1
|
Johnston J, Jeon H, Choi YY, Kim G, Shi T, Khong C, Chang HC, Myung NV, Wang Y. Stimulative piezoelectric nanofibrous scaffolds for enhanced small extracellular vesicle production in 3D cultures. Biomater Sci 2024; 12:5728-5741. [PMID: 39403853 PMCID: PMC11474809 DOI: 10.1039/d4bm00504j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Small extracellular vesicles (sEVs) have great promise as effective carriers for drug delivery. However, the challenges associated with the efficient production of sEVs hinder their clinical applications. Herein, we report a stimulative 3D culture platform for enhanced sEV production. The proposed platform consists of a piezoelectric nanofibrous scaffold (PES) coupled with acoustic stimulation to enhance sEV production of cells in a 3D biomimetic microenvironment. Combining cell stimulation with a 3D culture platform in this stimulative PES enables a 15.7-fold increase in the production rate per cell with minimal deviations in particle size and protein composition compared with standard 2D cultures. We find that the enhanced sEV production is attributable to the activation and upregulation of crucial sEV production steps through the synergistic effect of stimulation and the 3D microenvironment. Moreover, changes in cell morphology lead to cytoskeleton redistribution through cell-matrix interactions in the 3D cultures. This in turn facilitates intracellular EV trafficking, which impacts the production rate. Overall, our work provides a promising 3D cell culture platform based on piezoelectric biomaterials for enhanced sEV production. This platform is expected to accelerate the potential use of sEVs for drug delivery and broad biomedical applications.
Collapse
Affiliation(s)
- James Johnston
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Hyunsu Jeon
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Yun Young Choi
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Gaeun Kim
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Tiger Shi
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Courtney Khong
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Nosang Vincent Myung
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Yichun Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
2
|
Johnston J, Jeon H, Choi YY, Kim G, Shi T, Khong C, Chang HC, Myung NV, Wang Y. Stimulative piezoelectric nanofibrous scaffolds for enhanced small extracellular vesicle production in 3D cultures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589114. [PMID: 38659930 PMCID: PMC11042190 DOI: 10.1101/2024.04.12.589114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Small extracellular vesicles (sEVs) have great promise as effective carriers for drug delivery. However, the challenges associated with the efficient production of sEVs hinder their clinical applications. Herein, we report a stimulative 3D culture platform for enhanced sEV production. The proposed platform consists of a piezoelectric nanofibrous scaffold (PES) coupled with acoustic stimulation to enhance sEV production of cells in a 3D biomimetic microenvironment. Combining cell stimulation with a 3D culture platform in this stimulative PES enables a 15.7-fold increase in the production rate per cell with minimal deviations in particle size and protein composition compared with standard 2D cultures. We find that the enhanced sEV production is attributable to the activation and upregulation of crucial sEV production steps through the synergistic effect of stimulation and the 3D microenvironment. Moreover, changes in cell morphology lead to cytoskeleton redistribution through cell-matrix interactions in the 3D cultures. This in turn facilitates intracellular EV trafficking, which impacts the production rate. Overall, our work provides a promising 3D cell culture platform based on piezoelectric biomaterials for enhanced sEV production. This platform is expected to accelerate the potential use of sEVs for drug delivery and broad biomedical applications.
Collapse
|
3
|
Tokuoka Y, Ishida T. Local Microbubble Removal in Polydimethylsiloxane Microchannel by Balancing Negative and Atmospheric Pressures. MICROMACHINES 2023; 15:37. [PMID: 38258156 PMCID: PMC10819605 DOI: 10.3390/mi15010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024]
Abstract
Long-term experiments using organoids and tissues are crucial for drug development. Microfluidic devices have been regularly used in long-term experiments. However, microbubbles often form in these devices, and they may damage and starve cells. A method involving the application of negative pressure has been reported to remove microbubbles from microfluidic devices composed of polydimethylsiloxane; however, negative pressure affects the cells and tissues in microfluidic devices. In this study, a local microbubble removal method was developed using a microfluidic device with 0.5 mm thin polydimethylsiloxane sidewalls. The thin sidewalls counterbalanced the negative and atmospheric pressures, thereby localizing the negative pressure near the negatively pressurized chamber. Microbubbles were removed within 5 mm of the negatively pressurized chamber; however, those in an area 7 mm and more from the chamber were not removed. Using the local removal method, a long-term perfusion test was performed, and no contact was confirmed between the bubbles and the simulated tissue for 72 h.
Collapse
Affiliation(s)
- Yasunori Tokuoka
- Department of Mechanical Engineering, School of Engineering, Institute of Technology, Tokyo 226-8503, Japan
| | - Tadashi Ishida
- Department of Mechanical Engineering, School of Engineering, Institute of Technology, Tokyo 226-8503, Japan
| |
Collapse
|
4
|
Tian H, Ren J, Mou R, Jia Y. Application of organoids in precision immunotherapy of lung cancer (Review). Oncol Lett 2023; 26:484. [PMID: 37818130 PMCID: PMC10561155 DOI: 10.3892/ol.2023.14071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/18/2023] [Indexed: 10/12/2023] Open
Abstract
In immunotherapy, the immune system is modulated in order to treat cancer. Traditional two dimensional in vitro models and in vivo animal models are insufficient to simulate the complex tumor microenvironment (TME) in the original tumor. As tumor immunotherapy involves the immune system, additional tumor mimic models, such as patient-derived organoids, are required for the evaluation of the efficacy of immunotherapy. Furthermore, non-tumor components and host tumor cells in the TME may interact to promote cancer incidence, progression, drug resistance and metastasis. It is possible to produce organoid models for lung cancer by retaining endogenous stromal components (e.g., multiple immune cell types), supplying cancer-associated fibroblasts and exogenous immune cells, constructing tumor vasculature and adding other biological or chemical components that emulate the TME. Therefore, the lung cancer organoid culture platform may facilitate preclinical testing of immunotherapy drugs for lung cancer by mimicking immunotherapy responses. The present review summarizes current lung cancer organoid culture methods for TME modeling and discusses the use of lung cancer-derived organoids for the detection of lung cancer immunotherapy and individualized cancer immunotherapy.
Collapse
Affiliation(s)
- Huichuan Tian
- Department of Medical Oncology, The First Teaching Hospital of Tianjin University of Chinese Medicine, Tianjin 300381, P.R. China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin 300381, P.R. China
| | - Jiajun Ren
- Department of Medical Oncology, The First Teaching Hospital of Tianjin University of Chinese Medicine, Tianjin 300381, P.R. China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin 300381, P.R. China
| | - Ruiyu Mou
- Department of Medical Oncology, The First Teaching Hospital of Tianjin University of Chinese Medicine, Tianjin 300381, P.R. China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin 300381, P.R. China
| | - Yingjie Jia
- Department of Medical Oncology, The First Teaching Hospital of Tianjin University of Chinese Medicine, Tianjin 300381, P.R. China
- National Clinical Research Center of Chinese Acupuncture and Moxibustion, Tianjin 300381, P.R. China
| |
Collapse
|
5
|
Guan D, Liu X, Shi Q, He B, Zheng C, Meng X. Breast cancer organoids and their applications for precision cancer immunotherapy. World J Surg Oncol 2023; 21:343. [PMID: 37884976 PMCID: PMC10601270 DOI: 10.1186/s12957-023-03231-2] [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: 06/20/2023] [Accepted: 10/14/2023] [Indexed: 10/28/2023] Open
Abstract
Immunotherapy is garnering increasing attention as a therapeutic strategy for breast cancer (BC); however, the application of precise immunotherapy in BC has not been fully studied. Further studies on BC immunotherapy have a growing demand for preclinical models that reliably recapitulate the composition and function of the tumor microenvironment (TME) of BC. However, the classic two-dimensional in vitro and animal in vivo models inadequately recapitulate the intricate TME of the original tumor. Organoid models which allow the regular culture of primitive human tumor tissue are increasingly reported that they can incorporate immune components. Therefore, organoid platforms can be used to replicate the BC-TME to achieve the immunotherapeutic reaction modeling and facilitate relevant preclinical trial. In this study, we have investigated different organoid culture methods for BC-TME modeling and their applications for precision immunotherapy in BC.
