1
|
Watanabe T, Hatayama N, Guo M, Yuhara S, Shinoka T. Bridging the Gap: Advances and Challenges in Heart Regeneration from In Vitro to In Vivo Applications. Bioengineering (Basel) 2024; 11:954. [PMID: 39451329 PMCID: PMC11505552 DOI: 10.3390/bioengineering11100954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 10/26/2024] Open
Abstract
Cardiovascular diseases, particularly ischemic heart disease, area leading cause of morbidity and mortality worldwide. Myocardial infarction (MI) results in extensive cardiomyocyte loss, inflammation, extracellular matrix (ECM) degradation, fibrosis, and ultimately, adverse ventricular remodeling associated with impaired heart function. While heart transplantation is the only definitive treatment for end-stage heart failure, donor organ scarcity necessitates the development of alternative therapies. In such cases, methods to promote endogenous tissue regeneration by stimulating growth factor secretion and vascular formation alone are insufficient. Techniques for the creation and transplantation of viable tissues are therefore highly sought after. Approaches to cardiac regeneration range from stem cell injections to epicardial patches and interposition grafts. While numerous preclinical trials have demonstrated the positive effects of tissue transplantation on vasculogenesis and functional recovery, long-term graft survival in large animal models is rare. Adequate vascularization is essential for the survival of transplanted tissues, yet pre-formed microvasculature often fails to achieve sufficient engraftment. Recent studies report success in enhancing cell survival rates in vitro via tissue perfusion. However, the transition of these techniques to in vivo models remains challenging, especially in large animals. This review aims to highlight the evolution of cardiac patch and stem cell therapies for the treatment of cardiovascular disease, identify discrepancies between in vitro and in vivo studies, and discuss critical factors for establishing effective myocardial tissue regeneration in vivo.
Collapse
Affiliation(s)
- Tatsuya Watanabe
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
| | - Naoyuki Hatayama
- Department of Anatomy, Aichi Medical University, Nagakute 480-1195, Japan;
| | - Marissa Guo
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
- Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Satoshi Yuhara
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
| | - Toshiharu Shinoka
- Department of Surgery, Ohio State University, Columbus, OH 43210, USA
- Department of Cardiothoracic Surgery, The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
2
|
Kim SW, Baik S, Hyun J, Lee J, Lim D, Lee TJ, Jeong GJ, Im GB, Seo I, Kim YH, Pang C, Bhang SH. Facile Size Tunable Skin-Adaptive Patch for Accelerating Wound Healing. Adv Healthc Mater 2024:e2304435. [PMID: 39235562 DOI: 10.1002/adhm.202304435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/12/2024] [Indexed: 09/06/2024]
Abstract
Owing to the moist and curved interfaces of skin wounds, enhancing the adhesiveness while maintaining delivery efficacy of biomolecules has drawn significant attention in advanced wound dressings. Despite tremendous trials to load biomolecules with sound adhesiveness, the complicated fabricating processes and abnormal allergic responses that are attributed to chemical moiety-based adhesives remain as major problems. To this end, in this study a one-step fabrication process is developed to manufacture microstructures with both a therapeutic (cylindrical structure for embossed structure human adipose-derived stem cell sheet, ESS) and an adhesive part (octopi-inspired structure of adhesive, OIA), which ESOIA is called. OIA showed the highest adhesion strength in both dry (1.48 N cm-2) and wet pig skin conditions (0.81 N cm-2), maintaining the adhesive properties after repeated attach-detach trials. ESS from the therapeutic part of ESOIA also showed an enhanced angiogenic effect compared with the ones that are normally cultured in vitro. ESS also showed improved in vivo wound healing outcomes following enhanced cell engraftment compared to the cell injection group by means of intact cell-extracellular matrix interactions.
Collapse
Affiliation(s)
- Sung-Won Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sangyul Baik
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dohyun Lim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Tae-Jin Lee
- Department of Medical Biotechnology, Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon-si, 24341, Republic of Korea
| | - Gun-Jae Jeong
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Inwoo Seo
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Changhyun Pang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
3
|
Luca T, Pezzino S, Puleo S, Castorina S. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 2024; 22:764. [PMID: 39143643 PMCID: PMC11323604 DOI: 10.1186/s12967-024-05547-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024] Open
Abstract
Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy.
| | | | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
4
|
Zhang YS, Dolatshahi-Pirouz A, Orive G. Regenerative cell therapy with 3D bioprinting. Science 2024; 385:604-606. [PMID: 39116247 DOI: 10.1126/science.add8593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
In situ additive biomanufacturing of structures may boost regenerative medicine.
Collapse
Affiliation(s)
- Yu Shrike Zhang
- Division of Engineering of Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alireza Dolatshahi-Pirouz
- Technical University of Denmark, DTU Health Tech, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Lyngby, Denmark
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| |
Collapse
|
5
|
Seydel CM, Gonzaga BMDS, Coelho LL, Garzoni LR. Exploring the Dimensions of Pre-Clinical Research: 3D Cultures as an Investigative Model of Cardiac Fibrosis in Chagas Disease. Biomedicines 2024; 12:1410. [PMID: 39061986 PMCID: PMC11274318 DOI: 10.3390/biomedicines12071410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 07/28/2024] Open
Abstract
A three-dimensional (3D) cell culture can more precisely mimic tissues architecture and functionality, being a promising alternative model to study disease pathophysiology and drug screening. Chagas disease (CD) is a neglected parasitosis that affects 7 million people worldwide. Trypanosoma cruzi's (T. cruzi) mechanisms of invasion/persistence continue to be elucidated. Benznidazole (BZ) and Nifurtimox (NF) are trypanocidal drugs with few effects on the clinical manifestations of the chronic disease. Chronic Chagas cardiomyopathy (CCC) is the main manifestation of CD due to its frequency and severity. The development of fibrosis and hypertrophy in cardiac tissue can lead to heart failure and sudden death. Thus, there is an urgent need for novel therapeutic options. Our group has more than fifteen years of expertise using 3D primary cardiac cell cultures, being the first to reproduce fibrosis and hypertrophy induced by T. cruzi infection in vitro. These primary cardiac spheroids exhibit morphological and functional characteristics that are similar to heart tissue, making them an interesting model for studying CD cardiac fibrosis. Here, we aim to demonstrate that our primary cardiac spheroids are great preclinical models which can be used to develop new insights into CD cardiac fibrosis, presenting advances already achieved in the field, including disease modeling and drug screening.
Collapse
Affiliation(s)
| | | | | | - Luciana Ribeiro Garzoni
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (C.M.S.); (B.M.d.S.G.); (L.L.C.)
| |
Collapse
|
6
|
Teixeira Polez R, Huynh N, Pridgeon CS, Valle-Delgado JJ, Harjumäki R, Österberg M. Insights into spheroids formation in cellulose nanofibrils and Matrigel hydrogels using AFM-based techniques. Mater Today Bio 2024; 26:101065. [PMID: 38706731 PMCID: PMC11066555 DOI: 10.1016/j.mtbio.2024.101065] [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: 11/11/2023] [Revised: 03/30/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024] Open
Abstract
The recent FDA decision to eliminate animal testing requirements emphasises the role of cell models, such as spheroids, as regulatory test alternatives for investigations of cellular behaviour, drug responses, and disease modelling. The influence of environment on spheroid formation are incompletely understood, leading to uncertainty in matrix selection for scaffold-based 3D culture. This study uses atomic force microscopy-based techniques to quantify cell adhesion to Matrigel and cellulose nanofibrils (CNF), and cell-cell adhesion forces, and their role in spheroid formation of hepatocellular carcinoma (HepG2) and induced pluripotent stem cells (iPS(IMR90)-4). Results showed different cell behaviour in CNF and Matrigel cultures. Both cell lines formed compact spheroids in CNF but loose cell aggregates in Matrigel. Interestingly, the type of cell adhesion protein, and not the bond strength, appeared to be a key factor in the formation of compact spheroids. The gene expression of E- and N-cadherins, proteins on cell membrane responsible for cell-cell interactions, was increased in CNF culture, leading to formation of compact spheroids while Matrigel culture induced integrin-laminin binding and downregulated E-cadherin expression, resulting in looser cell aggregates. These findings enhance our understanding of cell-biomaterial interactions in 3D cultures and offer insights for improved 3D cell models, culture biomaterials, and applications in drug research.
Collapse
Affiliation(s)
- Roberta Teixeira Polez
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Ngoc Huynh
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Chris S. Pridgeon
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Juan José Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Riina Harjumäki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Monika Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| |
Collapse
|
7
|
Torres Chavez AG, McKenna MK, Balasubramanian K, Riffle L, Patel NL, Kalen JD, St. Croix B, Leen AM, Bajgain P. A dual-luciferase bioluminescence system for the assessment of cellular therapies. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200763. [PMID: 38596291 PMCID: PMC10869576 DOI: 10.1016/j.omton.2024.200763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/17/2023] [Accepted: 01/05/2024] [Indexed: 04/11/2024]
Abstract
Bioluminescence imaging is a well-established platform for evaluating engineered cell therapies in preclinical studies. However, despite the discovery of new luciferases and substrates, optimal combinations to simultaneously monitor two cell populations remain limited. This makes the functional assessment of cellular therapies cumbersome and expensive, especially in preclinical in vivo models. In this study, we explored the potential of using a green bioluminescence-emitting click beetle luciferase, CBG99, and a red bioluminescence-emitting firefly luciferase mutant, Akaluc, together to simultaneously monitor two cell populations. Using various chimeric antigen receptor T cells and tumor pairings, we demonstrate that these luciferases are suitable for real-time tracking of two cell types using 2D and 3D cultures in vitro and experimental models in vivo. Our data show the broad compatibility of this dual-luciferase (duo-luc) system with multiple bioluminescence detection equipment ranging from benchtop spectrophotometers to live animal imaging systems. Although this study focused on investigating complex CAR T cells and tumor cell interactions, this duo-luc system has potential utility for the simultaneous monitoring of any two cellular components-for example, to unravel the impact of a specific genetic variant on clonal dominance in a mixed population of tumor cells.
