1
|
Wheeler EE, Leach JK. Tissue-Engineered Three-Dimensional Platforms for Disease Modeling and Therapeutic Development. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39345164 DOI: 10.1089/ten.teb.2024.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Three-dimensional (3D) tissue-engineered models are under investigation to recapitulate tissue architecture and functionality, thereby overcoming limitations of traditional two-dimensional cultures and preclinical animal models. This review highlights recent developments in 3D platforms designed to model diseases in vitro that affect numerous tissues and organs, including cardiovascular, gastrointestinal, bone marrow, neural, reproductive, and pulmonary systems. We discuss current technologies for engineered tissue models, highlighting the advantages, limitations, and important considerations for modeling tissues and diseases. Lastly, we discuss future advancements necessary to enhance the reliability of 3D models of tissue development and disease.
Collapse
Affiliation(s)
- Erika E Wheeler
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, California, USA
| |
Collapse
|
2
|
Farhang Doost N, Srivastava SK. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. BIOSENSORS 2024; 14:225. [PMID: 38785699 PMCID: PMC11118005 DOI: 10.3390/bios14050225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that simulates an artificial organ within a microfluidic cell culture chip. Current cell biology research focuses on in vitro cell cultures due to various limitations of in vivo testing. Unfortunately, in-vitro cell culturing fails to provide an accurate microenvironment, and in vivo cell culturing is expensive and has historically been a source of ethical controversy. OOC aims to overcome these shortcomings and provide the best of both in vivo and in vitro cell culture research. The critical component of the OOC design is utilizing microfluidics to ensure a stable concentration gradient, dynamic mechanical stress modeling, and accurate reconstruction of a cellular microenvironment. OOC also has the advantage of complete observation and control of the system, which is impossible to recreate in in-vivo research. Multiple throughputs, channels, membranes, and chambers are constructed in a polydimethylsiloxane (PDMS) array to simulate various organs on a chip. Various experiments can be performed utilizing OOC technology, including drug delivery research and toxicology. Current technological expansions involve multiple organ microenvironments on a single chip, allowing for studying inter-tissue interactions. Other developments in the OOC technology include finding a more suitable material as a replacement for PDMS and minimizing artefactual error and non-translatable differences.
Collapse
Affiliation(s)
| | - Soumya K. Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
3
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [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: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
4
|
Barozzi D, Scielzo C. Emerging Strategies in 3D Culture Models for Hematological Cancers. Hemasphere 2023; 7:e932. [PMID: 37520775 PMCID: PMC10378728 DOI: 10.1097/hs9.0000000000000932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/16/2023] [Indexed: 08/01/2023] Open
Abstract
In vitro cell cultures are fundamental and necessary tools in cancer research and personalized drug discovery. Currently, most cells are cultured using two-dimensional (2D) methods, and drug testing is mainly performed in animal models. However, new and improved methods that implement three-dimensional (3D) cell-culturing techniques provide compelling evidence that more advanced experiments can be performed, yielding valuable new insights. In 3D cell-culture experiments, the cell environment can be manipulated to mimic the complexity and dynamicity of the human tissue microenvironment, possibly leading to more accurate representations of cell-to-cell interactions, tumor biology, and predictions of drug response. The 3D cell cultures can also potentially provide alternative ways to study hematological cancers and are expected to eventually bridge the gap between 2D cell culture and animal models. The present review provides an overview of the complexity of the lymphoid microenvironment and a summary of the currently used 3D models that aim at recreating it for hematological cancer research. We here dissect the differences and challenges between, and potential advantages of, different culture methods and present our vision of the most promising future strategies in the hematological field.