Collapse
Affiliation(s)
- Dandan Guan
- College of Medicine, Soochow University, Soochow, China
- General Surgery, Department of Breast Surgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema of Breast Cancer, Hangzhou, Zhejiang, China
| | - Xiaozhen Liu
- General Surgery, Department of Breast Surgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema of Breast Cancer, Hangzhou, Zhejiang, China
- Key Laboratory for Diagnosis and Treatment of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Qingyang Shi
- Department of Urology, Haining Central Hospital, Haining Branch of Zhejiang Provincial People's Hospital, Jiaxing, Zhejiang, China
| | - Bangjie He
- Department of General Surgery, Traditional Chinese Medicine Hospital of Zhuji, Zhuji, Zhejiang, China
| | - Chaopeng Zheng
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xuli Meng
- General Surgery, Department of Breast Surgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Key Laboratory for Diagnosis and Treatment of Upper Limb Edema of Breast Cancer, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Zhang R, Feng C, Luo D, Zhao R, Kannan PR, Yin Y, Iqbal MZ, Hu Y, Kong X. Metformin Hydrochloride Significantly Inhibits Rotavirus Infection in Caco2 Cell Line, Intestinal Organoids, and Mice. Pharmaceuticals (Basel) 2023; 16:1279. [PMID: 37765086 PMCID: PMC10536476 DOI: 10.3390/ph16091279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Rotavirus is one of the main pathogens that causes severe diarrhea in children under the age of 5, primarily infecting the enterocytes of the small intestine. Currently, there are no specific drugs available for oral rehydration and antiviral therapy targeting rotavirus. However, metformin hydrochloride, a drug known for its antiviral properties, shows promise as it accumulates in the small intestine and modulates the intestinal microbiota. Therefore, we formulated a hypothesis that metformin hydrochloride could inhibit rotavirus replication in the intestine. To validate the anti-rotavirus effect of metformin hydrochloride, we conducted infection experiments using different models, ranging from in vitro cells and organoids to small intestines in vivo. The findings indicate that a concentration of 0.5 mM metformin hydrochloride significantly inhibits the expression of rotavirus mRNA and protein in Caco-2 cells, small intestinal organoids, and suckling mice models. Rotavirus infections lead to noticeable pathological changes, but treatment with metformin has been observed to mitigate the lesions caused by rotavirus infection in the treated group. Our study establishes that metformin hydrochloride can inhibit rotavirus replication, while also affirming the reliability of organoids as a virus model for in vitro research.
Collapse
Affiliation(s)
- Rui Zhang
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Cui Feng
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Dandan Luo
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Ruibo Zhao
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Perumal Ramesh Kannan
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yuebang Yin
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Muhammad Zubair Iqbal
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yeting Hu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310030, China
| | - Xiangdong Kong
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.Z.); (C.F.); (D.L.); (R.Z.); (P.R.K.); (Y.Y.); (M.Z.I.)
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
7
|
SFRP4 + stromal cell subpopulation with IGF1 signaling in human endometrial regeneration. Cell Discov 2022; 8:95. [PMID: 36163341 PMCID: PMC9512788 DOI: 10.1038/s41421-022-00438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/17/2022] [Indexed: 11/08/2022] Open
Abstract
Our understanding of full-thickness endometrial regeneration after injury is limited by an incomplete molecular characterization of the cell populations responsible for the organ functions. To help fill this knowledge gap, we characterized 10,551 cells of full-thickness normal human uterine from two menstrual phases (proliferative and secretory phase) using unbiased single cell RNA-sequencing. We dissected cell heterogeneity of main cell types (epithelial, stromal, endothelial, and immune cells) of the full thickness uterine tissues, cell population architectures of human uterus cells across the menstrual cycle. We identified an SFRP4+ stromal cell subpopulation that was highly enriched in the regenerative stage of the human endometria during the menstrual cycle, and the SFRP4+ stromal cells could significantly enhance the proliferation of human endometrial epithelial organoid in vitro, and promote the regeneration of endometrial epithelial glands and full-thickness endometrial injury through IGF1 signaling pathway in vivo. Our cell atlas of full-thickness uterine tissues revealed the cellular heterogeneities, cell population architectures, and their cell-cell communications during the monthly regeneration of the human endometria, which provide insight into the biology of human endometrial regeneration and the development of regenerative medicine treatments against endometrial damage and intrauterine adhesion.
Collapse
|
8
|
|
9
|
The Effects of Transforming Growth Factor-β1 on the Differentiation of Cell Organoids Composed of Gingiva-Derived Stem Cells. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9818299. [PMID: 35872843 PMCID: PMC9303143 DOI: 10.1155/2022/9818299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022]
Abstract
This study was aimed at evaluating the effects of transforming growth factor-β on the differentiation and mRNA expression of organoids made out of human mesenchymal stem cells. Cell organoids composed of gingiva-derived stem cells were cultured in the presence of transforming growth factor-β1 at concentrations ranging from 0, 1, 10, to 20 ng/ml. Evaluations of the cell morphology of the organoids were performed on days 7, 9, 11, and 14. Quantitative cellular viability was completed on day 14. Alkaline phosphatase activity assays were performed to evaluate the differentiation of stem cells on day 14. Real-time polymerase chain reactions were used to determine the expression levels of TGF-β1, RUNX2, OCN, SOX9, and COL1A1 mRNA on day 14. The stem cells produced well-formed organoids on day 7, and the addition of transforming growth factor-β1 did not result in relevant changes in their shape. The organoids grew in size and became more intact with longer incubation times. On day 14, the diameters were 222.2 ± 9.6, 186.1 ± 4.8, 197.2 ± 9.6, and 211.1 ± 19.2 m for transforming growth factor-β1 at final concentrations of 0, 1, 10, and 20 ng/ml, respectively. Quantitative cell viability results from day 14 exhibited no significant difference between the groups (P > 0.05). There was significantly higher alkaline phosphatase activity with the addition of transforming growth factor-β1 with the highest value for the 1 ng/ml group (P < 0.05). Real-time polymerase chain reaction results demonstrated that the mRNA expression levels of RUNX2, OCN, and SOX were higher in 1 ng/ml but did not reach statistical significance. Treatment with 1 ng/ml of transforming growth factor-β1 significantly increased COL1A1 mRNA expression at day 14. The application of transforming growth factor-β1 increased differentiation, which was confirmed by alkaline phosphatase activity and mRNA expression while maintaining cell viability.
Collapse
|
10
|
Lu D, Yang Y, Zhang P, Ma Z, Li W, Song Y, Feng H, Yu W, Ren F, Li T, Zeng H, Wang J. Development and Application of Three-Dimensional Bioprinting Scaffold in the Repair of Spinal Cord Injury. Tissue Eng Regen Med 2022; 19:1113-1127. [PMID: 35767151 DOI: 10.1007/s13770-022-00465-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 01/04/2023] Open
Abstract
Spinal cord injury (SCI) is a disabling and destructive central nervous system injury that has not yet been successfully treated at this stage. Three-dimensional (3D) bioprinting has become a promising method to produce more biologically complex microstructures, which fabricate living neural constructs with anatomically accurate complex geometries and spatial distributions of neural stem cells, and this is critical in the treatment of SCI. With the development of 3D printing technology and the deepening of research, neural tissue engineering research using different printing methods, bio-inks, and cells to repair SCI has achieved certain results. Although satisfactory results have not yet been achieved, they have provided novel ideas for the clinical treatment of SCI. Considering the potential impact of 3D bioprinting technology on neural studies, this review focuses on 3D bioprinting methods widely used in SCI neural tissue engineering, and the latest technological applications of bioprinting of nerve tissues for the repair of SCI are discussed. In addition to introducing the recent progress, this work also describes the existing limitations and highlights emerging possibilities and future prospects in this field.