Collapse
Affiliation(s)
| | - Mary K. McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Lisa Riffle
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Nimit L. Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Brad St. Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pradip Bajgain
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
8
|
Chen S, Wang H, Yang P, Chen S, Ho C, Yang P, Kao Y, Liu S, Chiu H, Lin Y, Chuang E, Huang J, Kao H, Huang C. Schwann cells acquire a repair phenotype after assembling into spheroids and show enhanced in vivo therapeutic potential for promoting peripheral nerve repair. Bioeng Transl Med 2024; 9:e10635. [PMID: 38435829 PMCID: PMC10905550 DOI: 10.1002/btm2.10635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/24/2023] [Accepted: 12/05/2023] [Indexed: 03/05/2024] Open
Abstract
The prognosis for postinjury peripheral nerve regeneration remains suboptimal. Although transplantation of exogenous Schwann cells (SCs) has been considered a promising treatment to promote nerve repair, this strategy has been hampered in practice by the limited availability of SC sources and an insufficient postengraftment cell retention rate. In this study, to address these challenges, SCs were aggregated into spheroids before being delivered to an injured rat sciatic nerve. We found that the three-dimensional aggregation of SCs induced their acquisition of a repair phenotype, as indicated by enhanced levels of c-Jun expression/activation and decreased expression of myelin sheath protein. Furthermore, our in vitro results demonstrated the superior potential of the SC spheroid-derived secretome in promoting neurite outgrowth of dorsal root ganglion neurons, enhancing the proliferation and migration of endogenous SCs, and recruiting macrophages. Moreover, transplantation of SC spheroids into rats after sciatic nerve transection effectively increased the postinjury nerve structure restoration and motor functional recovery rates, demonstrating the therapeutic potential of SC spheroids. In summary, transplantation of preassembled SC spheroids may hold great potential for enhancing the cell delivery efficiency and the resultant therapeutic outcome, thereby improving SC-based transplantation approaches for promoting peripheral nerve regeneration.
Collapse
Affiliation(s)
- Shih‐Heng Chen
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
- School of MedicineCollege of Medicine, Chang Gung UniversityTaoyuanTaiwan
| | - Hsin‐Wen Wang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Pei‐Ching Yang
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Shih‐Shien Chen
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Chia‐Hsin Ho
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Pei‐Ching Yang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Ying‐Chi Kao
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Shao‐Wen Liu
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Han Chiu
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Yu‐Jie Lin
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Er‐Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, International Ph.D. Program in Biomedical Engineering, Taipei Medical UniversityTaipeiTaiwan
- Cell Physiology and Molecular Image Research CenterTaipei Medical University–Wan Fang HospitalTaipeiTaiwan
| | - Jen‐Huang Huang
- Department of Chemical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Huang‐Kai Kao
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
- School of MedicineCollege of Medicine, Chang Gung UniversityTaoyuanTaiwan
| | - Chieh‐Cheng Huang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
9
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
10
|
Garnique ADMB, Machado-Santelli GM. Characterization of 3D NSCLC Cell Cultures with Fibroblasts or Macrophages for Tumor Microenvironment Studies and Chemotherapy Screening. Cells 2023; 12:2790. [PMID: 38132110 PMCID: PMC10742261 DOI: 10.3390/cells12242790] [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: 07/07/2023] [Revised: 08/01/2023] [Accepted: 08/12/2023] [Indexed: 12/23/2023] Open
Abstract
The study of 3D cell culture has increased in recent years as a model that mimics the tumor microenvironment (TME), which is characterized by exhibiting cellular heterogeneity, allowing the modulation of different signaling pathways that enrich this microenvironment. The TME exhibits two main cell populations: cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAM). The aim of this study was to investigate 3D cell cultures of non-small cell lung cancer (NSCLC) alone and in combination with short-term cultured dermal fibroblasts (FDH) and with differentiated macrophages of the THP-1 cell line. Homotypic and heterotypic spheroids were morphologically characterized using light microscopy, immunofluorescence and transmission electron microscopy. Cell viability, cycle profiling and migration assay were performed, followed by the evaluation of the effects of some chemotherapeutic and potential compounds on homotypic and heterotypic spheroids. Both homotypic and heterotypic spheroids of NSCLC were generated with fibroblasts or macrophages. Heterotypic spheroids with fibroblast formed faster, while homotypic ones reached larger sizes. Different cell populations were identified based on spheroid zoning, and drug effects varied between spheroid types. Interestingly, heterotypic spheroids with fibroblasts showed similar responses to the treatment with different compounds, despite being smaller. Cellular viability analysis required multiple methods, since the responses varied depending on the spheroid type. Because of this, the complexity of the spheroid should be considered when analyzing compound effects. Overall, this study contributes to our understanding of the behavior and response of NSCLC cells in 3D microenvironments, providing valuable insights for future research and therapeutic development.
Collapse
Affiliation(s)
| | - Glaucia Maria Machado-Santelli
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, Ave., Prof, Lineu Prestes, 1524, Cidade Universitária, São Paulo 05508-000, SP, Brazil;
| |
Collapse
|
11
|
Koňáriková K, Girašková GM, Žitňanová I, Dvořáková M, Rollerová E, Scsuková S, Bizik J, Janubová M, Muchová J. Biological analyses of the effects of TiO2 and PEG-b-PLA nanoparticles on three-dimensional spheroid-based tumor. Physiol Res 2023; 72:S257-S266. [PMID: 37888969 PMCID: PMC10669953 DOI: 10.33549/physiolres.935152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 12/01/2023] Open
Abstract
The aim of our study was to monitor the antiproliferative/ cytotoxic and genotoxic effects of both, poly(ethylene glycol)-block-poly(lactic acid) (PEG-b-PLA) and titanium dioxide (TiO2) nanoparticles on the tumor (HT-29, MCF-7, U118MG) and healthy (HEK-293T) cell lines during 2D cultivation and during cultivation in the spheroid form (3D cultivation). Cells or spheroids were cultivated with nanoparticles (0.01, 0.1, 1, 10, 50, and 100 ?g/ml) for 72 hours. The cytotoxic effect was determined by the MTT test and the genotoxic effect by the comet assay. We found that 2D cultivation of tumor cell lines with PEG-b-PLA and TiO2 nanoparticles had an anti-proliferative effect on human colon cancer cell line HT-29, human breast cancer cell line MCF-7, human glioma cell line U-118MG during 72h cultivation, but not on control/healthy HEK-293T cells. At the concentrations used, the tested nanoparticles caused no cytotoxic effect on tumor cell lines. Nanoparticles PEG-b-PLA induced significant damage to DNA in HT-29 and MCF-7 cells, while TiO2 nanoparticles in MCF-7 and U-118MG cells. Only PEG-b-PLA nanoparticles caused cytotoxic (IC50 = 7 mikrog/ml) and genotoxic effects on the healthy cell line HEK-293T after 72h cultivation. The cells which were cultivated in spheroid forms were more sensitive to both types of nanoparticles. After 72h cultivation, we observed the cytotoxic effect on both, the tumor and healthy cell lines.
Collapse
Affiliation(s)
- K Koňáriková
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Seo JY, Park SB, Kim SY, Seo GJ, Jang HK, Lee TJ. Acoustic and Magnetic Stimuli-Based Three-Dimensional Cell Culture Platform for Tissue Engineering. Tissue Eng Regen Med 2023; 20:563-580. [PMID: 37052782 PMCID: PMC10313605 DOI: 10.1007/s13770-023-00539-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023] Open
Abstract
In a conventional two-dimensional (2D) culture method, cells are attached to the bottom of the culture dish and grow into a monolayer. These 2D culture methods are easy to handle, cost-effective, reproducible, and adaptable to growing many different types of cells. However, monolayer 2D cell culture conditions are far from those of natural tissue, indicating the need for a three-dimensional (3D) culture system. Various methods, such as hanging drop, scaffolds, hydrogels, microfluid systems, and bioreactor systems, have been utilized for 3D cell culture. Recently, external physical stimulation-based 3D cell culture platforms, such as acoustic and magnetic forces, were introduced. Acoustic waves can establish acoustic radiation force, which can induce suspended objects to gather in the pressure node region and aggregate to form clusters. Magnetic targeting consists of two components, a magnetically responsive carrier and a magnetic field gradient source. In a magnetic-based 3D cell culture platform, cells are aggregated by changing the magnetic force. Magnetic fields can manipulate cells through two different methods: positive magnetophoresis and negative magnetophoresis. Positive magnetophoresis is a way of imparting magnetic properties to cells by labeling them with magnetic nanoparticles. Negative magnetophoresis is a label-free principle-based method. 3D cell structures, such as spheroids, 3D network structures, and cell sheets, have been successfully fabricated using this acoustic and magnetic stimuli-based 3D cell culture platform. Additionally, fabricated 3D cell structures showed enhanced cell behavior, such as differentiation potential and tissue regeneration. Therefore, physical stimuli-based 3D cell culture platforms could be promising tools for tissue engineering.
Collapse
Affiliation(s)
- Ju Yeon Seo
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
- Department of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Song Bin Park
- Department of Bio-Health Technology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Seo Yeon Kim
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Gyeong Jin Seo
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Hyeon-Ki Jang
- Division of Chemical Engineering and Bioengineering, College of Art Culture and Engineering, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Tae-Jin Lee
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
| |
Collapse
|
13
|
Sousa LR, Oliveira AGS, Arantes A, Junqueira JGM, Alexandre GP, Severino VGP, Reis RM, Kim B, Ribeiro RIMA. Acetogenins-Rich Fractions of Annona coriacea Suppress Human Glioblastoma Viability and Migration by Regulating Necroptosis and MMP-2 Activity In Vitro. Molecules 2023; 28:molecules28093809. [PMID: 37175219 PMCID: PMC10179884 DOI: 10.3390/molecules28093809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 05/15/2023] Open
Abstract
Glioblastoma (GBM) is an incurable primary brain tumor with a poor prognosis. Resection, radiation therapy, and temozolomide (TMZ) are insufficient to increase survival, making the treatment limited. Thus, the search for more effective and specific treatments is essential, making plants a promising source for elucidating new anti-glioblastoma compounds. Accordingly, this study investigated the effects of four fractions of hexane and ethyl acetate extract of Annona coriacea Mart., enriched with acetogenins, against GBM cell lines. All four fractions were selectively cytotoxic to GBM cells when compared to TMZ. Moreover, A. coriacea fractions delayed cell migration; reduced cytoplasmic projections, the metalloproteinase 2 (MMP-2) activity; and induced morphological changes characteristic of necroptosis, possibly correlated with the increase in receptor-interacting protein kinase 1 and 3 (RIP-1 and RIP-3), apoptosis-inducing factor (AIF), and the non-activation of cleaved caspase 8. The present findings reinforce that fractions of A. coriacea Mart. should be considered for more studies focusing treatment of GBM.