Collapse
Affiliation(s)
- Dafne Barozzi
- Università degli Studi di Milano-Bicocca, School of Medicine and Surgery, PhD program in Molecular and Translational Medicine (DIMET), Milano, Italy
- Unit of Malignant B cells biology and 3D modelling, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Cristina Scielzo
- Unit of Malignant B cells biology and 3D modelling, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
5
|
The differentiation of human induced pluripotent stem cells into hematopoietic stem cells on 3D bone scaffold in a dynamic culture system. Tissue Cell 2023; 82:102044. [PMID: 36905860 DOI: 10.1016/j.tice.2023.102044] [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: 04/28/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Hematopoietic stem cell transplantation is used for cell-based therapy for many hematological disorders. However, difficulty in finding proper donors has limited this source of stem cells. For clinical application, the generation of these cells from induced pluripotent stem cells (iPSs) is a fascinating and endless source. One of the experimental methods to generate HSCs from iPSs is the mimicking of the hematopoietic niche. In the current study, as the first phase of differentiation, embryoid bodies were formed from iPSs. They were then cultured in different dynamic conditions in order to determine the appropriate settings for their differentiation into HSCs. The dynamic culture was composed of DBM Scaffold with or without growth factor. After ten days, the specific HSC markers (CD34, CD133, CD31 and CD45) were assessed using flow-cytometry. Our findings demonstrated that the dynamic conditions were significantly suitable than static ones. In addition, in 3D scaffold and dynamic system the expression of CXCR4, as a homing marker, was increased. These results suggest that the 3D culture bioreactor with DBM scaffold could provide a new approach for differentiation of iPSs into HSCs. Moreover, this system could provide maximum mimicry of bone marrow niche.
Collapse
|
6
|
Liu B, Jin M, Wang DA. In vitro expansion of hematopoietic stem cells in a porous hydrogel-based 3D culture system. Acta Biomater 2023; 161:67-79. [PMID: 36754271 DOI: 10.1016/j.actbio.2023.01.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 02/10/2023]
Abstract
Hematopoietic stem cell (HSC) transplantation remains the most effective therapy for hematologic and lymphoid disorders. However, as the primary therapeutic cells, the source of HSCs has been limited due to the scarcity of matched donors and difficulties in ex vivo expansion. Here, we described a facile method to attempt the expansion of HSCs in vitro through a porous alginate hydrogel-based 3D culture system. We used gelatin powders as the porogen to create submillimeter-scaled pores in alginate gel bulk while pre-embedding naïve HSCs in the gel phase. The results indicated that this porous hydrogel system performed significantly better than those cultured via conventional suspension or encapsulation in non-porous alginate hydrogels in maintaining the phenotype and renewability of HSCs. Only the porous hydrogel system achieved a two-fold growth of CD34+ cells within seven days of culture, while the number of CD34+ cells in the suspension system and nonporous hydrogel showed different degrees of attenuation. The expansion efficiency of the porous hydrogel for CD34+CD38- cells was more than 2.2 times that of the other two systems. Mechanistic study via biophysical analysis revealed that the porous alginate system was competent to reduce the electron capture caused by biomaterials, decrease cellular oxygen stress, avoid oxidative protection, thus maintaining the cellular phenotype of the CD34+ cells. The transcriptomic analysis further suggested that the porous alginate system also upregulated the TNF signaling pathway and activated the NF-κB signaling pathway to promote the CD34+ cells' survival and maintain cellular homeostasis so that renewability was substantially favoured. STATEMENT OF SIGNIFICANCE: • The reported porous hydrogel system performs significantly better in terms of maintaining the phenotype and renewability of HSCs than those cultured via conventional suspension or encapsulation in non-porous alginate hydrogel. • The reported porous alginate system is competent to reduce the electron capture caused by biomaterials, decrease cellular oxygen stress, avoid oxidative protection, and therefore maintain the cellular phenotype of the CD34+ cells. • The reported porous alginate system can also upregulate the TNF signaling pathway and activate the NF-κB signaling pathway to promote the CD34+ cells' survival and maintain cellular homeostasis so that the renewability is substantially favored..
Collapse
Affiliation(s)
- Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China; Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Min Jin
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China; Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China; Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
7
|
Krysko DV, Demuynck R, Efimova I, Naessens F, Krysko O, Catanzaro E. In Vitro Veritas: From 2D Cultures to Organ-on-a-Chip Models to Study Immunogenic Cell Death in the Tumor Microenvironment. Cells 2022; 11:3705. [PMID: 36429133 PMCID: PMC9688238 DOI: 10.3390/cells11223705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Immunogenic cell death (ICD) is a functionally unique form of cell death that promotes a T-cell-dependent anti-tumor immune response specific to antigens originating from dying cancer cells. Many anticancer agents and strategies induce ICD, but despite their robust effects in vitro and in vivo on mice, translation into the clinic remains challenging. A major hindrance in antitumor research is the poor predictive ability of classic 2D in vitro models, which do not consider tumor biological complexity, such as the contribution of the tumor microenvironment (TME), which plays a crucial role in immunosuppression and cancer evasion. In this review, we describe different tumor models, from 2D cultures to organ-on-a-chip technology, as well as spheroids and perfusion bioreactors, all of which mimic the different degrees of the TME complexity. Next, we discuss how 3D cell cultures can be applied to study ICD and how to increase the translational potential of the ICD inducers. Finally, novel research directions are provided regarding ICD in the 3D cellular context which may lead to novel immunotherapies for cancer.