Collapse
Affiliation(s)
- Dezhi Lu
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yang Yang
- Department of Rehabilitation Medicine, Shandong Provincial Third Hospital, Shandong, 250000, China
| | - Pingping Zhang
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yan Song
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Haiyang Feng
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Wenqiang Yu
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Fuchao Ren
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Tao Li
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| | - Hong Zeng
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
11
|
Mei J, Vasan A, Magaram U, Takemura K, Chalasani SH, Friend J. Well-free agglomeration and on-demand three-dimensional cell cluster formation using guided surface acoustic waves through a couplant layer. Biomed Microdevices 2022; 24:18. [PMID: 35596837 PMCID: PMC9124176 DOI: 10.1007/s10544-022-00617-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2022] [Indexed: 11/24/2022]
Abstract
Three-dimensional cell agglomerates are broadly useful in tissue engineering and drug testing. We report a well-free method to form large (1.4-mm) multicellular clusters using 100-MHz surface acoustic waves (SAW) without direct contact with the media or cells. A fluid couplant is used to transform the SAW into acoustic streaming in the cell-laden media held in a petri dish. The couplant transmits longitudinal sound waves, forming a Lamb wave in the petri dish that, in turn, produces longitudinal sound in the media. Due to recirculation, human embryonic kidney (HEK293) cells in the dish are carried to the center of the coupling location, forming a cluster in less than 10 min. A few minutes later, these clusters may then be translated and merged to form large agglomerations, and even repeatedly folded to produce a roughly spherical shape of over 1.4 mm in diameter for incubation-without damaging the existing intercellular bonds. Calcium ion signaling through these clusters and confocal images of multiprotein junctional complexes suggest a continuous tissue construct: intercellular communication. They may be formed at will, and the method is feasibly useful for formation of numerous agglomerates in a single petri dish.
Collapse
Affiliation(s)
- Jiyang Mei
- Medically Advanced Devices Laboratory, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Dr MC0411, La Jolla, San Diego, CA, 92093, USA
| | - Aditya Vasan
- Medically Advanced Devices Laboratory, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Dr MC0411, La Jolla, San Diego, CA, 92093, USA
| | - Uri Magaram
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, San Diego, CA, 92037, USA
| | - Kenjiro Takemura
- Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kouhoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Sreekanth H Chalasani
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, San Diego, CA, 92037, USA
| | - James Friend
- Medically Advanced Devices Laboratory, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Dr MC0411, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
12
|
Mahapatra C, Lee R, Paul MK. Emerging role and promise of nanomaterials in organoid research. Drug Discov Today 2021; 27:890-899. [PMID: 34774765 DOI: 10.1016/j.drudis.2021.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/27/2021] [Accepted: 11/04/2021] [Indexed: 12/30/2022]
Abstract
Organoids are 3D stem cell-derived self-organization of cells. Organoid bioengineering helps recreate and tailor their architecture in vitro to generate mini organ-like properties, providing the opportunity to study fundamental cell behavior in heterogeneous populations and as a tool to model various diseases. Nanomaterials (NMs) are becoming indispensable in regenerative medicine and in developing treatment modalities for various diseases. Therefore, organoid-NM interactions are set to gain traction for the development of advanced diagnostics and therapeutics. Here, we discuss the interactions of NMs with distinctive organoid types, organoid matrices, trafficking and cargo delivery, organs-on-a-chip, bioprinting, downstream therapeutic implications, and future approaches.
Collapse
Affiliation(s)
- Chinmaya Mahapatra
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh 492010, India
| | - Ruda Lee
- International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, Kumamoto 860-8555, Japan
| | - Manash K Paul
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| |
Collapse
|
13
|
Grönholm M, Feodoroff M, Antignani G, Martins B, Hamdan F, Cerullo V. Patient-Derived Organoids for Precision Cancer Immunotherapy. Cancer Res 2021; 81:3149-3155. [PMID: 33687948 PMCID: PMC7616950 DOI: 10.1158/0008-5472.can-20-4026] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/29/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy has revolutionized the way tumors are treated. Nevertheless, efficient and robust testing platforms are still missing, including clinically relevant human ex vivo tumor assays that allow pretreatment testing of cancer therapies and selection of the most efficient and safe therapy for a specific patient. In the case of immunotherapy, this testing platform would require not only cancer cells, but also the tumor microenvironment, including immune cells. Here, we discuss the applications of patient-derived tumor organoid cultures and the possibilities in using complex immune-organoid cultures to provide preclinical testing platforms for precision cancer immunotherapy.
Collapse
Affiliation(s)
- Mikaela Grönholm
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Michaela Feodoroff
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- FIMM, Institute for Molecular Medicine Finland, Helsinki Institute for Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Gabriella Antignani
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Beatriz Martins
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Firas Hamdan
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Medical Biotechnology and CEINGE, Naples University Federico II, Naples, Italy
| |
Collapse
|
14
|
Zamproni LN, Mundim MTVV, Porcionatto MA. Neurorepair and Regeneration of the Brain: A Decade of Bioscaffolds and Engineered Microtissue. Front Cell Dev Biol 2021; 9:649891. [PMID: 33898443 PMCID: PMC8058361 DOI: 10.3389/fcell.2021.649891] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/12/2021] [Indexed: 01/24/2023] Open
Abstract
Repairing the human brain remains a challenge, despite the advances in the knowledge of inflammatory response to injuries and the discovery of adult neurogenesis. After brain injury, the hostile microenvironment and the lack of structural support for neural cell repopulation, anchoring, and synapse formation reduce successful repair chances. In the past decade, we witnessed the rise of studies regarding bioscaffolds’ use as support for neuro repair. A variety of natural and synthetic materials is available and have been used to replace damaged tissue. Bioscaffolds can assume different shapes and may or may not carry a diversity of content, such as stem cells, growth factors, exosomes, and si/miRNA that promote specific therapeutic effects and stimulate brain repair. The use of these external bioscaffolds and the creation of cell platforms provide the basis for tissue engineering. More recently, researchers were able to engineer brain organoids, neural networks, and even 3D printed neural tissue. The challenge in neural tissue engineering remains in the fabrication of scaffolds with precisely controlled topography and biochemical cues capable of directing and controlling neuronal cell fate. The purpose of this review is to highlight the existing research in the growing field of bioscaffolds’ development and neural tissue engineering. Moreover, this review also draws attention to emerging possibilities and prospects in this field.
Collapse
Affiliation(s)
- Laura N Zamproni
- Molecular Neurobiology Laboratory, Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mayara T V V Mundim
- Molecular Neurobiology Laboratory, Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marimelia A Porcionatto
- Molecular Neurobiology Laboratory, Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Campostrini G, Meraviglia V, Giacomelli E, van Helden RW, Yiangou L, Davis RP, Bellin M, Orlova VV, Mummery CL. Generation, functional analysis and applications of isogenic three-dimensional self-aggregating cardiac microtissues from human pluripotent stem cells. Nat Protoc 2021; 16:2213-2256. [PMID: 33772245 PMCID: PMC7611409 DOI: 10.1038/s41596-021-00497-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/11/2021] [Indexed: 02/01/2023]
Abstract
Tissue-like structures from human pluripotent stem cells containing multiple cell types are transforming our ability to model and understand human development and disease. Here we describe a protocol to generate cardiomyocytes (CMs), cardiac fibroblasts (CFs) and cardiac endothelial cells (ECs), the three principal cell types in the heart, from human induced pluripotent stem cells (hiPSCs) and combine them in three-dimensional (3D) cardiac microtissues (MTs). We include details of how to differentiate, isolate, cryopreserve and thaw the component cells and how to construct and analyze the MTs. The protocol supports hiPSC-CM maturation and allows replacement of one or more of the three heart cell types in the MTs with isogenic variants bearing disease mutations. Differentiation of each cell type takes ~30 d, while MT formation and maturation requires another 20 d. No specialist equipment is needed and the method is inexpensive, requiring just 5,000 cells per MT.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ruben W.J. van Helden
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Biology, University of Padua, 35121 Padua, Italy,Veneto Institute of Molecular Medicine, 35129 Padua, Italy,Correspondence to , or
| | - Valeria V. Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Correspondence to , or
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands,Department of Applied Stem Cell Technologies, University of Twente, The Netherlands,Correspondence to , or
| |
Collapse
|
16
|
Contessi Negrini N, Angelova Volponi A, Higgins C, Sharpe P, Celiz A. Scaffold-based developmental tissue engineering strategies for ectodermal organ regeneration. Mater Today Bio 2021; 10:100107. [PMID: 33889838 PMCID: PMC8050778 DOI: 10.1016/j.mtbio.2021.100107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/15/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering (TE) is a multidisciplinary research field aiming at the regeneration, restoration, or replacement of damaged tissues and organs. Classical TE approaches combine scaffolds, cells and soluble factors to fabricate constructs mimicking the native tissue to be regenerated. However, to date, limited success in clinical translations has been achieved by classical TE approaches, because of the lack of satisfactory biomorphological and biofunctional features of the obtained constructs. Developmental TE has emerged as a novel TE paradigm to obtain tissues and organs with correct biomorphology and biofunctionality by mimicking the morphogenetic processes leading to the tissue/organ generation in the embryo. Ectodermal appendages, for instance, develop in vivo by sequential interactions between epithelium and mesenchyme, in a process known as secondary induction. A fine artificial replication of these complex interactions can potentially lead to the fabrication of the tissues/organs to be regenerated. Successful developmental TE applications have been reported, in vitro and in vivo, for ectodermal appendages such as teeth, hair follicles and glands. Developmental TE strategies require an accurate selection of cell sources, scaffolds and cell culture configurations to allow for the correct replication of the in vivo morphogenetic cues. Herein, we describe and discuss the emergence of this TE paradigm by reviewing the achievements obtained so far in developmental TE 3D scaffolds for teeth, hair follicles, and salivary and lacrimal glands, with particular focus on the selection of biomaterials and cell culture configurations.