Collapse
Affiliation(s)
- Lorena R Sousa
- Experimental Pathology Laboratory, Federal University of São João del Rei (UFSJ), 400, Sebastião Gonçalves Coelho, Chanadour, Divinópolis 35501-296, MG, Brazil
| | - Ana Gabriela S Oliveira
- Experimental Pathology Laboratory, Federal University of São João del Rei (UFSJ), 400, Sebastião Gonçalves Coelho, Chanadour, Divinópolis 35501-296, MG, Brazil
| | - Antônio Arantes
- Experimental Pathology Laboratory, Federal University of São João del Rei (UFSJ), 400, Sebastião Gonçalves Coelho, Chanadour, Divinópolis 35501-296, MG, Brazil
| | - João Gabriel M Junqueira
- Institute of Chemistry, Federal University of Goiás (UFG), University Campus, Goiânia 74968-755, GO, Brazil
| | - Gerso P Alexandre
- Institute of Chemistry, Federal University of Goiás (UFG), University Campus, Goiânia 74968-755, GO, Brazil
| | - Vanessa G P Severino
- Institute of Chemistry, Federal University of Goiás (UFG), University Campus, Goiânia 74968-755, GO, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Bonglee Kim
- College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Rosy I M A Ribeiro
- Experimental Pathology Laboratory, Federal University of São João del Rei (UFSJ), 400, Sebastião Gonçalves Coelho, Chanadour, Divinópolis 35501-296, MG, Brazil
| |
Collapse
|
14
|
Robledo F, González-Hodar L, Tapia P, Figueroa AM, Ezquer F, Cortés V. Spheroids derived from the stromal vascular fraction of adipose tissue self-organize in complex adipose organoids and secrete leptin. Stem Cell Res Ther 2023; 14:70. [PMID: 37024989 PMCID: PMC10080976 DOI: 10.1186/s13287-023-03262-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Adipose tissue-derived stromal vascular fraction (SVF) harbors multipotent cells with potential therapeutic relevance. We developed a method to form adipose spheroids (AS) from the SVF with complex organoid structure and enhanced leptin secretion upon insulin stimulation. METHODS SVF was generated from the interscapular brown adipose tissue of newborn mice. Immunophenotype and stemness of cultured SVF were determined by flow cytometry and in vitro differentiation, respectively. Spheroids were generated in hanging drops and non-adherent plates and compared by morphometric methods. The adipogenic potential was compared between preadipocyte monolayers and spheroids. Extracellular leptin was quantified by immunoassay. Lipolysis was stimulated with isoprenaline and quantified by colorimetric methods. AS viability and ultrastructure were determined by confocal and transmission electron microscopy analyses. RESULTS Cultured SVF contained Sca1 + CD29 + CD44 + CD11b- CD45- CD90- cells with adipogenic and chondrogenic but no osteogenic potential. Culture on non-adherent plates yielded the highest quantity and biggest size of spheroids. Differentiation of AS for 15 days in a culture medium supplemented with insulin and rosiglitazone resulted in greater Pparg, Plin1, and Lep expression compared to differentiated adipocytes monolayers. AS were viable and maintained leptin secretion even in the absence of adipogenic stimulation. Glycerol release after isoprenaline stimulation was higher in AS compared to adipocytes in monolayers. AS were composed of outer layers of unilocular mature adipocytes and an inner structure composed of preadipocytes, immature adipocytes and an abundant loose extracellular matrix. CONCLUSION Newborn mice adipose SVF can be efficiently differentiated into leptin-secreting AS. Prolonged stimulation with insulin and rosiglitazone allows the formation of structurally complex adipose organoids able to respond to adrenergic lipolytic stimulation.
Collapse
Affiliation(s)
- Fermín Robledo
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lila González-Hodar
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Tapia
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ana-María Figueroa
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, School of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Víctor Cortés
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
15
|
Otsuka H. Nanofabrication Technologies to Control Cell and Tissue Function in Three-Dimension. Gels 2023; 9:gels9030203. [PMID: 36975652 PMCID: PMC10048556 DOI: 10.3390/gels9030203] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/29/2023] Open
Abstract
In the 2000s, advances in cellular micropatterning using microfabrication contributed to the development of cell-based biosensors for the functional evaluation of newly synthesized drugs, resulting in a revolutionary evolution in drug screening. To this end, it is essential to utilize cell patterning to control the morphology of adherent cells and to understand contact and paracrine-mediated interactions between heterogeneous cells. This suggests that the regulation of the cellular environment by means of microfabricated synthetic surfaces is not only a valuable endeavor for basic research in biology and histology, but is also highly useful to engineer artificial cell scaffolds for tissue regeneration. This review particularly focuses on surface engineering techniques for the cellular micropatterning of three-dimensional (3D) spheroids. To establish cell microarrays, composed of a cell adhesive region surrounded by a cell non-adherent surface, it is quite important to control a protein-repellent surface in the micro-scale. Thus, this review is focused on the surface chemistries of the biologically inspired micropatterning of two-dimensional non-fouling characters. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single-cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., fibers and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. These important approaches to cell engineering result in their applications to tissue regeneration, where the cell-biomaterial composite is injected into diseased area. This approach allows the operating surgeon to implant the cell and polymer combinations with minimum invasiveness. The polymers utilized in hydrogels are structurally similar to components of the extracellular matrix in vivo, and are considered biocompatible. This review will provide an overview of the critical design to make hydrogels when used as cell scaffolds for tissue engineering. In addition, the new strategy of injectable hydrogel will be discussed as future directions.
Collapse
Affiliation(s)
- Hidenori Otsuka
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, 1-3 Kagurazaka, Shinjuku-ku, Tokyo 162-8601, Japan
| |
Collapse
|
16
|
Kim W, Kim G. Hybrid cell constructs consisting of bioprinted cell-spheroids. Bioeng Transl Med 2023; 8:e10397. [PMID: 36925682 PMCID: PMC10013803 DOI: 10.1002/btm2.10397] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/18/2022] [Accepted: 08/16/2022] [Indexed: 11/11/2022] Open
Abstract
Bioprinted cell constructs have been investigated for regeneration of various tissues. However, poor cell-cell interactions have limited their utility. Although cell-spheroids offer an alternative for efficient cell-cell interactions, they complicate bioprinting. Here, we introduce a new cell-printing process, fabricating cell-spheroids and cell-loaded constructs together without preparation of cell-spheroids in advance. Cells in mineral oil droplets self-assembled to form cell-spheroids due to the oil-aqueous interaction, exhibiting similar biological functions to the conventionally prepared cell-spheroids. By controlling printing parameters, spheroid diameter and location could be manipulated. To demonstrate the feasibility of this process, we fabricated hybrid cell constructs, consisting of endothelial cell-spheroids and stem cells loaded decellularized extracellular matrix/β-tricalcium phosphate struts for regenerating vascularized bone. The hybrid cell constructs exhibited strong angiogenic/osteogenic activities as a result of increased secretion of signaling molecules and synergistic crosstalk between the cells.
Collapse
Affiliation(s)
- WonJin Kim
- Department of Biomechatronic Engineering, College of Biotechnology and BioengineeringSungkyunkwan University (SKKU)SuwonSouth Korea
| | - GeunHyung Kim
- Department of Biomechatronic Engineering, College of Biotechnology and BioengineeringSungkyunkwan University (SKKU)SuwonSouth Korea
- Biomedical Institute for Convergence at SKKU (BICS)Sungkyunkwan UniversitySuwonSouth Korea
| |
Collapse
|
17
|
Jiang Z, Xu Y, Fu M, Zhu D, Li N, Yang G. Genetically modified cell spheroids for tissue engineering and regenerative medicine. J Control Release 2023; 354:588-605. [PMID: 36657601 DOI: 10.1016/j.jconrel.2023.01.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023]
Abstract
Cell spheroids offer cell-to-cell interactions and show advantages in survival rate and paracrine effect to solve clinical and biomedical inquiries ranging from tissue engineering and regenerative medicine to disease pathophysiology. Therefore, cell spheroids are ideal vehicles for gene delivery. Genetically modified spheroids can enhance specific gene expression to promote tissue regeneration. Gene deliveries to cell spheroids are via viral vectors or non-viral vectors. Some new technologies like CRISPR/Cas9 also have been used in genetically modified methods to deliver exogenous gene to the host chromosome. It has been shown that genetically modified cell spheroids had the potential to differentiate into bone, cartilage, vascular, nerve, cardiomyocytes, skin, and skeletal muscle as well as organs like the liver to replace the diseased organ in the animal and pre-clinical trials. This article reviews the recent articles about genetically modified spheroid cells and explains the fabrication, applications, development timeline, limitations, and future directions of genetically modified cell spheroid.
Collapse
Affiliation(s)
- Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yi Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Na Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
18
|
Kim W, Gwon Y, Park S, Kim H, Kim J. Therapeutic strategies of three-dimensional stem cell spheroids and organoids for tissue repair and regeneration. Bioact Mater 2023; 19:50-74. [PMID: 35441116 PMCID: PMC8987319 DOI: 10.1016/j.bioactmat.2022.03.039] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) stem cell culture systems have attracted considerable attention as a way to better mimic the complex interactions between individual cells and the extracellular matrix (ECM) that occur in vivo. Moreover, 3D cell culture systems have unique properties that help guide specific functions, growth, and processes of stem cells (e.g., embryogenesis, morphogenesis, and organogenesis). Thus, 3D stem cell culture systems that mimic in vivo environments enable basic research about various tissues and organs. In this review, we focus on the advanced therapeutic applications of stem cell-based 3D culture systems generated using different engineering techniques. Specifically, we summarize the historical advancements of 3D cell culture systems and discuss the therapeutic applications of stem cell-based spheroids and organoids, including engineering techniques for tissue repair and regeneration.
Collapse
Affiliation(s)
- Woochan Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yonghyun Gwon
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunho Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyoseong Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jangho Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| |
Collapse
|
19
|
Griffin KH, Fok SW, Kent Leach J. Strategies to capitalize on cell spheroid therapeutic potential for tissue repair and disease modeling. NPJ Regen Med 2022; 7:70. [PMID: 36494368 PMCID: PMC9734656 DOI: 10.1038/s41536-022-00266-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Cell therapies offer a tailorable, personalized treatment for use in tissue engineering to address defects arising from trauma, inefficient wound repair, or congenital malformation. However, most cell therapies have achieved limited success to date. Typically injected in solution as monodispersed cells, transplanted cells exhibit rapid cell death or insufficient retention at the site, thereby limiting their intended effects to only a few days. Spheroids, which are dense, three-dimensional (3D) aggregates of cells, enhance the beneficial effects of cell therapies by increasing and prolonging cell-cell and cell-matrix signaling. The use of spheroids is currently under investigation for many cell types. Among cells under evaluation, spheroids formed of mesenchymal stromal cells (MSCs) are particularly promising. MSC spheroids not only exhibit increased cell survival and retained differentiation, but they also secrete a potent secretome that promotes angiogenesis, reduces inflammation, and attracts endogenous host cells to promote tissue regeneration and repair. However, the clinical translation of spheroids has lagged behind promising preclinical outcomes due to hurdles in their formation, instruction, and use that have yet to be overcome. This review will describe the current state of preclinical spheroid research and highlight two key examples of spheroid use in clinically relevant disease modeling. It will highlight techniques used to instruct the phenotype and function of spheroids, describe current limitations to their use, and offer suggestions for the effective translation of cell spheroids for therapeutic treatments.