Collapse
Affiliation(s)
- Dmitri V. Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Robin Demuynck
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Iuliia Efimova
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Faye Naessens
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Catanzaro
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
8
|
Liu X, Jiang Z, Xing D, Yang Y, Li Z, Sun Z. Recent progress in nanocomposites of carbon dioxide fixation derived reproducible biomedical polymers. Front Chem 2022; 10:1035825. [PMID: 36277338 PMCID: PMC9585172 DOI: 10.3389/fchem.2022.1035825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
In recent years, the environmental problems accompanying the extensive application of biomedical polymer materials produced from fossil fuels have attracted more and more attentions. As many biomedical polymer products are disposable, their life cycle is relatively short. Most of the used or overdue biomedical polymer products need to be burned after destruction, which increases the emission of carbon dioxide (CO2). Developing biomedical products based on CO2 fixation derived polymers with reproducible sources, and gradually replacing their unsustainable fossil-based counterparts, will promote the recycling of CO2 in this field and do good to control the greenhouse effect. Unfortunately, most of the existing polymer materials from renewable raw materials have some property shortages, which make them unable to meet the gradually improved quality and property requirements of biomedical products. In order to overcome these shortages, much time and effort has been dedicated to applying nanotechnology in this field. The present paper reviews recent advances in nanocomposites of CO2 fixation derived reproducible polymers for biomedical applications, and several promising strategies for further research directions in this field are highlighted.
Collapse
Affiliation(s)
- Xin Liu
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhiwen Jiang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Zhiwen Jiang, ; Zhiying Li,
| | - Dejun Xing
- Tumor Hospital of Jilin Province, Changchun, China
| | - Yan Yang
- Tumor Hospital of Jilin Province, Changchun, China
| | - Zhiying Li
- Tumor Hospital of Jilin Province, Changchun, China
- *Correspondence: Zhiwen Jiang, ; Zhiying Li,
| | - Zhiqiang Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
9
|
Current insights into the bone marrow niche: From biology in vivo to bioengineering ex vivo. Biomaterials 2022; 286:121568. [DOI: 10.1016/j.biomaterials.2022.121568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 11/21/2022]
|
10
|
Lee JW, Kim HS, Yon SJ, Matsumoto T, Lee SK, Lee KY. In vitro culture of hematopoietic stem cell niche using angiopoietin-1-coupled alginate hydrogel. Int J Biol Macromol 2022; 209:1893-1899. [PMID: 35489624 DOI: 10.1016/j.ijbiomac.2022.04.163] [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: 08/09/2021] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 11/05/2022]
Abstract
Stem cells exist and maintain their quiescence and pluripotency in stem cell niche. Here, we hypothesized that regulation of cell-cell interactions using a polymeric scaffold as synthetic extracellular matrix (ECM) could be critical in creating a hematopoietic stem cell (HSC) niche in vitro. Angiopoietin-1 (Ang1) binds to the tyrosine kinase receptor (Tie2), and regulation of the Tie2/Ang1 interaction is important in maintaining the quiescence of HSCs in vivo. Alginate hydrogel was thus modified with Ang1 as a synthetic ECM to mimic the HSC niche. Long-term HSCs (CD34-, CD135-, and CD150+) were isolated from mouse femurs and cultured on Ang1-modified alginate hydrogel. The percentage of LT-HSCs in G0 phase was 46.8 ± 1.8%, which was comparable to that of LT-HSCs co-cultured with osteoblasts (46.8 ± 2.1%). Ang1-coupled alginate gels were useful to provide a niche for HSC quiescence without a co-culture system. Polymeric scaffolds containing biomimetic and cell-instructive characteristics for stem cell phenotype regulation might help create HSC niches in vitro and be useful to engineer tissues and transplant stem cells.