Collapse
Affiliation(s)
| | - A. Angelova Volponi
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - C.A. Higgins
- Department of Bioengineering, Imperial College London, London, UK
| | - P.T. Sharpe
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - A.D. Celiz
- Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
17
|
Abstract
Supramolecular biopolymers (SBPs) are those polymeric units derived from macromolecules that can assemble with each other by noncovalent interactions. Macromolecular structures are commonly found in living systems such as proteins, DNA/RNA, and polysaccharides. Bioorganic chemistry allows the generation of sequence-specific supramolecular units like SBPs that can be tailored for novel applications in tissue engineering (TE). SBPs hold advantages over other conventional polymers previously used for TE; these materials can be easily functionalized; they are self-healing, biodegradable, stimuli-responsive, and nonimmunogenic. These characteristics are vital for the further development of current trends in TE, such as the use of pluripotent cells for organoid generation, cell-free scaffolds for tissue regeneration, patient-derived organ models, and controlled delivery systems of small molecules. In this review, we will analyse the 3 subtypes of SBPs: peptide-, nucleic acid-, and oligosaccharide-derived. Then, we will discuss the role that SBPs will be playing in TE as dynamic scaffolds, therapeutic scaffolds, and bioinks. Finally, we will describe possible outlooks of SBPs for TE.
Collapse
|
18
|
Li Q, Yang G, Li J, Ding M, Zhou N, Dong H, Mou Y. Stem cell therapies for periodontal tissue regeneration: a network meta-analysis of preclinical studies. Stem Cell Res Ther 2020; 11:427. [PMID: 33008471 PMCID: PMC7531120 DOI: 10.1186/s13287-020-01938-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022] Open
Abstract
Background Periodontal tissue regeneration (PTR) is the ultimate goal of periodontal therapy. Currently, stem cell therapy is considered a promising strategy for achieving PTR. However, there is still no conclusive comparison that distinguishes clear hierarchies among different kinds of stem cells. Methods A systematic review and network meta-analysis (NMA) was performed using MEDLINE (via PubMed), EMBASE, and Web of Science up to February 2020. Preclinical studies assessing five types of stem cells for PTR were included; the five types of stem cells included periodontal ligament-derived stem cells (PDLSCs), bone marrow-derived stem cells (BMSCs), adipose tissue-derived stem cells (ADSCs), dental pulp-derived stem cells (DPSCs), and gingival-derived stem cells (GMSCs). The primary outcomes were three histological indicators with continuous variables: newly formed alveolar bone (NB), newly formed cementum (NC), and newly formed periodontal ligament (NPDL). We performed pairwise meta-analyses using a random-effects model and then performed a random-effects NMA using a multivariate meta-analysis model. Results Sixty preclinical studies assessing five different stem cell-based therapies were identified. The NMA showed that in terms of NB, PDLSCs (standardized mean difference 1.87, 95% credible interval 1.24 to 2.51), BMSCs (1.88, 1.17 to 2.59), and DPSCs (1.69, 0.64 to 2.75) were statistically more efficacious than cell carriers (CCs). In addition, PDLSCs were superior to GMSCs (1.49, 0.04 to 2.94). For NC, PDLSCs (2.18, 1.48 to 2.87), BMSCs (2.11, 1.28 to 2.94), and ADSCs (1.55, 0.18 to 2.91) were superior to CCs. For NPDL, PDLSCs (1.69, 0.92 to 2.47) and BMSCs (1.41, 0.56 to 2.26) were more efficacious than CCs, and PDLSCs (1.26, 0.11 to 2.42) were superior to GMSCs. The results of treatment hierarchies also demonstrated that the two highest-ranked interventions were PDLSCs and BMSCs. Conclusion PDLSCs and BMSCs were the most effective and well-documented stem cells for PTR among the five kinds of stem cells evaluated in this study, and there was no statistical significance between them. To translate the stem cell therapies for PTR successfully in the clinic, future studies should utilize robust experimental designs and reports.
Collapse
Affiliation(s)
- Qiang Li
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guangwen Yang
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jialing Li
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Meng Ding
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Na Zhou
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Heng Dong
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China. .,Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Yongbin Mou
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
19
|
Liao J, Lu X, Shao X, Zhu L, Fan X. Uncovering an Organ's Molecular Architecture at Single-Cell Resolution by Spatially Resolved Transcriptomics. Trends Biotechnol 2020; 39:43-58. [PMID: 32505359 DOI: 10.1016/j.tibtech.2020.05.006] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/17/2023]
Abstract
Revealing fine-scale cellular heterogeneity among spatial context and the functional and structural foundations of tissue architecture is fundamental within biological research and pharmacology. Unlike traditional approaches involving single molecules or bulk omics, cutting-edge, spatially resolved transcriptomics techniques offer near-single-cell or even subcellular resolution within tissues. Massive information across higher dimensions along with position-coordinating labels can better map the whole 3D transcriptional landscape of tissues. In this review, we focus on developments and strategies in spatially resolved transcriptomics, compare the cell and gene throughput and spatial resolution in detail for existing methods, and highlight the enormous potential in biomedical research.
Collapse
Affiliation(s)
- Jie Liao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xin Shao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Ling Zhu
- The Save Sight Institute, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW 2000, Australia
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; The Save Sight Institute, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW 2000, Australia.
| |
Collapse
|
20
|
Yang Z, Xu H, Zhao X. Designer Self-Assembling Peptide Hydrogels to Engineer 3D Cell Microenvironments for Cell Constructs Formation and Precise Oncology Remodeling in Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903718. [PMID: 32382486 PMCID: PMC7201262 DOI: 10.1002/advs.201903718] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/08/2020] [Indexed: 02/05/2023]
Abstract
Designer self-assembling peptides form the entangled nanofiber networks in hydrogels by ionic-complementary self-assembly. This type of hydrogel has realistic biological and physiochemical properties to serve as biomimetic extracellular matrix (ECM) for biomedical applications. The advantages and benefits are distinct from natural hydrogels and other synthetic or semisynthetic hydrogels. Designer peptides provide diverse alternatives of main building blocks to form various functional nanostructures. The entangled nanofiber networks permit essential compositional complexity and heterogeneity of engineering cell microenvironments in comparison with other hydrogels, which may reconstruct the tumor microenvironments (TMEs) in 3D cell cultures and tissue-specific modeling in vitro. Either ovarian cancer progression or recurrence and relapse are involved in the multifaceted TMEs in addition to mesothelial cells, fibroblasts, endothelial cells, pericytes, immune cells, adipocytes, and the ECM. Based on the progress in common hydrogel products, this work focuses on the diverse designer self-assembling peptide hydrogels for instructive cell constructs in tissue-specific modeling and the precise oncology remodeling for ovarian cancer, which are issued by several research aspects in a 3D context. The advantages and significance of designer peptide hydrogels are discussed, and some common approaches and coming challenges are also addressed in current complex tumor diseases.