Collapse
Affiliation(s)
- Katherine H Griffin
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
- School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Shierly W Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA.
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
20
|
Li L, Sheng Q, Zeng H, Li W, Wang Q, Ma G, Qiu M, Zhang W, Shan C. Engineering a functional thyroid as a potential therapeutic substitute for hypothyroidism treatment: A systematic review. Front Endocrinol (Lausanne) 2022; 13:1065410. [PMID: 36531472 PMCID: PMC9755335 DOI: 10.3389/fendo.2022.1065410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
Background Hypothyroidism is a common hormone deficiency disorder. Although hormone supplemental therapy can be easily performed by daily levothyroxine administration, a proportion of patients suffer from persisting complaints due to unbalanced hormone levels, leaving room for new therapeutic strategies, such as tissue engineering and regenerative medicine. Methods Electronic searches of databases for studies of thyroid regeneration or thyroid organoids were performed. A systematic review including both in vitro and in vivo models of thyroid regenerative medicine was conducted. Results Sixty-six independent studies published between 1959 and May 1st, 2022 were included in the current systematic review. Among these 66 studies, the most commonly involved species was human (19 studies), followed by mouse (18 studies), swine (14 studies), rat (13 studies), calf/bovine (4 studies), sheep/lamb (4 studies) and chick (1 study). In addition, in these experiments, the most frequently utilized tissue source was adult thyroid tissue (46 studies), followed by embryonic stem cells (ESCs)/pluripotent stem cells (iPSCs) (10 studies), rat thyroid cell lines (7 studies), embryonic thyroid tissue (2 studies) and newborn or fetal thyroid tissue (2 studies). Sixty-three studies reported relevant thyroid follicular regeneration experiments in vitro, while 21 studies showed an in vivo experiment section that included transplanting engineered thyroid tissue into recipients. Together, 12 studies were carried out using 2D structures, while 50 studies constructed 3D structures. Conclusions Each aspect of thyroid regenerative medicine was comprehensively described in this review. The recovery of optimal hormonal equilibrium by the transplantation of an engineered functional thyroid holds great therapeutic promise.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei Zhang
- Department of Thyroid and Breast Surgery of Changzheng Hospital Affiliated with Naval Military Medical University, Shanghai, China
| | - Chengxiang Shan
- Department of Thyroid and Breast Surgery of Changzheng Hospital Affiliated with Naval Military Medical University, Shanghai, China
| |
Collapse
|
21
|
Tsujimura M, Kusamori K, Takamura K, Ito T, Kaya T, Shimizu K, Konishi S, Nishikawa M. Quality evaluation of cell spheroids for transplantation by monitoring oxygen consumption using an on-chip electrochemical device. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2022; 36:e00766. [PMID: 36245695 PMCID: PMC9562952 DOI: 10.1016/j.btre.2022.e00766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/19/2022] [Accepted: 10/01/2022] [Indexed: 11/18/2022]
Abstract
Three-dimensional cell spheroids are superior cell-administration form for cell-based therapy which generally exhibit superior functionality and long-term survival after transplantation. Here, we nondestructively measured the oxygen consumption rate of cell spheroids using an on-chip electrochemical device (OECD) and examined whether this rate can be used as a marker to estimate the quality of cell spheroids. Cell spheroids containing NanoLuc luciferase-expressing mouse mesenchymal stem cell line C3H10T1/2 (C3H10T1/2/Nluc) were prepared. Spheroids of high or low quality were prepared by altering the medium change frequency. After transplantation into mice, the high-quality C3H10T1/2/Nluc spheroids exhibited a higher survival rate than the low-quality ones. The oxygen consumption rate of the high-quality C3H10T1/2/Nluc spheroids was maintained at high levels, whereas that of the low-quality spheroids decreased with time. These results indicate that OECD-based measurement of the oxygen consumption rate can be used to estimate the quality of cell spheroids without destructive analysis of the spheroids.
Collapse
Affiliation(s)
- Mari Tsujimura
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
- Corresponding author.
| | - Kodai Takamura
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Temmei Ito
- KONICA MINOLTA, INC., No.1 Sakura-machi, Hino-shi, Tokyo, 191-8511, Japan
| | - Takatoshi Kaya
- KONICA MINOLTA, INC., No.1 Sakura-machi, Hino-shi, Tokyo, 191-8511, Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8603, Japan
| | - Satoshi Konishi
- Department of Mechanical Engineering, Graduate School of Science and Engineering, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| |
Collapse
|
22
|
Correlation of the regenerative potential of dermal fibroblasts in 2D culture with the biological properties of fibroblast-derived tissue spheroids. Cell Tissue Res 2022; 390:453-464. [PMID: 36129531 DOI: 10.1007/s00441-022-03690-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/14/2022] [Indexed: 11/02/2022]
Abstract
In situ 3D bioprinting is a new emerging therapeutic modality for treating human skin diseases. The tissue spheroids have been previously suggested as a powerful tool in rapidly expanding bioprinting technology. It has been demonstrated that the regenerative potential of human dermal fibroblasts could be quantitatively evaluated in 2D cell culture and confirmed after implantation in vivo. However, the development of unbiassed quantitative criteria of the regenerative potential of 3D tissue spheroids in vitro before their in situ bioprinting remains to be investigated. Here it has been demonstrated for the first time that specific correlations exist between the regenerative potential of human dermal fibroblasts cultured in vitro as 2D cell monolayer with biological properties of 3D tissue spheroids fabricated from these fibroblasts. In vitro assessment of biological properties included diameter, spreading and fusion kinetics, and biomechanical properties of 3D tissue spheroids. This comprehensive characterization could be used to predict tissue spheroids' regenerative potential in vivo.
Collapse
|
23
|
Park J, An G, Lim W, Song G. Dinitramine induces implantation failure by cell cycle arrest and mitochondrial dysfunction in porcine trophectoderm and luminal epithelial cells. JOURNAL OF HAZARDOUS MATERIALS 2022; 435:128927. [PMID: 35489316 DOI: 10.1016/j.jhazmat.2022.128927] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 06/14/2023]
Abstract
The herbicide market is growing rapidly, as weed control is a significant challenge in agriculture. Many studies have reported the toxicity of herbicides to non-target organisms. Dinitramine is a dinitroaniline herbicide that is particularly toxic to aquatic organisms. However, little is known about the effects of dinitramine on the female reproductive system. Therefore, in the present study, we utilized porcine trophectoderm (pTr) cells and porcine endometrial luminal epithelial (pLE) cells to verify the reproductive toxicity of dinitramine. Dinitramine reduced the viability of both cell types, by triggering cell cycle arrest, especially at the sub-G1 phase, and increasing apoptosis, inhibiting DNA replication. Dinitramine disrupted intracellular calcium homeostasis and induced oxidative stress by producing reactive oxygen species, leading to the loss of mitochondrial membrane potential and alteration of mitochondrial respiration. Mitogen-activated protein kinase pathways were altered, and migration decreased in pTr and pLE cells after dinitramine treatment; the expression of pregnancy-related genes in these cells was decreased. Thus, dinitramine reduced the viability and migratory capacity of both cell types, and this could interrupt the early stages of pregnancy.
Collapse
Affiliation(s)
- Junho Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
24
|
Mohr E, Thum T, Bär C. Accelerating Cardiovascular Research: Recent Advances in Translational 2D and 3D Heart Models. Eur J Heart Fail 2022; 24:1778-1791. [PMID: 35867781 DOI: 10.1002/ejhf.2631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/30/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022] Open
Abstract
In vitro modelling the complex (patho-) physiological conditions of the heart is a major challenge in cardiovascular research. In recent years, methods based on three-dimensional (3D) cultivation approaches have steadily evolved to overcome the major limitations of conventional adherent monolayer cultivation (2D). These 3D approaches aim to study, reproduce or modify fundamental native features of the heart such as tissue organization and cardiovascular microenvironment. Therefore, these systems have great potential for (patient-specific) disease research, for the development of new drug screening platforms, and for the use in regenerative and replacement therapy applications. Consequently, continuous improvement and adaptation is required with respect to fundamental limitations such as cardiomyocyte maturation, scalability, heterogeneity, vascularization, and reproduction of native properties. In this review, 2D monolayer culturing and the 3D in vitro systems of cardiac spheroids, organoids, engineered cardiac microtissue and bioprinting as well as the ex vivo technique of myocardial slicing are introduced with their basic concepts, advantages, and limitations. Furthermore, recent advances of various new approaches aiming to extend as well as to optimize these in vitro and ex vivo systems are presented. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Elisa Mohr
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| |
Collapse
|
25
|
Liao FC, Wang YK, Cheng MY, Tu TY. A Preliminary Investigation of Embedding In Vitro HepaRG Spheroids into Recombinant Human Collagen Type I for the Promotion of Liver Differentiation. Polymers (Basel) 2022; 14:polym14091923. [PMID: 35567092 PMCID: PMC9103061 DOI: 10.3390/polym14091923] [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: 04/18/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022] Open
Abstract
Background: In vitro three-dimensional (3D) hepatic spheroid culture has shown great promise in toxicity testing because it better mimics the cell–cell and cell–matrix interactions found in in vivo conditions than that of the traditional two-dimensional (2D) culture. Despite embedding HepaRG spheroids with collagen type I (collagen I) extracellular matrix (ECM) revealed a much better differentiation capability, almost all the collagen utilized in in vitro hepatocytes cultures is animal-derived collagen that may limit its use in human toxicity testing. Method: Here, a preliminary investigation of HepaRG cells cultured in different dimensionalities and with the addition of ECM was performed. Comparisons of conventional 2D culture with 3D spheroid culture were performed based on their functional or structural differences over 7 days. Rat tail collagen (rtCollagen) I and recombinant human collagen (rhCollagen) I were investigated for their ability in promoting HepaRG spheroid differentiation. Results: An immunofluorescence analysis of the hepatocyte-specific functional protein albumin suggested that HepaRG spheroids demonstrated better hepatic function than spheroids from 2D culture, and the function of HepaRG spheroids improved in a time-dependent manner. The fluorescence intensities per unit area of spheroids formed by 1000 cells on days 7 and 10 were 25.41 and 45.38, respectively, whereas almost undetectable fluorescence was obtained with 2D cells. In addition, the embedding of HepaRG spheroids into rtCollagen and rhCollagen I showed that HepaRG differentiation can be accelerated relative to the differentiation of spheroids grown in suspension, demonstrating the great promise of HepaRG spheroids. Conclusions: The culture conditions established in this study provide a potentially novel alternative for promoting the differentiation of HepaRG spheroids into mature hepatocytes through a collagen-embedded in vitro liver spheroid model. This culture method is envisioned to provide insights for future drug toxicology.