Collapse
Affiliation(s)
- Jae Won Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyun Seung Kim
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Soo-Jeong Yon
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Sang-Kyung Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea
| | - Kuen Yong Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
11
|
In Vitro Models of Bone Marrow Remodelling and Immune Dysfunction in Space: Present State and Future Directions. Biomedicines 2022; 10:biomedicines10040766. [PMID: 35453515 PMCID: PMC9031916 DOI: 10.3390/biomedicines10040766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Spaceflight affects the body on every level. Reports on astronaut health identify bone marrow remodelling and dysfunction of the innate immune system as significant health risks of long-term habitation in space. Microgravity-induced alterations of the bone marrow induce physical changes to the bone marrow stem cell niche. Downstream effects on innate immunity are expected due to impaired hematopoiesis and myelopoiesis. To date, few studies have investigated these effects in real microgravity and the sparsely available literature often reports contrasting results. This emphasizes a need for the development of physiologically relevant in vitro models of the bone marrow stem cell niche, capable of delivering appropriate sample sizes for robust statistics. Here, we review recent findings on the impact of spaceflight conditions on innate immunity in in vitro and animal models and discusses the latest in vitro models of the bone marrow stem cell niche and their potential translatability to gravitational biology research.
Collapse
|
12
|
Sui C, Zilberberg J, Lee W. Microfluidic device engineered to study the trafficking of multiple myeloma cancer cells through the sinusoidal niche of bone marrow. Sci Rep 2022; 12:1439. [PMID: 35087109 PMCID: PMC8795452 DOI: 10.1038/s41598-022-05520-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 12/13/2021] [Indexed: 02/04/2023] Open
Abstract
Multiple myeloma (MM) is an incurable B cell malignancy characterized by the accumulation of monoclonal abnormal plasma cells in the bone marrow (BM). It has been a significant challenge to study the spatiotemporal interactions of MM cancer cells with the embedded microenvironments of BM. Here we report a microfluidic device which was designed to mimic several physiological features of the BM niche: (1) sinusoidal circulation, (2) sinusoidal endothelium, and (3) stroma. The endothelial and stromal compartments were constructed and used to demonstrate the device's utility by spatiotemporally characterizing the CXCL12-mediated egression of MM cells from the BM stroma and its effects on the barrier function of endothelial cells (ECs). We found that the egression of MM cells resulted in less organized and loosely connected ECs, the widening of EC junction pores, and increased permeability through ECs, but without significantly affecting the number density of viable ECs. The results suggest that the device can be used to study the physical and secreted factors determining the trafficking of cancer cells through BM. The sinusoidal flow feature of the device provides an integral element for further creating systemic models of cancers that reside or metastasize to the BM niche.
Collapse
Affiliation(s)
- Chao Sui
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point on Hudson, Hoboken, NJ, 07030, USA
| | - Jenny Zilberberg
- Hackensack Meridian Health, Center for Discovery and Innovation, Nutley, NJ, 07110, USA
| | - Woo Lee
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point on Hudson, Hoboken, NJ, 07030, USA. .,Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point On Hudson, Hoboken, NJ, 07030, USA.
| |
Collapse
|
13
|
Zmrhal V, Svoradova A, Batik A, Slama P. Three-Dimensional Avian Hematopoietic Stem Cell Cultures as a Model for Studying Disease Pathogenesis. Front Cell Dev Biol 2022; 9:730804. [PMID: 35127695 PMCID: PMC8811169 DOI: 10.3389/fcell.2021.730804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
Three-dimensional (3D) cell culture is attracting increasing attention today because it can mimic tissue environments and provide more realistic results than do conventional cell cultures. On the other hand, very little attention has been given to using 3D cell cultures in the field of avian cell biology. Although mimicking the bone marrow niche is a classic challenge of mammalian stem cell research, experiments have never been conducted in poultry on preparing in vitro the bone marrow niche. It is well known, however, that all diseases cause immunosuppression and target immune cells and their development. Hematopoietic stem cells (HSC) reside in the bone marrow and constitute a source for immune cells of lymphoid and myeloid origins. Disease prevention and control in poultry are facing new challenges, such as greater use of alternative breeding systems and expanding production of eggs and chicken meat in developing countries. Moreover, the COVID-19 pandemic will draw greater attention to the importance of disease management in poultry because poultry constitutes a rich source of zoonotic diseases. For these reasons, and because they will lead to a better understanding of disease pathogenesis, in vivo HSC niches for studying disease pathogenesis can be valuable tools for developing more effective disease prevention, diagnosis, and control. The main goal of this review is to summarize knowledge about avian hematopoietic cells, HSC niches, avian immunosuppressive diseases, and isolation of HSC, and the main part of the review is dedicated to using 3D cell cultures and their possible use for studying disease pathogenesis with practical examples. Therefore, this review can serve as a practical guide to support further preparation of 3D avian HSC niches to study the pathogenesis of avian diseases.