Collapse
Affiliation(s)
- Zehong Yang
- West China School of Basic Medical Sciences and Forensic MedicineSichuan UniversityChengduSichuan610041P. R. China
- Institute for Nanobiomedical Technology and Membrane BiologyWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Hongyan Xu
- GL Biochem (Shanghai) Ltd.519 Ziyue Rd.Shanghai200241P. R. China
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane BiologyWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
- Wenzhou InstituteUniversity of Chinese Academy of Sciences (Wenzhou Institute of Biomaterials & Engineering)WenzhouZhejiang325001P. R. China
| |
Collapse
|
21
|
Mueller M, Rasoulinejad S, Garg S, Wegner SV. The Importance of Cell-Cell Interaction Dynamics in Bottom-Up Tissue Engineering: Concepts of Colloidal Self-Assembly in the Fabrication of Multicellular Architectures. NANO LETTERS 2020; 20:2257-2263. [PMID: 31751141 PMCID: PMC7146848 DOI: 10.1021/acs.nanolett.9b04160] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/19/2019] [Indexed: 06/10/2023]
Abstract
Building tissue from cells as the basic building block based on principles of self-assembly is a challenging and promising approach. Understanding how far principles of self-assembly and self-sorting known for colloidal particles apply to cells remains unanswered. In this study, we demonstrate that not just controlling the cell-cell interactions but also their dynamics is a crucial factor that determines the formed multicellular structure, using photoswitchable interactions between cells that are activated with blue light and reverse in the dark. Tuning dynamics of the cell-cell interactions by pulsed light activation results in multicellular architectures with different sizes and shapes. When the interactions between cells are dynamic, compact and round multicellular clusters under thermodynamic control form, while otherwise branched and loose aggregates under kinetic control assemble. These structures parallel what is known for colloidal assemblies under reaction- and diffusion-limited cluster aggregation, respectively. Similarly, dynamic interactions between cells are essential for cells to self-sort into distinct groups. Using four different cell types, which expressed two orthogonal cell-cell interaction pairs, the cells sorted into two separate assemblies. Bringing concepts of colloidal self-assembly to bottom-up tissue engineering provides a new theoretical framework and will help in the design of more predictable tissue-like structures.
Collapse
Affiliation(s)
- Marc Mueller
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | | | - Sukant Garg
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Seraphine V. Wegner
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
- Institute
of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| |
Collapse
|
22
|
Nguyen R, Bae SDW, Zhou G, Read SA, Ahlenstiel G, George J, Qiao L. Application of organoids in translational research of human diseases with a particular focus on gastrointestinal cancers. Biochim Biophys Acta Rev Cancer 2020; 1873:188350. [PMID: 32007597 DOI: 10.1016/j.bbcan.2020.188350] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/15/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers constitute the largest portion of all human cancers and represent a significant health burden on modern society. Conventional therapeutic approaches such as chemotherapy and surgical resections often fail due to poor treatment response or tumour relapse. Unfortunately, drug discovery for GI cancers has stalled as current cancer models fail to recapitulate critical features of the parent tumour, leading to poor translation from bench to bedside. Recent advances in three-dimensional (3D) cell culturing techniques have driven the surge of interest in stem cell-derived organoid models, a promising platform with a plethora of potential applications due to its ability to retain crucial architectural, genomic and transcriptional properties of the native tissue. In this review article, we discuss current applications and advantages of organoid models in the translational research of GI cancers with a particular focus on primary liver cancer that currently lack effective curative treatments.
Collapse
Affiliation(s)
- Romario Nguyen
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Sarah Da Won Bae
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Gang Zhou
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Scott A Read
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia; Blacktown Medical School, Western Sydney University, Blacktown, NSW, Australia
| | - Golo Ahlenstiel
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia; Blacktown Medical School, Western Sydney University, Blacktown, NSW, Australia; Blacktown Hospital, Blacktown, NSW, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia.
| |
Collapse
|
23
|
Galzi JL, Jouault T, Amédée J. Les organoïdes : des mini-organes au service de la biomédecine. Med Sci (Paris) 2019; 35:467-469. [DOI: 10.1051/medsci/2019091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
24
|
Preparation of hair beads and hair follicle germs for regenerative medicine. Biomaterials 2019; 212:55-63. [PMID: 31103946 DOI: 10.1016/j.biomaterials.2019.05.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/24/2022]
Abstract
Hair regenerative medicine is a promising approach for hair loss, during which autologous follicular stem cells are transplanted into regions of hair loss to regenerate hairs. Because cells transplanted as a single cell suspension scarcely generate hairs, the engineering of three-dimensional (3D) tissues before transplantation has been explored to improve this process. Here, we propose an approach to fabricate collagen-enriched cell aggregates, named hair beads (HBs), through the spontaneous constriction of cell-encapsulated collagen drops. Mouse embryonic mesenchymal cells or human dermal papilla cells were encapsulated in 2-μl collagen microgels, which were concentrated >10-fold in volume during 3 days of culture. Interestingly, HB constriction was attributed to attraction forces driven by myosin II and involved the upregulation of follicular genes. Single HBs with epithelial cells seeded in U-shaped microwells formed dumbbell-like structures comprising respective aggregates (named bead-based hair follicle germs, bbHFGs), during 3 days of culture. bbHFGs efficiently generated hair follicles upon intracutaneous transplantation into the backs of nude mice. Using an automated spotter, this approach was scalable to prepare a large number of bbHFGs, which is important for clinical applications. Therefore, this could represent a robust and practical approach for the preparation of germ-like tissues for hair regenerative medicine.
Collapse
|
25
|
Yüz SG, Rasoulinejad S, Mueller M, Wegner AE, Wegner SV. Blue Light Switchable Cell-Cell Interactions Provide Reversible and Spatiotemporal Control Towards Bottom-Up Tissue Engineering. ADVANCED BIOSYSTEMS 2019; 3:e1800310. [PMID: 32627428 DOI: 10.1002/adbi.201800310] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/14/2018] [Indexed: 12/22/2022]
Abstract
Controlling cell-cell interactions is central for understanding key cellular processes and bottom-up tissue assembly from single cells. The challenge is to control cell-cell interactions dynamically and reversibly with high spatiotemporal precision noninvasively and sustainably. In this study, cell-cell interactions are controlled with visible light using an optogenetic approach by expressing the blue light switchable proteins CRY2 or CIBN on the surfaces of cells. CRY2 and CIBN expressing cells form specific heterophilic interactions under blue light providing precise control in space and time. Further, these interactions are reversible in the dark and can be repeatedly and dynamically switched on and off. Unlike previous approaches, these genetically encoded proteins allow for long-term expression of the interaction domains and respond to nontoxic low intensity blue light. In addition, these interactions are suitable to assemble cells into 3D multicellular architectures. Overall, this approach captures the dynamic and reversible nature of cell-cell interactions and controls them noninvasively and sustainably both in space and time. This provides a new way of studying cell-cell interactions and assembling cellular building blocks into tissues with unmatched flexibility.
Collapse
Affiliation(s)
- Simge G Yüz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
| | - Samaneh Rasoulinejad
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Marc Mueller
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Anatol E Wegner
- Department of Statistical Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Seraphine V Wegner
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| |
Collapse
|
26
|
Bejoy J, Wang Z, Bijonowski B, Yang M, Ma T, Sang QX, Li Y. Differential Effects of Heparin and Hyaluronic Acid on Neural Patterning of Human Induced Pluripotent Stem Cells. ACS Biomater Sci Eng 2018; 4:4354-4366. [PMID: 31572767 PMCID: PMC6768405 DOI: 10.1021/acsbiomaterials.8b01142] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A lack of well-established animal models that can efficiently represent human brain pathology has led to the development of human induced pluripotent stem cell (hiPSC)-derived brain tissues. Brain organoids have enhanced our ability to understand the developing human brain and brain disorders (e.g., Schizophrenia, microcephaly), but the organoids still do not accurately recapitulate the anatomical organization of the human brain. Therefore, it is important to evaluate and optimize induction and signaling factors in order to engineer the next generation of brain organoids. In this study, the impact of hyaluronic acid (HA), a major brain extracellular matrix (ECM) component that interacts with cells through ligand-binding receptors, on the patterning of brain organoids from hiPSCs was evaluated. To mediate HA- binding capacity of signaling molecules, heparin was added in addition to HA or conjugated to HA to form hydrogels (with two different moduli). The neural cortical spheroids derived from hiPSCs were treated with either HA or heparin plus HA (Hep- HA) and were analyzed for ECM impacts on neural patterning. The results indicate that Hep-HA has a caudalizing effect on hiPSC-derived neural spheroids, in particular for stiff Hep-HA hydrogels. Wnt and Hippo/Yes-associated protein (YAP) signaling was modulated (using Wnt inhibitor IWP4 or actin disruption agent Cytochalasin D respectively) to understand the underlying mechanism. IWP4 and cytochalasin D promote forebrain identity. The results from this study should enhance the understanding of influence of biomimetic ECM factors for brain organoid generation.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Zhe Wang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States
| | - Brent Bijonowski
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Mo Yang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Qing-Xiang Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, United States
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
27
|
Wang H. Modeling Neurological Diseases With Human Brain Organoids. Front Synaptic Neurosci 2018; 10:15. [PMID: 29937727 PMCID: PMC6002496 DOI: 10.3389/fnsyn.2018.00015] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/22/2018] [Indexed: 12/18/2022] Open
Abstract
The complexity and delicacy of human brain make it challenging to recapitulate its development, function and disorders. Brain organoids derived from human pluripotent stem cells (PSCs) provide a new tool to model both normal and pathological human brain, and greatly enhance our ability to study brain biology and diseases. Currently, human brain organoids are increasingly used in modeling neurological disorders and relative therapeutic discovery. This review article focuses on recent advances in human brain organoid system and its application in disease modeling. It also discusses the limitations and future perspective of human brain organoids in modeling neurological diseases.