Collapse
Affiliation(s)
- Fang-Chun Liao
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 70101, Taiwan; (F.-C.L.); (M.-Y.C.)
| | - Yang-Kao Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Ming-Yang Cheng
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 70101, Taiwan; (F.-C.L.); (M.-Y.C.)
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 70101, Taiwan; (F.-C.L.); (M.-Y.C.)
- Medical Device Innovation Center, National Cheng Kung University, Tainan 70101, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 70101, Taiwan
- Correspondence:
| |
Collapse
|
26
|
Svistushkin MV, Kotova S, Shpichka A, Starostina S, Shekhter A, Bikmulina P, Nikiforova A, Zolotova A, Royuk V, Kochetkov PA, Timashev S, Fomin V, Vosough M, Svistushkin V, Timashev P. Stem cell therapy for vocal fold regeneration after scarring: a review of experimental approaches. Stem Cell Res Ther 2022; 13:176. [PMID: 35505357 PMCID: PMC9066721 DOI: 10.1186/s13287-022-02853-9] [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: 11/24/2021] [Accepted: 04/13/2022] [Indexed: 11/12/2022] Open
Abstract
This review aims at becoming a guide which will help to plan the experimental design and to choose adequate methods to assess the outcomes when testing cell-based products in the treatment of the damaged vocal folds. The requirements to preclinical trials of cell-based products remain rather hazy and dictated by the country regulations. Most parameters like the way the cells are administered, selection of the cell source, selection of a carrier, and design of in vivo studies are decided upon by each research team and may differ essentially between studies. The review covers the methodological aspects of preclinical studies such as experimental models, characterization of cell products, assessment of the study outcome using molecular, morphological and immunohistochemical analyses, as well as measuring the tissue physical properties. The unified recommendations to perform preclinical trials could significantly facilitate the translation of cell-based products into the clinical practice.
Collapse
Affiliation(s)
- Mikhail V Svistushkin
- Department for ENT Diseases, Sechenov University, Moscow, Russia.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
| | - Svetlana Kotova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia Shpichka
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia. .,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia. .,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia.
| | | | - Anatoliy Shekhter
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Polina Bikmulina
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Anna Nikiforova
- Department for ENT Diseases, Sechenov University, Moscow, Russia
| | - Anna Zolotova
- Department for ENT Diseases, Sechenov University, Moscow, Russia
| | - Valery Royuk
- University Hospital No 1, Sechenov University, Moscow, Russia
| | - P A Kochetkov
- Department for ENT Diseases, Sechenov University, Moscow, Russia
| | - Serge Timashev
- National Research Nuclear University «MEPhI», Moscow, Russia
| | - Victor Fomin
- Department of Internal Medicine No 1, Sechenov University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | | | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia. .,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia. .,Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, Russia. .,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
27
|
Lu Y, Yu CH, Yang G, Sun N, Jiang F, Zhou M, Wu X, Luo J, Huang C, Zhang W, Jiang X. A rapidly magnetically assembled stem cell microtissue with "hamburger" architecture and enhanced vascularization capacity. Bioact Mater 2021; 6:3756-3765. [PMID: 33898876 PMCID: PMC8044908 DOI: 10.1016/j.bioactmat.2021.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 12/19/2022] Open
Abstract
With the development of magnetic manipulation technology based on magnetic nanoparticles (MNPs), scaffold-free microtissues can be constructed utilizing the magnetic attraction of MNP-labeled cells. The rapid in vitro construction and in vivo vascularization of microtissues with complex hierarchical architectures are of great importance to the viability and function of stem cell microtissues. Endothelial cells are indispensable for the formation of blood vessels and can be used in the prevascularization of engineered tissue constructs. Herein, safe and rapid magnetic labeling of cells was achieved by incubation with MNPs for 1 h, and ultrathick scaffold-free microtissues with different sophisticated architectures were rapidly assembled, layer by layer, in 5 min intervals. The in vivo transplantation results showed that in a stem cell microtissue with trisection architecture, the two separated human umbilical vein endothelial cell (HUVEC) layers would spontaneously extend to the stem cell layers and connect with each other to form a spatial network of functional blood vessels, which anastomosed with the host vasculature. The "hamburger" architecture of stem cell microtissues with separated HUVEC layers could promote vascularization and stem cell survival. This study will contribute to the construction and application of structural and functional tissues or organs in the future.
Collapse
Affiliation(s)
- Yuezhi Lu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chun-Hua Yu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Guangzheng Yang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ningjia Sun
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fei Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Mingliang Zhou
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaolin Wu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jiaxin Luo
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedical Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
28
|
Mizukami Y, Takahashi Y, Shimizu K, Konishi S, Takakura Y, Nishikawa M. Calcium Peroxide-Containing Polydimethylsiloxane-Based Microwells for Inhibiting Cell Death in Spheroids through Improved Oxygen Supply. Biol Pharm Bull 2021; 44:1458-1464. [PMID: 34602554 DOI: 10.1248/bpb.b21-00269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Multicellular spheroids are expected to be used for in vivo-like tissue models and cell transplantation. Microwell devices are useful for the fabrication of multicellular spheroids to improve productivity and regulate their size. However, the high cell density in microwell devices leads to accelerated cell death. In this study, we developed O2-generating microwells by incorporating calcium peroxide (CaO2) into polydimethylsiloxane (PDMS)-based microwells. The CaO2-containing PDMS was shown to generate O2 for 3 d. Then, CaO2-containing PDMS was used to fabricate O2-generating microwells using a micro-molding technique. When human hepatocellular carcinoma (HepG2) spheroids were prepared using the conventional microwells, the O2 concentration in the culture medium reduced to approx. 67% of the cell-free level. In contrast, the O2-generating microwells maintained O2 at constant levels. The HepG2 spheroids prepared using the O2-generating microwells had a larger number of live cells than those prepared using the conventional microwells. In addition, the O2-generating microwells rescued hypoxia in the HepG2 spheroids and increased cell viability. Lastly, the O2-generating microwells were also useful for the preparation of multicellular spheroids of other cell types (i.e., MIN6, B16-BL6, and adipose-derived stem cells) with high cell viability. These results showed that the O2-generating microwells are useful for preparing multicellular spheroids with high cell viability.
Collapse
Affiliation(s)
- Yuya Mizukami
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University
| | - Satoshi Konishi
- Department of Mechanical Engineering, Graduate School of Science and Engineering, Ritsumeikan University
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Makiya Nishikawa
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University.,Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| |
Collapse
|
29
|
Maciel MM, Correia TR, Henriques M, Mano JF. Microparticles orchestrating cell fate in bottom-up approaches. Curr Opin Biotechnol 2021; 73:276-281. [PMID: 34597880 DOI: 10.1016/j.copbio.2021.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 12/30/2022]
Abstract
The modulation of cells in tissue formation is still one of the hardest tasks to achieve in Tissue Engineering. To control the cell response when undergoing their normal functions such as adhesion, differentiation, assembly, or maturation is vital the development of more successful solutions. Herein, we discuss how microparticles are being overlooked in their potential for controlling the cellular response. Until now, their role was quite often restricted to a reservoir of chemical compounds or as carriers for cell expansion. Nevertheless, microparticles design with the introduction of biophysical and biochemical cues can effectively modulate cell response.
Collapse
Affiliation(s)
- Marta M Maciel
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Complexo de Laboratórios Tecnológicos, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Tiago R Correia
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Complexo de Laboratórios Tecnológicos, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Mariana Henriques
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - João F Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Complexo de Laboratórios Tecnológicos, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
30
|
Shin DS, Anseth KS. Recent advances in 3D models of tumor invasion. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 19:100310. [PMID: 34308009 PMCID: PMC8294077 DOI: 10.1016/j.cobme.2021.100310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This review presents recent advances in the design of in vitro cancer models to study tumor cell migration, metastasis, and invasion in three-dimensions (3D). These cancer models are divided into two categories based on the biophysiological processes and structures simulated, namely (i) spheroid invasion models or (ii) vascularization models. Some recent advances to spheroid invasion models include new methods to make them amenable to high-throughput settings. In vascularization models, cancer cell extravasation, intravasation, and angiogenesis have been emulated. Finally, 3D bioprinting and microfluidic technologies are allowing researchers to recapitulate some of the complex architectural and microenvironmental changes that can drive cancer cells migration from the extracellular matrix and basement membrane to blood vessels.
Collapse
Affiliation(s)
- Della S. Shin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA 80303
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA 80303
| |
Collapse
|
31
|
Gilmour AD, Reshamwala R, Wright AA, Ekberg JAK, St John JA. Optimizing Olfactory Ensheathing Cell Transplantation for Spinal Cord Injury Repair. J Neurotrauma 2021; 37:817-829. [PMID: 32056492 DOI: 10.1089/neu.2019.6939] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell transplantation constitutes an important avenue for development of new treatments for spinal cord injury (SCI). These therapies are aimed at supporting neural repair and/or replacing lost cells at the injury site. To date, various cell types have been trialed, with most studies focusing on different types of stem cells or glial cells. Here, we review commonly used cell transplantation approaches for spinal cord injury (SCI) repair, with focus on transplantation of olfactory ensheathing cells (OECs), the glial cells of the primary olfactory nervous system. OECs are promising candidates for promotion of neural repair given that they support continuous regeneration of the olfactory nerve that occurs throughout life. Further, OECs can be accessed from the nasal mucosa (olfactory neuroepithelium) at the roof of the nasal cavity and can be autologously transplanted. OEC transplantation has been trialed in many animal models of SCI, as well as in human clinical trials. While several studies have been promising, outcomes are variable and the method needs improvement to enhance aspects such as cell survival, integration, and migration. As a case study, we include the approaches used by our team (the Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia) to address the current problems with OEC transplantation and discuss how the therapeutic potential of OEC transplantation can be improved. Our approach includes discovery research to improve our knowledge of OEC biology, identifying natural and synthetic compounds to stimulate the neural repair properties of OECs, and designing three-dimensional cell constructs to create stable and transplantable cell structures.
