ectoderm (NESTIN, TUBB3, and NEFH), mesoderm (MSX1, BMP4, and T), and endoderm (GATA6, AFP, and SOX17) lineages was detected by RT-PCR with both coating systems. We conclude that the QES maintains the stemness of iPSCs and is a promising platform to provide the number of cells necessary to recellularize small human-sized organ scaffolds for clinical purposes.
Collapse
|
27
|
Abelseth E, Abelseth L, De la Vega L, Beyer ST, Wadsworth SJ, Willerth SM. 3D Printing of Neural Tissues Derived from Human Induced Pluripotent Stem Cells Using a Fibrin-Based Bioink. ACS Biomater Sci Eng 2018; 5:234-243. [DOI: 10.1021/acsbiomaterials.8b01235] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | | | - Simon T. Beyer
- Aspect Biosystems, 1781 W 75th Avenue, Vancouver, British Columbia V6P 6P2, Canada
| | - Samuel J. Wadsworth
- Aspect Biosystems, 1781 W 75th Avenue, Vancouver, British Columbia V6P 6P2, Canada
| | | |
Collapse
|
28
|
Li Y, Jiang X, Li L, Chen ZN, Gao G, Yao R, Sun W. 3D printing human induced pluripotent stem cells with novel hydroxypropyl chitin bioink: scalable expansion and uniform aggregation. Biofabrication 2018; 10:044101. [PMID: 29952313 DOI: 10.1088/1758-5090/aacfc3] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) are more likely to successfully avoid the immunological rejection and ethical problems that are often encountered by human embryonic stem cells in various stem cell studies and applications. To transfer hiPSCs from the laboratory to clinical applications, researchers must obtain sufficient cell numbers. In this study, 3D cell printing was used as a novel method for iPSC scalable expansion. Hydroxypropyl chitin (HPCH), utilized as a new type of bioink, and a set of optimized printing parameters were shown to achieve high cell survival (>90%) after the printing process and high proliferation efficiency (∼32.3 folds) during subsequent 10 d culture. After the culture, high levels of pluripotency maintenance were recognized by both qualitative and quantitative detections. Compared with static suspension culture, hiPSC aggregates formed in 3D-printed constructs showed a higher uniformity in size. Using a novel dual-fluorescent labeling method, hiPSC aggregates in the constructs were found more inclined to form by in situ proliferation rather than multicellular aggregation. This study revealed unique advantages of non-ionic crosslinking bioink material HPCH, including high gel strength and rapid temperature response in hiPSC printing, and achieved primed state hiPSC printing for the first time. Features achieved in this study, such as high cell yield, high pluripotency maintenance and uniform aggregation provide good foundations for further hiPSC studies on 3D micro-tissue differentiation and drug screening.
Collapse
Affiliation(s)
- Yang Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China. 111 'Biomanufacturing and Engineering Living Systems' Innovation International Talents Base, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
29
|
Cui H, Miao S, Esworthy T, Zhou X, Lee SJ, Liu C, Yu ZX, Fisher JP, Mohiuddin M, Zhang LG. 3D bioprinting for cardiovascular regeneration and pharmacology. Adv Drug Deliv Rev 2018; 132:252-269. [PMID: 30053441 PMCID: PMC6226324 DOI: 10.1016/j.addr.2018.07.014] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/22/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Compared to traditional therapeutic strategies, three-dimensional (3D) bioprinting is one of the most advanced techniques for creating complicated cardiovascular implants with biomimetic features, which are capable of recapitulating both the native physiochemical and biomechanical characteristics of the cardiovascular system. The present review provides an overview of the cardiovascular system, as well as describes the principles of, and recent advances in, 3D bioprinting cardiovascular tissues and models. Moreover, this review will focus on the applications of 3D bioprinting technology in cardiovascular repair/regeneration and pharmacological modeling, further discussing current challenges and perspectives.
Collapse
Affiliation(s)
- Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Shida Miao
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Chengyu Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, USA
| | | | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
30
|
Madl CM, Heilshorn SC, Blau HM. Bioengineering strategies to accelerate stem cell therapeutics. Nature 2018; 557:335-342. [PMID: 29769665 PMCID: PMC6773426 DOI: 10.1038/s41586-018-0089-z] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023]
Abstract
Although only a few stem cell-based therapies are currently available to patients, stem cells hold tremendous regenerative potential, and several exciting clinical applications are on the horizon. Biomaterials with tuneable mechanical and biochemical properties can preserve stem cell function in culture, enhance survival of transplanted cells and guide tissue regeneration. Rapid progress with three-dimensional hydrogel culture platforms provides the opportunity to grow patient-specific organoids, and has led to the discovery of drugs that stimulate endogenous tissue-specific stem cells and enabled screens for drugs to treat disease. Therefore, bioengineering technologies are poised to overcome current bottlenecks and revolutionize the field of regenerative medicine.
Collapse
Affiliation(s)
- Christopher M Madl
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
31
|
Muncie JM, Weaver VM. The Physical and Biochemical Properties of the Extracellular Matrix Regulate Cell Fate. Curr Top Dev Biol 2018; 130:1-37. [PMID: 29853174 DOI: 10.1016/bs.ctdb.2018.02.002] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The extracellular matrix is a complex network of hydrated macromolecular proteins and sugars that, in concert with bound soluble factors, comprise the acellular stromal microenvironment of tissues. Rather than merely providing structural information to cells, the extracellular matrix plays an instructive role in development and is critical for the maintenance of tissue homeostasis. In this chapter, we review the composition of the extracellular matrix and summarize data illustrating its importance in embryogenesis, tissue-specific development, and stem cell differentiation. We discuss how the biophysical and biochemical properties of the extracellular matrix ligate specific transmembrane receptors to activate intracellular signaling that alter cell shape and cytoskeletal dynamics to modulate cell growth and viability, and direct cell migration and cell fate. We present examples describing how the extracellular matrix functions as a highly complex physical and chemical entity that regulates tissue organization and cell behavior through a dynamic and reciprocal dialogue with the cellular constituents of the tissue. We suggest that the extracellular matrix not only transmits cellular and tissue-level force to shape development and tune cellular activities that are key for coordinated tissue behavior, but that it is itself remodeled such that it temporally evolves to maintain the integrated function of the tissue. Accordingly, we argue that perturbations in extracellular matrix composition and structure compromise key developmental events and tissue homeostasis, and promote disease.
Collapse
Affiliation(s)
- Jonathon M Muncie
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA, United States; Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, United States
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|