Collapse
Affiliation(s)
- Hansen Wang
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Saheli M, Sepantafar M, Pournasr B, Farzaneh Z, Vosough M, Piryaei A, Baharvand H. Three-dimensional liver-derived extracellular matrix hydrogel promotes liver organoids function. J Cell Biochem 2018; 119:4320-4333. [PMID: 29247536 DOI: 10.1002/jcb.26622] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/12/2017] [Indexed: 12/25/2022]
Abstract
An important advantage of employing extracellular matrix (ECM)-derived biomaterials in tissue engineering is the ability to tailor the biochemical and biophysical microenvironment of the cells. This study aims to assess whether three-dimensional (3D) liver-derived ECM hydrogel (LEMgel) promotes physiological function of liver organoids generated by self-organization of human hepatocarcinoma cells together with human mesenchymal and endothelial cells. We have optimized the decellularization method to fabricate liver ECM derived from sheep to preserve the greatest content of glycosaminoglycans, collagen, laminin, and fibronectin in produced LEMgel. During gelation, complex viscoelasticity modulus of the LEMgel (3 mg/mL) increased from 186.7 to 1570.5 Pa and Tan Delta decreased from 0.27 to 0.18. Scanning electron microscopy (SEM) determined that the LEMgel had a pore size of 382 ± 71 µm. Hepatocarcinoma cells in the self-organized liver organoids in 3D LEMgel (LEMgel organoids) showed an epithelial phenotype and expressed ALB, CYP3A4, E-cadherin, and ASGPR. The LEMgel organoid had significant upregulation of transcripts of ALB, CYP3A4, CYP3A7, and TAT as well as downregulation of AFP compared to collagen type I- and hydrogel-free-organoids or organoids in solubilized LEM and 2D culture of hepatocarcinoma cells. Generated 3D LEMgel organoids had significantly more ALB and AAT secretion, urea production, CYP3A4 enzyme activity, and inducibility. In conclusion, 3D LEMgel enhanced the functional activity of self-organized liver organoids compared to traditional 2D, 3D, and collagen gel cultures. Our novel 3D LEMgel organoid could potentially be used in liver tissue engineering, drug discovery, toxicology studies, or bio-artificial liver fabrication.
Collapse
Affiliation(s)
- Mona Saheli
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadmajid Sepantafar
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Behshad Pournasr
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technology in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
29
|
The Synergy of Scaffold-Based and Scaffold-Free Tissue Engineering Strategies. Trends Biotechnol 2018; 36:348-357. [PMID: 29475621 DOI: 10.1016/j.tibtech.2018.01.005] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/24/2017] [Accepted: 01/11/2018] [Indexed: 02/07/2023]
Abstract
Tissue engineering (TE) is a highly interdisciplinary research field driven by the goal to restore, replace, or regenerate defective tissues. Throughout more than two decades of intense research, different technological approaches, which can be principally categorized into scaffold-based and scaffold-free strategies, have been developed. In this opinion article, we discuss the emergence of a third strategy in TE. This synergetic strategy integrates the advantages of both of these traditional approaches, while being clearly distinct from them. Its characteristic attributes, numerous practical benefits, and recent literature reports supporting our opinion, are discussed in detail.
Collapse
|
30
|
Jin MZ, Han RR, Qiu GZ, Ju XC, Lou G, Jin WL. Organoids: An intermediate modeling platform in precision oncology. Cancer Lett 2018; 414:174-180. [PMID: 29174804 DOI: 10.1016/j.canlet.2017.11.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023]
Abstract
Cancer harbors variable heterogeneity and plasticity. Thus far, our comprehension is greatly based on cell lines, organoids, and patient-derived tumor xenografts (PDTXs). Organoids are a three-dimensional in vitro culture platform constructed from self-organizing stem cells. They can almost accurately recapitulate tumor heterogeneity and microenvironment "in a dish," which surpass established cell lines and are not as expensive and time-consuming as PDTXs. As an intermediate model, tumor organoids are also used to study the fundamental issues of tumorigenesis and metastasis. They are specifically applied for drug testing and stored as "living biobanks." In this review, we highlight the translational applications of organoid technologies in tumor research and precision medicine, discuss the advantages and limitations compared with other mentioned methods, and provide our outlook on its future.
Collapse
Affiliation(s)
- Ming-Zhu Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Run-Run Han
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China
| | - Guan-Zhong Qiu
- Department of Neurosurgery, General Hospital of Jinan Military Command, Jinan 250031, PR China
| | - Xiang-Chun Ju
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China.
| | - Ge Lou
- Department of Gynecology Oncology, The Tumor Hospital, Harbin Medical University, Harbin, 150086, PR China.
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China; National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Shaanxi Key Laboratory of Brain Disorders and School of Basic Medical Sciences, Xi'an Medical University, Xi'an 710021, PR China.
| |
Collapse
|
31
|
Ahrens CC, Dong Z, Li W. Engineering cell aggregates through incorporated polymeric microparticles. Acta Biomater 2017; 62:64-81. [PMID: 28782721 DOI: 10.1016/j.actbio.2017.08.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 12/16/2022]
Abstract
Ex vivo cell aggregates must overcome significant limitations in the transport of nutrients, drugs, and signaling proteins compared to vascularized native tissue. Further, engineered extracellular environments often fail to sufficiently replicate tethered signaling cues and the complex architecture of native tissue. Co-cultures of cells with microparticles (MPs) is a growing field directed towards overcoming many of these challenges by providing local and controlled presentation of both soluble and tethered proteins and small molecules. Further, co-cultured MPs offer a mechanism to better control aggregate architecture and even to report key characteristics of the local microenvironment such as pH or oxygen levels. Herein, we provide a brief introduction to established and developing strategies for MP production including the choice of MP materials, fabrication techniques, and techniques for incorporating additional functionality. In all cases, we emphasize the specific utility of each approach to form MPs useful for applications in cell aggregate co-culture. We review established techniques to integrate cells and MPs. We highlight those strategies that promote targeted heterogeneity or homogeneity, and we describe approaches to engineer cell-particle and particle-particle interactions that enhance aggregate stability and biological response. Finally, we review advances in key application areas of MP aggregates and future areas of development. STATEMENT OF SIGNIFICANT Cell-scaled polymer microparticles (MPs) integrated into cellular aggregates have been shown to be a powerful tool to direct cell response. MPs have supported the development of healthy cartilage, islets, nerves, and vasculature by the maintenance of soluble gradients as well as by the local presentation of tethered cues and diffusing proteins and small molecules. MPs integrated with pluripotent stem cells have directed in vivo expansion and differentiation. Looking forward, MPs are expected to support both the characterization and development of in vitro tissue systems for applications such as drug testing platforms. However, useful co-cultures must be designed keeping in mind the limitations and attributes of each material strategy within the context of the overall tissue biology. The present review integrates prospectives from materials development, drug delivery, and tissue engineering to provide a toolbox for the development and application of MPs useful for long-term co-culture within cell aggregates.