Collapse
Affiliation(s)
- Aaron D Gilmour
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Ronak Reshamwala
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Alison A Wright
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Jenny A K Ekberg
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - James A St John
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
32
|
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy.
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | | |
Collapse
|
33
|
Lee NH, Bayaraa O, Zechu Z, Kim HS. Biomaterials-assisted spheroid engineering for regenerative therapy. BMB Rep 2021; 54:356-367. [PMID: 34154700 PMCID: PMC8328824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/23/2021] [Accepted: 06/15/2021] [Indexed: 04/04/2024] Open
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy. [BMB Reports 2021; 54(7): 356-367].
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
34
|
Zhang J, Xu W, Li C, Meng F, Guan Y, Liu X, Zhao J, Peng J, Wang Y. Tissue Engineering Microtissue: Construction, Optimization, and Application. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:393-404. [PMID: 33719547 DOI: 10.1089/ten.teb.2020.0370] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Until now, there is no clear definition of microtissue; it usually refers to the microtissue formed by the aggregation of seed cells under the action of cell-cell or cell-extracellular matrix (ECM). Compared with traditional cell monolayer culture, cells are cultivated into a three-dimensional microstructure in a specific way. The microstructure characteristics of microtissue are similar to natural tissues and can promote cell proliferation and differentiation. Therefore, it has a broader range of biomedical applications in tissue engineering. The traditional tissue engineering strategy is to add high-density seed cells and biomolecules on a preformed scaffold to construct a tissue engineering graft. However, due to the destruction of the ECM of the cells cultured in a monolayer during the digestion process with trypsin, the uneven distribution of the cells in the scaffold, and the damage of various adverse factors after the cells are implanted in the scaffold, this strategy is often ineffective, and the subsequent applications still face challenges. This article reviews the latest researches of a new strategy-tissue engineering microtissue strategy; discuss several traditional construction methods, structure, and function optimization; and practical application of microtissue. The review aims to provide a reference for future research on tissue engineering microtissue. Impact statement The traditional tissue engineering strategies have several disadvantages, researchers have conducted extensive research on tissue engineering microtissues in recent years, and they make significant progress. Microtissue is a kind of microtissue with three-dimensional structure, its microstructure is similar to that of natural tissue. In addition, microtissue implantation can protect cells from mechanical interference, inflammation, and other adverse factors. Furthermore, it improves the survival rate of cells and the therapeutic effect of tissue-engineered grafts. However, the practical conditions, advantages, and disadvantages of tissue engineering microtissue have not been fully elucidated. The purpose of this review is to discuss the latest research progress of microtissue and provide a reference for future research.
Collapse
Affiliation(s)
- Jian Zhang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Wenjing Xu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Chaochao Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Fanqi Meng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Department of Spine Surgery, Peking University People's Hospital, Beijing, P.R. China
| | - Yanjun Guan
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Xiuzhi Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Jie Zhao
- Beijing Tsinghua Changgeng Hospital Affiliated to Tsinghua University, Tsinghua University Clinical School, Beijing, P.R. China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China
| |
Collapse
|
35
|
Park Y, Huh KM, Kang SW. Applications of Biomaterials in 3D Cell Culture and Contributions of 3D Cell Culture to Drug Development and Basic Biomedical Research. Int J Mol Sci 2021; 22:2491. [PMID: 33801273 PMCID: PMC7958286 DOI: 10.3390/ijms22052491] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 01/10/2023] Open
Abstract
The process of evaluating the efficacy and toxicity of drugs is important in the production of new drugs to treat diseases. Testing in humans is the most accurate method, but there are technical and ethical limitations. To overcome these limitations, various models have been developed in which responses to various external stimuli can be observed to help guide future trials. In particular, three-dimensional (3D) cell culture has a great advantage in simulating the physical and biological functions of tissues in the human body. This article reviews the biomaterials currently used to improve cellular functions in 3D culture and the contributions of 3D culture to cancer research, stem cell culture and drug and toxicity screening.
Collapse
Affiliation(s)
- Yujin Park
- Department of Polymer Science and Engineering & Chemical Engineering and Applied Chemistry, Chungnam National University, Daejeon 34134, Korea;
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering & Chemical Engineering and Applied Chemistry, Chungnam National University, Daejeon 34134, Korea;
| | - Sun-Woong Kang
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon 34114, Korea
- Human and Environmental Toxicology Program, University of Science and Technology, Daejeon 34114, Korea
| |
Collapse
|
36
|
Wang A, Madden LA, Paunov VN. Advanced biomedical applications based on emerging 3D cell culturing platforms. J Mater Chem B 2020; 8:10487-10501. [PMID: 33136103 DOI: 10.1039/d0tb01658f] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It is of great value to develop reliable in vitro models for cell biology and toxicology. However, ethical issues and the decreasing number of donors restrict the further use of traditional animal models in various fields, including the emerging fields of tissue engineering and regenerative medicine. The huge gap created by the restrictions in animal models has pushed the development of the increasingly recognized three-dimensional (3D) cell culture, which enables cells to closely simulate authentic cellular behaviour such as close cell-to-cell interactions and can achieve higher functionality. Furthermore, 3D cell culturing is superior to the traditional 2D cell culture, which has obvious limitations and cannot closely mimic the structure and architecture of tissues. In this study, we review several methods used to form 3D multicellular spheroids. The extracellular microenvironment of 3D spheroids plays a role in many aspects of biological sciences, including cell signalling, cell growth, cancer cell generation, and anti-cancer drugs. More recently, they have been explored as basic construction units for tissue and organ engineering. We review this field with a focus on the previous research in different areas using spheroid models, emphasizing aqueous two-phase system (ATPS)-based techniques. Multi-cellular spheroids have great potential in the study of biological systems and can closely mimic the in vivo environment. New technologies to form and analyse spheroids such as the aqueous two-phase system and magnetic levitation are rapidly overcoming the technical limitations of spheroids and expanding their applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Anheng Wang
- Department of Chemistry, University of Hull, Hull, HU6 7RX, UK.
| | | | | |
Collapse
|
37
|
Thermoresponsive poly(N-isopropylacrylamide) hydrogel substrates micropatterned with poly(ethylene glycol) hydrogel for adipose mesenchymal stem cell spheroid formation and retrieval. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111128. [DOI: 10.1016/j.msec.2020.111128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/06/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022]
|
38
|
|
39
|
Mizukami Y, Moriya A, Takahashi Y, Shimizu K, Konishi S, Takakura Y, Nishikawa M. Incorporation of Gelatin Microspheres into HepG2 Human Hepatocyte Spheroids for Functional Improvement through Improved Oxygen Supply to Spheroid Core. Biol Pharm Bull 2020; 43:1220-1225. [PMID: 32741942 DOI: 10.1248/bpb.b20-00141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The multicellular spheroid three-dimensional cell culture system can be used as a formulation for cell-based therapy. However, the viability and functions of the cells in the core region of the spheroid tend to decrease because of limited oxygen supply. In this study, we incorporated gelatin microspheres (GMS) into HepG2 human hepatocyte spheroids to allow oxygen to reach the spheroid core. GMS with an approximate diameter of 37 µm were fabricated by water-in-oil emulsification followed by freeze drying. GMS-containing HepG2 spheroids (GMS/HepG2 spheroids) were prepared by incubation of the cells with GMS at various mixing ratios in agarose gel-based microwells. Increasing the GMS ratio increased the diameter of the spheroids, and few spheroids formed with excess GMS. HepG2 cells in the GMS/HepG2 spheroids were more oxygenated than those in the GMS-free spheroids. GMS incorporation increased the viability of HepG2 cells in the spheroids and increased the CYP1A1 activity of the cells to metabolize 7-ethoxyresorufin, although mRNA expression of the CYP1A1 gene was hardly affected by GMS incorporation. These results indicate that incorporating GMS into HepG2 spheroids improves the hypoxic microenvironment in the spheroids and increases cell viability and CYP1A1 metabolic activity.
Collapse
Affiliation(s)
- Yuya Mizukami
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Ai Moriya
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University
| | - Satoshi Konishi
- Department of Mechanical Engineering, Graduate School of Science and Engineering, Ritsumeikan University
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Makiya Nishikawa
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University.,Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| |
Collapse
|
40
|
Doulgkeroglou MN, Di Nubila A, Niessing B, König N, Schmitt RH, Damen J, Szilvassy SJ, Chang W, Csontos L, Louis S, Kugelmeier P, Ronfard V, Bayon Y, Zeugolis DI. Automation, Monitoring, and Standardization of Cell Product Manufacturing. Front Bioeng Biotechnol 2020; 8:811. [PMID: 32766229 PMCID: PMC7381146 DOI: 10.3389/fbioe.2020.00811] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Although regenerative medicine products are at the forefront of scientific research, technological innovation, and clinical translation, their reproducibility and large-scale production are compromised by automation, monitoring, and standardization issues. To overcome these limitations, new technologies at software (e.g., algorithms and artificial intelligence models, combined with imaging software and machine learning techniques) and hardware (e.g., automated liquid handling, automated cell expansion bioreactor systems, automated colony-forming unit counting and characterization units, and scalable cell culture plates) level are under intense investigation. Automation, monitoring and standardization should be considered at the early stages of the developmental cycle of cell products to deliver more robust and effective therapies and treatment plans to the bedside, reducing healthcare expenditure and improving services and patient care.