Collapse
Affiliation(s)
- Vladimir Zmrhal
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Andrea Svoradova
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
- NPPC, Research Institute for Animal Production in Nitra, Luzianky, Slovak Republic
| | - Andrej Batik
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| |
Collapse
|
14
|
Culturing patient-derived malignant hematopoietic stem cells in engineered and fully humanized 3D niches. Proc Natl Acad Sci U S A 2021; 118:2114227118. [PMID: 34580200 DOI: 10.1073/pnas.2114227118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 01/13/2023] Open
Abstract
Human malignant hematopoietic stem and progenitor cells (HSPCs) reside in bone marrow (BM) niches, which remain challenging to explore due to limited in vivo accessibility and constraints with humanized animal models. Several in vitro systems have been established to culture patient-derived HSPCs in specific microenvironments, but they do not fully recapitulate the complex features of native bone marrow. Our group previously reported that human osteoblastic BM niches (O-N), engineered by culturing mesenchymal stromal cells within three-dimensional (3D) porous scaffolds under perfusion flow in a bioreactor system, are capable of maintaining, expanding, and functionally regulating healthy human cord blood-derived HSPCs. Here, we first demonstrate that this 3D O-N can sustain malignant CD34+ cells from acute myeloid leukemia (AML) and myeloproliferative neoplasm patients for up to 3 wk. Human malignant cells distributed in the bioreactor system mimicking the spatial distribution found in native BM tissue, where most HSPCs remain linked to the niches and mature cells are released to the circulation. Using human adipose tissue-derived stromal vascular fraction cells, we then generated a stromal-vascular niche and demonstrated that O-N and stromal-vascular niche differentially regulate leukemic UCSD-AML1 cell expansion, immunophenotype, and response to chemotherapy. The developed system offers a unique platform to investigate human leukemogenesis and response to drugs in customized environments, mimicking defined features of native hematopoietic niches and compatible with the establishment of personalized settings.
Collapse
|
15
|
Ravichandran A, Meinert C, Bas O, Hutmacher DW, Bock N. Engineering a 3D bone marrow adipose composite tissue loading model suitable for studying mechanobiological questions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112313. [PMID: 34474864 DOI: 10.1016/j.msec.2021.112313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Tissue engineering strategies are widely used to model and study the bone marrow microenvironment in healthy and pathological conditions. Yet, while bone function highly depends on mechanical stimulation, the effects of biomechanical stimuli on the bone marrow niche, specifically on bone marrow adipose tissue (BMAT) is poorly understood due to a lack of representative in vitro loading models. Here, we engineered a BMAT analog made of a GelMA (gelatin methacryloyl) hydrogel/medical-grade polycaprolactone (mPCL) scaffold composite to structurally and biologically mimic key aspects of the bone marrow microenvironment, and exploited an innovative bioreactor to study the effects of mechanical loading. Highly reproducible BMAT analogs facilitated the successful adipogenesis of human mesenchymal bone marrow stem cells. Upon long-term intermittent stimulation (1 Hz, 2 h/day, 3 days/week, 3 weeks) in the novel bioreactor, cellular proliferation and lipid accumulation were similar to unloaded controls, yet there was a significant reduction in the secretion of adipokines including leptin and adiponectin, in line with clinical evidence of reduced adipokine expression following exercise/activity. Ultimately, this innovative loading platform combined with reproducibly engineered BMAT analogs provide opportunities to study marrow physiology in greater complexity as it accounts for the dynamic mechanical microenvironment context.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia
| | - Christoph Meinert
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Metro North Hospital and Health Service, Herston 4029, QLD, Australia
| | - Onur Bas
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove 4059, QLD, Australia
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; Bone and Joint Disorders Program, School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane 4000, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia; ARC Industrial Transformation Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing, QUT, Kelvin Grove 4059, QLD, Australia.