Collapse
Affiliation(s)
- Caroline C Ahrens
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States
| | - Ziye Dong
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States
| | - Wei Li
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States.
| |
Collapse
|
32
|
Liquid Marble as Bioreactor for Engineering Three-Dimensional Toroid Tissues. Sci Rep 2017; 7:12388. [PMID: 28959016 PMCID: PMC5620055 DOI: 10.1038/s41598-017-12636-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/13/2017] [Indexed: 11/09/2022] Open
Abstract
Liquid marble is a liquid droplet coated with hydrophobic powder that can be used as a bioreactor. This paper reports the three-dimensional self-assembly and culture of a cell toroid in a slow-releasing, non-adhesive and evaporation-reducing bioreactor platform based on a liquid marble. The bioreactor is constructed by embedding a hydrogel sphere containing growth factor into a liquid marble filled with a suspension of dissociated cells. The hydrogel maintains the water content and concurrently acts as a slow-release carrier. The concentration gradient of growth factor induces cell migration and assembly into toroidal aggregates. An optimum cell concentration resulted in the toroidal (doughnut-like) tissue after 12 hours. The harvested cell toroids showed rapid closure of the inner opening when treated with the growth factor. We also present a geometric growth model to describe the shape of the toroidal tissue over time. In analogy to the classical two-dimensional scratch assay, we propose that the cell toroids reported here open up new possibilities to screen drugs affecting cell migration in three dimensions.
Collapse
|
33
|
Picollet-D'hahan N, Dolega ME, Freida D, Martin DK, Gidrol X. Deciphering Cell Intrinsic Properties: A Key Issue for Robust Organoid Production. Trends Biotechnol 2017; 35:1035-1048. [PMID: 28927991 DOI: 10.1016/j.tibtech.2017.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 07/18/2017] [Accepted: 08/08/2017] [Indexed: 02/07/2023]
Abstract
We highlight the disposition of various cell types to self-organize into complex organ-like structures without necessarily the support of any stromal cells, provided they are placed into permissive 3D culture conditions. The goal of generating organoids reproducibly and efficiently has been hampered by poor understanding of the exact nature of the intrinsic cell properties at the origin of organoid generation, and of the signaling pathways governing their differentiation. Using microtechnologies like microfluidics to engineer organoids would create opportunities for single-cell genomics and high-throughput functional genomics to exhaustively characterize cell intrinsic properties. A more complete understanding of the development of organoids would enhance their relevance as models to study organ morphology, function, and disease and would open new avenues in drug development and regenerative medicine.
Collapse
Affiliation(s)
| | - Monika E Dolega
- Université Grenoble Alpes, INSERM, CEA, BIG, F-38000 Grenoble, France
| | - Delphine Freida
- Université Grenoble Alpes, INSERM, CEA, BIG, F-38000 Grenoble, France
| | - Donald K Martin
- Université Grenoble Alpes, F-38000 Grenoble, France; TIMC-IMAG/CNRS UMR 5525, F-38041 Grenoble, France
| | - Xavier Gidrol
- Université Grenoble Alpes, INSERM, CEA, BIG, F-38000 Grenoble, France.
| |
Collapse
|
34
|
Ozdemir T, Srinivasan PP, Zakheim DR, Harrington DA, Witt RL, Farach-Carson MC, Jia X, Pradhan-Bhatt S. Bottom-up assembly of salivary gland microtissues for assessing myoepithelial cell function. Biomaterials 2017; 142:124-135. [PMID: 28734180 DOI: 10.1016/j.biomaterials.2017.07.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 11/15/2022]
Abstract
Myoepithelial cells are flat, stellate cells present in exocrine tissues including the salivary glands. While myoepithelial cells have been studied extensively in mammary and lacrimal gland tissues, less is known of the function of myoepithelial cells derived from human salivary glands. Several groups have isolated tumorigenic myoepithelial cells from cancer specimens, however, only one report has demonstrated isolation of normal human salivary myoepithelial cells needed for use in salivary gland tissue engineering applications. Establishing a functional organoid model consisting of myoepithelial and secretory acinar cells is therefore necessary for understanding the coordinated action of these two cell types in unidirectional fluid secretion. Here, we developed a bottom-up approach for generating salivary gland microtissues using primary human salivary myoepithelial cells (hSMECs) and stem/progenitor cells (hS/PCs) isolated from normal salivary gland tissues. Phenotypic characterization of isolated hSMECs confirmed that a myoepithelial cell phenotype consistent with that from other exocrine tissues was maintained over multiple passages of culture. Additionally, hSMECs secreted basement membrane proteins, expressed adrenergic and cholinergic neurotransmitter receptors, and released intracellular calcium [Ca2+i] in response to parasympathetic agonists. In a collagen I contractility assay, activation of contractile machinery was observed in isolated hSMECs treated with parasympathetic agonists. Recombination of hSMECs with assembled hS/PC spheroids in a microwell system was used to create microtissues resembling secretory complexes of the salivary gland. We conclude that the engineered salivary gland microtissue complexes provide a physiologically relevant model for both mechanistic studies and as a building block for the successful engineering of the salivary gland for restoration of salivary function in patients suffering from hyposalivation.
Collapse
Affiliation(s)
- Tugba Ozdemir
- Department of Materials Sciences and Engineering, University of Delaware, Newark, DE, USA
| | - Padma Pradeepa Srinivasan
- Department of Biological Sciences, University of Delaware, Newark, DE, USA; Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA
| | - Daniel R Zakheim
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Daniel A Harrington
- BioSciences, Rice University, Houston, TX, USA; Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| | - Robert L Witt
- Department of Biological Sciences, University of Delaware, Newark, DE, USA; Otolaryngology - Head & Neck Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mary C Farach-Carson
- Department of Biological Sciences, University of Delaware, Newark, DE, USA; BioSciences, Rice University, Houston, TX, USA; Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| | - Xinqiao Jia
- Department of Materials Sciences and Engineering, University of Delaware, Newark, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| | - Swati Pradhan-Bhatt
- Department of Biological Sciences, University of Delaware, Newark, DE, USA; Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
35
|
Liu Y, Gill E, Shery Huang YY. Microfluidic on-chip biomimicry for 3D cell culture: a fit-for-purpose investigation from the end user standpoint. Future Sci OA 2017; 3:FSO173. [PMID: 28670465 PMCID: PMC5481809 DOI: 10.4155/fsoa-2016-0084] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/19/2017] [Indexed: 12/13/2022] Open
Abstract
A plethora of 3D and microfluidics-based culture models have been demonstrated in the recent years with the ultimate aim to facilitate predictive in vitro models for pharmaceutical development. This article summarizes to date the progress in the microfluidics-based tissue culture models, including organ-on-a-chip and vasculature-on-a-chip. Specific focus is placed on addressing the question of what kinds of 3D culture and system complexities are deemed desirable by the biological and biomedical community. This question is addressed through analysis of a research survey to evaluate the potential use of microfluidic cell culture models among the end users. Our results showed a willingness to adopt 3D culture technology among biomedical researchers, although a significant gap still exists between the desired systems and existing 3D culture options. With these results, key challenges and future directions are highlighted.
Collapse
Affiliation(s)
- Ye Liu
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, UK, CB2 1PZ
| | - Elisabeth Gill
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, UK, CB2 1PZ
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, UK, CB2 1PZ
| |
Collapse
|
36
|
Sasaki K, Akagi T, Asaoka T, Eguchi H, Fukuda Y, Iwagami Y, Yamada D, Noda T, Wada H, Gotoh K, Kawamoto K, Doki Y, Mori M, Akashi M. Construction of three-dimensional vascularized functional human liver tissue using a layer-by-layer cell coating technique. Biomaterials 2017; 133:263-274. [PMID: 28448819 DOI: 10.1016/j.biomaterials.2017.02.034] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/19/2017] [Accepted: 02/26/2017] [Indexed: 12/14/2022]
Abstract
The creation of artificial liver tissue is an active area of research due to the shortage of donors for liver transplantation. Here we investigated whether a simple and efficient cell coating technique developed in our laboratory could be used to generate functional vascularized liver tissue. This technique creates three-dimensional tissue by loading cells sterically onto other cells that have been coated with layer-by-layer (LbL) nanofilms of fibronectin and gelatin, two extracellular matrix proteins. We used this technique to construct homogenous, dense, well-vascularized liver tissue from cryopreserved human primary hepatocytes, human umbilical vein endothelial cells, and normal human dermal fibroblasts. Using LbL cell coating technique resulted in higher cellular function in terms of human albumin production (P < 0.01) and cytochrome P450 activity (P < 0.01) in vitro. Furthermore, after being transplanted subcutaneously into NOD/SCID mice, the vascularized liver tissue showed greater albumin production in the early stage than non-vascularized tissue or a hepatocyte suspension (P < 0.01). Histological examination demonstrated that compare to non-vascularized tissue, there were many less-morphologically changed and intact hepatocytes in the vascularized tissue. This cell coating technique would be applicable to the generation of vascularized functional liver tissue for regenerative medicine in the future.