Collapse
Affiliation(s)
- Meletios-Nikolaos Doulgkeroglou
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Alessia Di Nubila
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | - Niels König
- Fraunhofer Institute for Production Technology, Aachen, Germany
| | - Robert H Schmitt
- Production Engineering Cluster, RWTH Aachen University, Aachen, Germany
| | - Jackie Damen
- STEMCELL Technologies Inc., Vancouver, BC, Canada
| | | | - Wing Chang
- STEMCELL Technologies Ltd., Cambridge, United Kingdom
| | - Lynn Csontos
- STEMCELL Technologies Ltd., Cambridge, United Kingdom
| | - Sharon Louis
- STEMCELL Technologies Inc., Vancouver, BC, Canada
| | | | - Vincent Ronfard
- College System of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States.,Cutiss AG, Zurich, Switzerland.,HairClone, Manchester, United Kingdom
| | - Yves Bayon
- Medtronic - Sofradim Production, Trévoux, France
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
41
|
Becer E, Soykut G, Kabadayi H, Mammadov E, ÇaliŞ İ, Vatansever S. Obtaining Stem Cell Spheroids from Foreskin Tissue and the Effect of Corchorus olitorius L. on Spheroid Proliferation. Turk J Pharm Sci 2020; 17:265-270. [PMID: 32636703 DOI: 10.4274/tjps.galenos.2019.05658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/07/2019] [Indexed: 12/01/2022]
Abstract
Objectives Mesenchymal stem cells are self-renewing stem cells. The human foreskin has potential to be used as a source of stem cells. The aim of the study was to obtain spheroid formation of human foreskin cells (hnFSSCs) isolated from newborn human foreskin tissue. In addition, the apoptotic and proliferative effects of a traditional plant, Corchorus olitorius L. (C. olitorius), on hnFSSC spheroids were investigated. Materials and Methods After a routine circumcision procedure the cells were isolated and cultured in suitable medium. The plant leaves was extracted with ethanol and their composition was analyzed by liquid chromatography coupled with mass spectrometry (LC-MS/MS). The foreskin stem cells were characterized immunocytochemically by CD45, CD34, and CD90 antibodies. hnFSSC spheroids were formed using the hanging drop technique. Immunofluorescence staining was used on the obtained spheroids to determine the distribution of caspase-3 and Ki-67 after being treated with C. olitorius extract for 48 h. Results Immunostaining analysis showed that hnFSSCs were positive for CD45 and CD34 and negative for CD90. According to LC-MS/MS C. olitorius was rich in flavanols and hydrocinnamic acid derivatives. Although the spheroids obtained were loose and floating, the cells interacted with each other. Caspase-3 activity was higher in the control group than in the extract-treated group and Ki-67 was higher in the extract-treated group than in the control group, suggesting that the plant might have the capacity to increase stem cell proliferation due to its rich polyphenolic content. Conclusion The results suggest that hnFSSCs and spheroids might be used in stem cell generation, tissue repair and renewal as human foreskin tissue has potential to be used as a stem cell source. C. olitorius also increased proliferation of hnFSSCs, showing that polyphenols might increase proliferation of stem cells.
Collapse
Affiliation(s)
- Eda Becer
- Near East University Faculty of Pharmacy, Department of Biochemistry, Nicosia, North Cyprus
| | - Günsu Soykut
- Near East University Faculty of Health Sciences, Department of Nutrition and Dietetics, Nicosia, North Cyprus
| | - Hilal Kabadayi
- Manisa Celal Bayar University Faculty of Medicine, Department of Histology and Embryology, Manisa, Turkey
| | - Emil Mammadov
- Near East University, Faculty of Medicine, Department of Pediatric Surgery, Nicosia, North Cyprus
| | - İhsan ÇaliŞ
- Near East University Faculty of Pharmacy, Department of Pharmacognosy, Nicosia, North Cyprus
| | - Seda Vatansever
- Manisa Celal Bayar University Faculty of Medicine, Department of Histology and Embryology, Manisa, Turkey
| |
Collapse
|
42
|
Zhao Z, Vizetto-Duarte C, Moay ZK, Setyawati MI, Rakshit M, Kathawala MH, Ng KW. Composite Hydrogels in Three-Dimensional in vitro Models. Front Bioeng Biotechnol 2020; 8:611. [PMID: 32656197 PMCID: PMC7325910 DOI: 10.3389/fbioe.2020.00611] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
3-dimensional (3D) in vitro models were developed in order to mimic the complexity of real organ/tissue in a dish. They offer new possibilities to model biological processes in more physiologically relevant ways which can be applied to a myriad of applications including drug development, toxicity screening and regenerative medicine. Hydrogels are the most relevant tissue-like matrices to support the development of 3D in vitro models since they are in many ways akin to the native extracellular matrix (ECM). For the purpose of further improving matrix relevance or to impart specific functionalities, composite hydrogels have attracted increasing attention. These could incorporate drugs to control cell fates, additional ECM elements to improve mechanical properties, biomolecules to improve biological activities or any combinations of the above. In this Review, recent developments in using composite hydrogels laden with cells as biomimetic tissue- or organ-like constructs, and as matrices for multi-cell type organoid cultures are highlighted. The latest composite hydrogel systems that contain nanomaterials, biological factors, and combinations of biopolymers (e.g., proteins and polysaccharide), such as Interpenetrating Networks (IPNs) and Soft Network Composites (SNCs) are also presented. While promising, challenges remain. These will be discussed in light of future perspectives toward encompassing diverse composite hydrogel platforms for an improved organ environment in vitro.
Collapse
Affiliation(s)
- Zhitong Zhao
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Catarina Vizetto-Duarte
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Zi Kuang Moay
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | | | - Moumita Rakshit
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | | | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
- Environmental Chemistry & Materials Centre, Nanyang Environment and Water Research Institute (NEWRI), Nanyang Technological University, Singapore, Singapore
- Skin Research Institute of Singapore, Singapore, Singapore
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, United States
| |
Collapse
|
43
|
Zhao N, Coyne J, Abune L, Shi P, Lian XL, Zhang G, Wang Y. Exogenous Signaling Molecules Released from Aptamer-Functionalized Hydrogels Promote the Survival of Mesenchymal Stem Cell Spheroids. ACS APPLIED MATERIALS & INTERFACES 2020; 12:24599-24610. [PMID: 32384232 PMCID: PMC7883300 DOI: 10.1021/acsami.0c05681] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Mesenchymal stem cells (MSCs) have a very low survival rate after in vivo delivery, which limits their great promise for treating human diseases. Various strategies have been studied to overcome this challenge. However, an overlooked but important potential is to apply exogenous signaling molecules as biochemical cues to promote MSC survival, presumably because it is well-known that MSCs themselves can release a variety of potent signaling molecules. Thus, the purpose of this work was to examine and understand whether the release of exogenous signaling molecules from hydrogels can promote the survival of MSC spheroids. Our data show that more vascular endothelial growth factor (VEGF) but not platelet-derived growth factor BB (PDGF-BB) were released from MSC spheroids in comparison with 2D cultured MSCs. Aptamer-functionalized fibrin hydrogel (aFn) could release exogenous VEGF and PDGF-BB in a sustained manner. PDGF-BB-loaded aFn promoted MSC survival by ∼70% more than VEGF-loaded aFn under the hypoxic condition in vitro. Importantly, PDGF-BB-loaded aFn could double the survival rate of MSC spheroids in comparison with VEGF-loaded aFn during the one-week test in vivo. Therefore, this work demonstrated that defined exogenous signaling molecules (e.g., PDGF-BB) can function as biochemical cues for promoting the survival of MSC spheroids in vivo.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - James Coyne
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Lidya Abune
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Peng Shi
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Xiaojun Lance Lian
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
44
|
Murata Y, Jo JI, Yukawa H, Tsumaki N, Baba Y, Tabata Y. Visualization of Human Induced Pluripotent Stem Cells-Derived Three-Dimensional Cartilage Tissue by Gelatin Nanospheres. Tissue Eng Part C Methods 2020; 26:244-252. [PMID: 32143549 DOI: 10.1089/ten.tec.2020.0029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Recently, many studies on the three-dimensional (3D) fabrication of cells have been performed. Under these circumstances, it is indispensable to develop the imaging technologies and methodologies for noninvasive visualization of 3D cells fabricated. The objective of this study is to develop the labeling method of human induced pluripotent stem (iPS) cells-derived 3D cartilage tissue with gelatin nanospheres coincorporating three kinds of quantum dots (QD) and iron oxide nanoparticles (IONP) (GNSQD+IONP). In this study, two labeling methods were performed. One is that a cartilage tissue was labeled directly by incubating with octaarginine (R8)-treated GNSQD+IONP (direct labeling method). The other one is a "dissociation and labeling method." First, the cartilage tissue was dissociated to cells in a single dispersed state. Then, the cells were incubated with R8-GNSQD+IONP in a monolayer culture. Finally, the cells labeled were fabricated to 3D pellets or cell sheets. By the direct labeling method, only cells residing in the surrounding site of cartilage tissue were labeled. On the contrary, the 3D cartilage pellets and the cell sheets were homogenously labeled and maintained fluorescently visualized over 4 weeks. In addition, the cartilage properties were histologically detected even after the process of dissociation and labeling. Homogenous labeling and visualization of human iPS cells-derived 3D cartilage tissue was achieved by the dissociation and labeling method with GNSQD+IONP. Impact statement The homogenous labeling and visualization of human iPS cells-derived three-dimensional (3D) cartilage tissue was achieved over 4 weeks by the dissociation and labeling method with gelatin nanospheres coincorporating quantum dots (QD) and iron oxide nanoparticles (IONP) (GNSQD+IONP). The cartilage properties of cells treated were maintained. It is concluded that the dissociation and labeling method with GNSQD+IONP is a promising to visualize the human iPS cells-derived 3D cartilage tissue.
Collapse
Affiliation(s)
- Yuki Murata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroshi Yukawa
- Institute of Innovation for Future Society and Nagoya University, Nagoya, Japan.,Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Noriyuki Tsumaki
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yoshinobu Baba
- Institute of Innovation for Future Society and Nagoya University, Nagoya, Japan.,Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
45
|
Miao S, Cui H, Esworthy T, Mahadik B, Lee S, Zhou X, Hann SY, Fisher JP, Zhang LG. 4D Self-Morphing Culture Substrate for Modulating Cell Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902403. [PMID: 32195081 PMCID: PMC7080541 DOI: 10.1002/advs.201902403] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/30/2020] [Indexed: 05/08/2023]
Abstract
As the most versatile and promising cell source, stem cells have been studied in regenerative medicine for two decades. Currently available culturing techniques utilize a 2D or 3D microenvironment for supporting the growth and proliferation of stem cells. However, these culture systems fail to fully reflect the supportive biological environment in which stem cells reside in vivo, which contain dynamic biophysical growth cues. Herein, a 4D programmable culture substrate with a self-morphing capability is presented as a means to enhance dynamic cell growth and induce differentiation of stem cells. To function as a model system, a 4D neural culture substrate is fabricated using a combination of printing and imprinting techniques keyed to the different biological features of neural stem cells (NSCs) at different differentiation stages. Results show the 4D culture substrate demonstrates a time-dependent self-morphing process that plays an essential role in regulating NSC behaviors in a spatiotemporal manner and enhances neural differentiation of NSCs along with significant axonal alignment. This study of a customized, dynamic substrate revolutionizes current stem cell therapies, and can further have a far-reaching impact on improving tissue regeneration and mimicking specific disease progression, as well as other impacts on materials and life science research.