| |
Collapse
|
16
|
Chen L, Zhou D, Li X, Yang B, Xu T. Bioprinting of Human Cord Blood-Derived CD34+ Cells and Exploration of the Multilineage Differentiation Ability in Vitro. ACS Biomater Sci Eng 2021; 7:2592-2604. [PMID: 33939424 DOI: 10.1021/acsbiomaterials.0c01297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The three-dimensional (3D) marrow microenvironment plays an essential role in regulating human cord blood-derived CD34+ cells (hCB-CD34+) migration, proliferation, and differentiation. Extensive in vitro and in vivo studies have aimed to recapitulate the main components of the bone marrow (BM) niche. Nonetheless, the models are limited by a lack of heterogeneity and compound structure. Here, we fabricated coaxial extruded core-shell tubular scaffolds and extrusion-based bioprinted cell-laden mesh scaffolds to mimic the functional niche in vitro. A multicellular mesh scaffold and two different core-shell tubular scaffolds were developed with human bone marrow-derived mesenchymal stromal cells (BMSCs) in comparison with a conventional 2D coculture system. A clear cell-cell connection was established in all three bioprinted constructs. Cell distribution and morphology were observed in different systems with scanning electron microscopy (SEM). Collected hCB-CD34+ cells were characterized by various stem cell-specific and lineage-specific phenotypic parameters. The results showed that compared with hCB-CD34+ cells cocultured with BMSCs in Petri dishes, the self-renewal potential of hCB-CD34+ cells was stronger in the tubular scaffolds after 14 days. Besides, cells in these core-shell constructs tended to obtain stronger differentiation potential of lymphoid and megakaryocytes, while cells encapsulated in mesh scaffolds obtained stronger differentiation tendency into erythroid cells. Consequently, 3D bioprinting technology could partially simulate the niche of human hematopoietic stem cells. The three models have their potential in stemness maintenance and multilineage differentiation. This study can provide initial effective guidance in the directed differentiation research and related screening of drug models for hematological diseases.
Collapse
Affiliation(s)
- Lidan Chen
- Centre of Maxillofacial Surgery and Digital Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Xinda Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Bin Yang
- Centre of Maxillofacial Surgery and Digital Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, People's Republic of China
| | - Tao Xu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Department of Precision Medicine and Healthcare, Tsinghua Berkeley Shenzhen Institute, Shenzhen 518055, People's Republic of China
| |
Collapse
|
17
|
Carreras P, González I, Gallardo M, Ortiz-Ruiz A, Morales ML, Encinas J, Martínez-López J. Long-Term Human Hematopoietic Stem Cell Culture in Microdroplets. MICROMACHINES 2021; 12:90. [PMID: 33467039 PMCID: PMC7830102 DOI: 10.3390/mi12010090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/16/2020] [Accepted: 01/12/2021] [Indexed: 12/28/2022]
Abstract
We previously reported a new approach for micromanipulation and encapsulation of human stem cells using a droplet-based microfluidic device. This approach demonstrated the possibility of encapsulating and culturing difficult-to-preserve primary human hematopoietic stem cells using an engineered double-layered bead composed by an inner layer of alginate and an outer layer of Puramatrix. We also demonstrated the maintenance and expansion of Multiple Myeloma cells in this construction. Here, the presented microfluidic technique is applied to construct a 3D biomimetic model to recapitulate the human hematopoietic stem cell niche using double-layered hydrogel beads cultured in 10% FBS culture medium. In this model, the long-term maintenance of the number of cells and expansion of hHSCS encapsulated in the proposed structures was observed. Additionally, a phenotypic characterization of the human hematopoietic stem cells generated in the presented biomimetic model was performed in order to assess their long-term stemness maintenance. Results indicate that the ex vivo cultured human CD34+ cells from bone marrow were viable, maintained, and expanded over a time span of eight weeks. This novel long-term stem cell culture methodology could represent a novel breakthrough to improve Hematopoietic Progenitor cell Transplant (HPT) as well as a novel tool for further study of the biochemical and biophysical factors influencing stem cell behavior. This technology opens a myriad of new applications as a universal stem cell niche model potentially able to expand other types of cells.