Collapse
Affiliation(s)
- Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Takami Akagi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Japan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Yasunari Fukuda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Hiroshi Wada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Kunihito Gotoh
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Koichi Kawamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Japan
| | - Mitsuru Akashi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Japan.
| |
Collapse
|
37
|
Zakikhan K, Pournasr B, Vosough M, Nassiri-Asl M. In Vitro Generated Hepatocyte-Like Cells: A Novel Tool in Regenerative Medicine and Drug Discovery. CELL JOURNAL 2017; 19:204-217. [PMID: 28670513 PMCID: PMC5412779 DOI: 10.22074/cellj.2016.4362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/05/2016] [Indexed: 12/19/2022]
Abstract
Hepatocyte-like cells (HLCs) are generated from either various human pluripotent stem
cells (hPSCs) including induced pluripotent stem cells (iPSCs) and embryonic stem cells
(ESCs), or direct cell conversion, mesenchymal stem cells as well as other stem cells like
gestational tissues. They provide potential cell sources for biomedical applications. Liver
transplantation is the gold standard treatment for the patients with end stage liver disease,
but there are many obstacles limiting this process, like insufficient number of donated
healthy livers. Meanwhile, the number of patients receiving a liver organ transplant for
a better life is increasing. In this regard, HLCs may provide an adequate cell source to
overcome these shortages. New molecular engineering approaches such as CRISPR/
Cas system applying in iPSCs technology provide the basic principles of gene correction
for monogenic inherited metabolic liver diseases, as another application of HLCs. It has
been shown that HLCs could replace primary human hepatocytes in drug discovery and
hepatotoxicity tests. However, generation of fully functional HLCs is still a big challenge;
several research groups have been trying to improve current differentiation protocols to
achieve better HLCs according to morphology and function of cells. Large-scale generation
of functional HLCs in bioreactors could make a new opportunity in producing enough
hepatocytes for treating end-stage liver patients as well as other biomedical applications
such as drug studies. In this review, regarding the biomedical value of HLCs, we focus
on the current and efficient approaches for generating hepatocyte-like cells in vitro and
discuss about their applications in regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Kobra Zakikhan
- Cellular and Molecular Research Center, Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Behshad Pournasr
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marjan Nassiri-Asl
- Cellular and Molecular Research Center, Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.,Cellular and Molecular Research Center, Department of Pharmacology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
38
|
|
39
|
Ng WL, Lee JM, Yeong WY, Win Naing M. Microvalve-based bioprinting – process, bio-inks and applications. Biomater Sci 2017; 5:632-647. [DOI: 10.1039/c6bm00861e] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
DOD microvalve-based bioprinting system provides a highly advanced manufacturing platform that facilitates precise control over the cellular and biomaterial deposition in a highly reproducible and reliable manner. This article highlights promising directions to transform microvalve-based bioprinting into an enabling technology that will potentially drive significant advances in the field of TERM.
Collapse
Affiliation(s)
- Wei Long Ng
- Singapore Centre for 3D Printing (SC3DP)
- School of Mechanical and Aerospace Engineering
- Nanyang Technological University (NTU)
- Singapore 639798
- Singapore
| | - Jia Min Lee
- Singapore Centre for 3D Printing (SC3DP)
- School of Mechanical and Aerospace Engineering
- Nanyang Technological University (NTU)
- Singapore 639798
- Singapore
| | - Wai Yee Yeong
- Singapore Centre for 3D Printing (SC3DP)
- School of Mechanical and Aerospace Engineering
- Nanyang Technological University (NTU)
- Singapore 639798
- Singapore
| | - May Win Naing
- Singapore Institute of Manufacturing Technology (SIMTech)
- Agency for Science
- Technology and Research
- Singapore 637662
| |
Collapse
|
40
|
Compaan AM, Christensen K, Huang Y. Inkjet Bioprinting of 3D Silk Fibroin Cellular Constructs Using Sacrificial Alginate. ACS Biomater Sci Eng 2016; 3:1519-1526. [DOI: 10.1021/acsbiomaterials.6b00432] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Ashley M. Compaan
- Department
of Materials Science and Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Kyle Christensen
- Department of
Mechanical and Aerospace
Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Yong Huang
- Department
of Materials Science and Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department of
Mechanical and Aerospace
Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
41
|
Yeh CH, Chen PC, Chen CH, Hsu CF, Huang RL, Ding DC, Chu TY. Platelet-Derived Growth Factor in the Ovarian Follicle Attracts the Stromal Cells of the Fallopian Tube Fimbriae. PLoS One 2016; 11:e0158266. [PMID: 27379403 PMCID: PMC4933398 DOI: 10.1371/journal.pone.0158266] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 06/13/2016] [Indexed: 01/09/2023] Open
Abstract
During human ovulation, the fallopian tube fimbriae must move to the ovulation site to catch the oocyte. As the tissue-of-origin of the majority of ovarian high-grade serous carcinoma (HGSC), the fallopian tube fimbriae carrying a precursor cancer lesion may also approach the ovulatory site for metastasis. We hypothesize that platelet-derived growth factor (PDGF) in mature follicle fluid (FF) attracts the migration of PDGFR-expressing fimbriae toward the ovulating follicle. We observed that more PDGFR-β was expressed in the distal part than in the proximal parts of the fallopian tube, particularly in stromal cells in the lamina propria. The stromal cells, but not the epithelial cells, from normal fimbriae and fallopian tube HGSC were highly chemotactic to mature FF. The chemotactic activities were positively correlated with PDGF-BB and estradiol levels in FF and were abolished by a blocking antibody of PDGFR-β and by tyrosine kinase inhibitor imatinib. When PDGF-BB/AB was depleted from the FF, more than 80% of chemotaxis activities were diminished. This study suggests an ovarian follicle-directed and PDGF-dependent attraction of fallopian tube fimbriae before ovulation. The same mechanism may also be crucial for the ovarian homing of HGSC, which largely originates in the fimbriae.
Collapse
Affiliation(s)
- Ching-Hua Yeh
- Cervical Cancer Prevention Center, Tzu Chi General Hospital, Hualien, Hualien, 970, Taiwan, ROC
| | - Pao-Chu Chen
- Department of Obstetrics and Gynecology,Tzu Chi General Hospital, Hualien, Hualien, 970, Taiwan, ROC
- Institute of Medical Sciences, Tzu Chi University, Hualien, 970, Taiwan, ROC
| | - Chiu-Hua Chen
- Cervical Cancer Prevention Center, Tzu Chi General Hospital, Hualien, Hualien, 970, Taiwan, ROC
| | - Che-Fang Hsu
- Cervical Cancer Prevention Center, Tzu Chi General Hospital, Hualien, Hualien, 970, Taiwan, ROC
| | - Rui-Len Huang
- Department of Obstetrics and Gynecology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
| | - Dah-Ching Ding
- Cervical Cancer Prevention Center, Tzu Chi General Hospital, Hualien, Hualien, 970, Taiwan, ROC
| | - Tang-Yuan Chu
- Cervical Cancer Prevention Center, Tzu Chi General Hospital, Hualien, Hualien, 970, Taiwan, ROC
- Department of Obstetrics and Gynecology,Tzu Chi General Hospital, Hualien, Hualien, 970, Taiwan, ROC
- Institute of Medical Sciences, Tzu Chi University, Hualien, 970, Taiwan, ROC
- * E-mail:
| |
Collapse
|