Collapse
Affiliation(s)
- Shida Miao
- Department of Mechanical and Aerospace EngineeringThe George Washington University3590 Science and Engineering Hall, 800 22nd Street NWWashingtonDC20052USA
| | - Haitao Cui
- Department of Mechanical and Aerospace EngineeringThe George Washington University3590 Science and Engineering Hall, 800 22nd Street NWWashingtonDC20052USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace EngineeringThe George Washington University3590 Science and Engineering Hall, 800 22nd Street NWWashingtonDC20052USA
| | - Bhushan Mahadik
- Fischell Department of BioengineeringUniversity of Maryland3238 Jeong H. Kim Engineering BuildingCollege ParkMD20742USA
| | - Se‐jun Lee
- Department of Mechanical and Aerospace EngineeringThe George Washington University3590 Science and Engineering Hall, 800 22nd Street NWWashingtonDC20052USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace EngineeringThe George Washington University3590 Science and Engineering Hall, 800 22nd Street NWWashingtonDC20052USA
| | - Sung Yun Hann
- Department of Mechanical and Aerospace EngineeringThe George Washington University3590 Science and Engineering Hall, 800 22nd Street NWWashingtonDC20052USA
| | - John P. Fisher
- Fischell Department of BioengineeringUniversity of Maryland3238 Jeong H. Kim Engineering BuildingCollege ParkMD20742USA
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace EngineeringThe George Washington University3590 Science and Engineering Hall, 800 22nd Street NWWashingtonDC20052USA
- Department of Electrical and Computer EngineeringDepartment of MedicineDepartment of Biomedical EngineeringThe George Washington UniversityWashingtonDC20052USA
| |
Collapse
|
46
|
Ong CS, Pitaktong I, Hibino N. Principles of Spheroid Preparation for Creation of 3D Cardiac Tissue Using Biomaterial-Free Bioprinting. Methods Mol Biol 2020; 2140:183-197. [PMID: 32207113 DOI: 10.1007/978-1-0716-0520-2_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biomaterial-free three-dimensional (3D) bioprinting is a relatively new field within 3D bioprinting, where 3D tissues are created from the fusion of 3D multicellular spheroids, without requiring biomaterial. This is in contrast to traditional 3D bioprinting, which requires biomaterials to carry the cells to be bioprinted, such as a hydrogel or decellularized extracellular matrix. Here, we discuss principles of spheroid preparation for biomaterial-free 3D bioprinting of cardiac tissue. In addition, we discuss principles of using spheroids as building blocks in biomaterial-free 3D bioprinting, including spheroid dislodgement, spheroid transfer, and spheroid fusion. These principles are important considerations, to create the next generation of biomaterial-free spheroid-based 3D bioprinters.
Collapse
Affiliation(s)
- Chin Siang Ong
- Division of Cardiac Surgery, The Johns Hopkins Hospital, Baltimore, MD, USA.,Division of Cardiology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Isaree Pitaktong
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Narutoshi Hibino
- Division of Cardiac Surgery, The Johns Hopkins Hospital, Baltimore, MD, USA. .,Section of Cardiac Surgery, Department of Surgery, The University of Chicago, Advocate Children's Hospital, Chicago, IL, USA.
| |
Collapse
|
47
|
Takeuchi H, Ikeguchi R, Aoyama T, Oda H, Yurie H, Mitsuzawa S, Tanaka M, Ohta S, Akieda S, Miyazaki Y, Nakayama K, Matsuda S. A scaffold-free Bio 3D nerve conduit for repair of a 10-mm peripheral nerve defect in the rats. Microsurgery 2019; 40:207-216. [PMID: 31724780 DOI: 10.1002/micr.30533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 08/13/2019] [Accepted: 10/25/2019] [Indexed: 12/28/2022]
Abstract
INTRODUCTION A Bio 3D printed nerve conduit was reported to promote nerve regeneration in a 5 mm nerve gap model. The purpose of this study was to fabricate Bio 3D nerve conduits suitable for a 10 mm nerve gap and to evaluate their capacity for nerve regeneration in a rat sciatic nerve defect model. MATERIALS AND METHODS Eighteen F344 rats with immune deficiency (9-10 weeks old; weight, 200-250 g) were divided into three groups: a Bio 3D nerve conduit group (Bio 3D, n = 6), a nerve graft group (NG, n = 6), and a silicon tube group (ST, n = 6). A 12-mm Bio 3D nerve conduit or silicon tube was transplanted into the 10-mm defect of the right sciatic nerve. In the nerve graft group, reverse autografting was performed with an excised 10-mm nerve segment. Assessments were performed at 8 weeks after the surgery. RESULTS In the region distal to the suture site, the number of myelinated axons in the Bio 3D group were significantly larger compared with the silicon group (2,548 vs. 950, p < .05). The myelinated axon diameter (MAD) and the myelin thickness (MT) of the regenerated axons in the Bio 3D group were significantly larger compared with those of the ST group (MAD: 3.09 vs. 2.36 μm; p < .01; MT: 0.59 vs. 0.40 μm, p < .01). CONCLUSIONS This study indicates that a Bio 3D nerve conduit can enhance peripheral nerve regeneration even in a 10 mm nerve defect model.
Collapse
Affiliation(s)
- Hisataka Takeuchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Ikeguchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Physical Therapy, Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroki Oda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirofumi Yurie
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sadaki Mitsuzawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mai Tanaka
- Department of Physical Therapy, Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Souichi Ohta
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering Faculty of Medicine, Saga University, Saga, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
48
|
Qiao Y, Xu Z, Yu Y, Hou S, Geng J, Xiao T, Liang Y, Dong Q, Mei Y, Wang B, Qiao H, Dai J, Suo G. Single cell derived spheres of umbilical cord mesenchymal stem cells enhance cell stemness properties, survival ability and therapeutic potential on liver failure. Biomaterials 2019; 227:119573. [PMID: 31670080 DOI: 10.1016/j.biomaterials.2019.119573] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/05/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023]
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) have shown great potentials in regenerative medicine for their extensive sources, multilineage differentiation potential, low immunogenicity and self-renewal ability. However, the clinical application of UCMSCs still confronts many challenges including the requirement of large quantity of cells, low survival ability in vivo and the loss of main original characteristics due to two-dimensional (2D) culture. The traditional three-dimensional (3D)-spheroid culture can mimic in vivo conditions, but still has limitations in clinical application due to large size of spheroid against direct injection and inner cell death. Based on self-renewal tenet, we produced single cell derived sphere (SCDS) of UCMSCs through combining single cell pattern on chip with 3D culture. Compared with the 2D and traditional 3D culture, SCDS culture has many advantages to meet clinical requirements, including small size, higher abilities of survival and migration, and stronger hypoxia resistance and stemness maintenance. Furthermore, SCDS culture promotes angiogenesis in UCMSCs-xenografts and displays greater therapeutic potential on acute liver failure (ALF) in vivo. Our results suggest that SCDS culture may serve as a simple and effective strategy for UCMSCs optimization to meet clinical demand.
Collapse
Affiliation(s)
- Yong Qiao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China
| | - Zhongjuan Xu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yanzhen Yu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shulan Hou
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; School of Pharmacy, Xi'an Jiaotong University, Shaanxi, 710061, China
| | - Junsa Geng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Tongqian Xiao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu Liang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Qun Dong
- Department of Pathology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu, 210046, China
| | - Yan Mei
- Greepharma Inc., 211100, Nanjing, Jiangsu, China
| | - Bin Wang
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Hong Qiao
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jianwu Dai
- State Key Laboratory of Molecular, Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Guangli Suo
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China.
| |
Collapse
|
49
|
Cutrona MB, Simpson JC. A High-Throughput Automated Confocal Microscopy Platform for Quantitative Phenotyping of Nanoparticle Uptake and Transport in Spheroids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902033. [PMID: 31334922 DOI: 10.1002/smll.201902033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/24/2019] [Indexed: 05/23/2023]
Abstract
There is a high demand for advanced, image-based, automated high-content screening (HCS) approaches to facilitate phenotypic screening in 3D cell culture models. A major challenge lies in retaining the resolution of fine cellular detail but at the same time imaging multicellular structures at a large scale. In this study, a confocal microscopy-based HCS platform in optical multiwell plates that enables the quantitative morphological profiling of populations of nonuniform spheroids obtained from HT-29 human colorectal cancer cells is described. This platform is then utilized to demonstrate a quantitative dissection of the penetration of synthetic nanoparticles (NP) in multicellular 3D spheroids at multiple levels of scale. A pilot RNA interference-based screening validates this methodology and identifies a subset of RAB GTPases that regulate NP trafficking in these spheroids. This technology is suitable for high-content phenotyping in 3D cell-based screening, providing a framework for nanomedicine drug development as applied to translational oncology.
Collapse
Affiliation(s)
- Meritxell B Cutrona
- School of Biology and Environmental Science & Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), D04 N2E5, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), Galway, H91 W2TY, Ireland
| | - Jeremy C Simpson
- School of Biology and Environmental Science & Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), D04 N2E5, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), Galway, H91 W2TY, Ireland
| |
Collapse
|
50
|
Jafari J, Han XL, Palmer J, Tran PA, O'Connor AJ. Remote Control in Formation of 3D Multicellular Assemblies Using Magnetic Forces. ACS Biomater Sci Eng 2019; 5:2532-2542. [PMID: 33405759 DOI: 10.1021/acsbiomaterials.9b00297] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell constructs have been utilized as building blocks in tissue engineering to closely mimic the natural tissue and also overcome some of the limitations caused by two-dimensional cultures or using scaffolds. External forces can be used to enhance the cells' adhesion and interaction and thus provide better control over production of these structures compared to methods like cell seeding and migration. In this paper, we demonstrate an efficient method to generate uniform, three-dimensional cell constructs using magnetic forces. This method produced spheroids with higher densities and more symmetrical structures than the commonly used centrifugation method for production of cell spheroids. It was also shown that shape of the cell constructs could be changed readily by using different patterns of magnetic field. The application of magnetic fields to impart forces on the cells enhanced the fusion of these spheroids, which could be used to produce larger and more complicated structures for future tissue engineering applications.
Collapse
Affiliation(s)
- Javad Jafari
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Grattan St., Parkville, Victoria 3010, Australia
| | - Xiao-Lian Han
- O'Brien Institute Department, St. Vincent's Institute, 42 Fitzroy Street, Fitzroy, Victoria 3065, Australia
| | - Jason Palmer
- O'Brien Institute Department, St. Vincent's Institute, 42 Fitzroy Street, Fitzroy, Victoria 3065, Australia
| | - Phong A Tran
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Grattan St., Parkville, Victoria 3010, Australia.,Interface Science and Materials Engineering Group, School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology (QUT), 2 George St., Brisbane, Queensland 4000, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Grattan St., Parkville, Victoria 3010, Australia
| |
Collapse
|