Collapse
Affiliation(s)
- Pilar Carreras
- CSIC, Spanish National Research Council, 28006 Madrid, Spain;
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
| | - Itziar González
- CSIC, Spanish National Research Council, 28006 Madrid, Spain;
| | - Miguel Gallardo
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Alejandra Ortiz-Ruiz
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Maria Luz Morales
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Jessica Encinas
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Joaquín Martínez-López
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
- UCM, Medical Faculty, Complutense University Madrid, 28040 Madrid, Spain
| |
Collapse
|
18
|
Bioactive and Topographically-Modified Electrospun Membranes for the Creation of New Bone Regeneration Models. Processes (Basel) 2020. [DOI: 10.3390/pr8111341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Bone injuries that arise from trauma, cancer treatment, or infection are a major and growing global challenge. An increasingly ageing population plays a key role in this, since a growing number of fractures are due to diseases such as osteoporosis, which place a burden on healthcare systems. Current reparative strategies do not sufficiently consider cell-substrate interactions that are found in healthy tissues; therefore, the need for more complex models is clear. The creation of in vitro defined 3D microenvironments is an emerging topographically-orientated approach that provides opportunities to apply knowledge of cell migration and differentiation mechanisms to the creation of new cell substrates. Moreover, introducing biofunctional agents within in vitro models for bone regeneration has allowed, to a certain degree, the control of cell fate towards osteogenic pathways. In this research, we applied three methods for functionalizing spatially-confined electrospun artificial microenvironments that presented relevant components of the native bone stem cell niche. The biological and osteogenic behaviors of mesenchymal stromal cells (MSCs) were investigated on electrospun micro-fabricated scaffolds functionalized with extracellular matrix (ECM) proteins (collagen I), glycosaminoglycans (heparin), and ceramic-based materials (bioglass). Collagen, heparin, and bioglass (BG) were successfully included in the models without modifying the fibrous structures offered by the polycaprolactone (PCL) scaffolds. Mesenchymal stromal cells (MSCs) were successfully seeded in all the biofunctional scaffolds and they showed an increase in alkaline phosphatase production when exposed to PCL/BG composites. This research demonstrates the feasibility of manufacturing smart and hierarchical artificial microenvironments for studying stem cell behavior and ultimately the potential of incorporating these artificial microenvironments into multifunctional membranes for bone tissue regeneration
Collapse
|
19
|
Afflerbach AK, Kiri MD, Detinis T, Maoz BM. Mesenchymal Stem Cells as a Promising Cell Source for Integration in Novel In Vitro Models. Biomolecules 2020; 10:E1306. [PMID: 32927777 PMCID: PMC7565384 DOI: 10.3390/biom10091306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
The human-relevance of an in vitro model is dependent on two main factors-(i) an appropriate human cell source and (ii) a modeling platform that recapitulates human in vivo conditions. Recent years have brought substantial advancements in both these aspects. In particular, mesenchymal stem cells (MSCs) have emerged as a promising cell source, as these cells can differentiate into multiple cell types, yet do not raise the ethical and practical concerns associated with other types of stem cells. In turn, advanced bioengineered in vitro models such as microfluidics, Organs-on-a-Chip, scaffolds, bioprinting and organoids are bringing researchers ever closer to mimicking complex in vivo environments, thereby overcoming some of the limitations of traditional 2D cell cultures. This review covers each of these advancements separately and discusses how the integration of MSCs into novel in vitro platforms may contribute enormously to clinical and fundamental research.
Collapse
Affiliation(s)
- Ann-Kristin Afflerbach
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Faculty of Biosciences, Universität Heidelberg, 69120 Heidelberg, Germany
| | - Mark D. Kiri
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Tahir Detinis
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
20
|
Bello-Rodriguez C, Wittig O, Diaz-Solano D, Bolaños P, Cardier JE. A 3D construct based on mesenchymal stromal cells, collagen microspheres and plasma clot supports the survival, proliferation and differentiation of hematopoietic cells in vivo. Cell Tissue Res 2020; 382:499-507. [PMID: 32789682 DOI: 10.1007/s00441-020-03265-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 07/22/2020] [Indexed: 01/09/2023]
Abstract
The hematopoietic niche is a specialized microenvironment that supports the survival, proliferation and differentiation of hematopoietic stem progenitor cells (HSPCs). Three-dimensional (3D) models mimicking hematopoiesis might allow in vitro and in vivo studies of the hematopoietic (HP) process. Here, we investigate the capacity of a 3D construct based on non-adherent murine bone marrow mononuclear cells (NA-BMMNCs), mesenchymal stromal cells (MSCs) and collagen microspheres (CMs), all embedded into plasma clot (PC) to support in vitro and in vivo hematopoiesis. Confocal analysis of the 3D hematopoietic construct (3D-HPC), cultured for 24 h, showed MSC lining the CM and the NA-BMMNCs closely associated with MSC. In vivo hematopoiesis was examined in 3D-HPC subcutaneously implanted in mice and harvested at different intervals. Hematopoiesis in the 3D-HPC was evaluated by histology, cell morphology, flow cytometry, confocal microscopy and hematopoietic colony formation assay. 3D-HPC implants were integrated and vascularized in the host tissue, after 3 months of implantation. Histological studies showed the presence of hematopoietic tissue with the presence of mature blood cells. Cells from 3D-HPC showed viability greater than 90%, expressed HSPCs markers, and formed hematopoietic colonies, in vitro. Confocal microscopy studies showed that MSCs adhered to the CM and NA-BMMNCs were scattered across the 3D-HPC area and in close association with MSC. In conclusion, the 3D-HPC mimics a hematopoietic niche supporting the survival, proliferation and differentiation of HSPCs, in vivo. 3D-HPC may allow evaluation of regulatory mechanisms involved in hematopoiesis.
Collapse
Affiliation(s)
- Carlos Bello-Rodriguez
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas, 1020-A, Venezuela.,Facultad de Ciencias, Universidad Central de Venezuela, Caracas, 1080, Venezuela
| | - Olga Wittig
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas, 1020-A, Venezuela
| | - Dylana Diaz-Solano
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas, 1020-A, Venezuela
| | - Pura Bolaños
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, 1020-A, Venezuela
| | - Jose E Cardier
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas, 1020-A, Venezuela. .,Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 20632, Caracas, 1020-A, Venezuela.
| |
Collapse
|
21
|
Gilchrist AE, Harley BA. Connecting secretome to hematopoietic stem cell phenotype shifts in an engineered bone marrow niche. Integr Biol (Camb) 2020; 12:175-187. [PMID: 32556172 PMCID: PMC7384206 DOI: 10.1093/intbio/zyaa013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/21/2020] [Accepted: 05/08/2020] [Indexed: 01/06/2023]
Abstract
Hematopoietic stem cells (HSCs) primarily reside in the bone marrow, where they receive external cues from their local microenvironment. The complex milieu of biophysical cues, cellular components and cell-secreted factors regulates the process by which HSC produce the blood and immune system. We previously showed direct coculture of primary murine hematopoietic stem and progenitor cells with a population of marrow-derived mesenchymal stromal and progenitor cells (MSPCs) in a methacrylamide-functionalized gelatin (GelMA) hydrogel improves hematopoietic progenitor maintenance. However, the mechanism by which MSPCs influenced HSC fate decisions remained unknown. Herein, we report the use of proteomic analysis to correlate HSC phenotype to a broad candidate pool of 200 soluble factors produced by combined mesenchymal and hematopoietic progeny. Partial least squares regression (PLSR), along with an iterative filter method, identified TGFβ-1, MMP-3, c-RP and TROY as positively correlated with HSC maintenance. Experimentally, we then observe exogenous stimulation of HSC monocultures in GelMA hydrogels with these combined cytokines increases the ratio of hematopoietic progenitors to committed progeny after a 7-day culture 7.52 ± 3.65-fold compared to non-stimulated monocultures. Findings suggest a cocktail of the downselected cytokines amplifies hematopoietic maintenance potential of HSCs beyond that of MSPC-secreted factors alone. This work integrates empirical and computation methods to identify cytokine combinations to improve HSC maintenance within an engineered HSC niche, suggesting a route toward identifying feeder-free culture platforms for HSC expansion. Insight Hematopoietic stem cells within an artificial niche receive maintenance cues in the form of soluble factors from hematopoietic and mesenchymal progeny. Applying a proteomic regression analysis, we identify a reduced set of soluble factors correlated to maintenance of a hematopoietic phenotype during culture in a biomaterial model of the bone marrow niche. We identify a minimum factor cocktail that promotes hematopoietic maintenance potential in a gelatin-based culture, regardless of the presence of mesenchymal feeder cells. By combining empirical and computational methods, we report an experimentally feasible number of factors from a large dataset, enabling exogenous integration of soluble factors into an engineered hematopoietic stem cell for enhanced maintenance potential of a quiescent stem cell population.
Collapse
Affiliation(s)
- Aidan E. Gilchrist
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
22
|
Kwak JG, Lee J. Thermoresponsive Inverted Colloidal Crystal Hydrogel Scaffolds for Lymphoid Tissue Engineering. Adv Healthc Mater 2020; 9:e1901556. [PMID: 32017462 PMCID: PMC7103457 DOI: 10.1002/adhm.201901556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/05/2020] [Indexed: 12/15/2022]
Abstract
Inverted colloidal crystal (ICC) hydrogel scaffolds represent unique opportunities in modeling lymphoid tissues and expanding hematopoietic-lymphoid cells. Fully interconnected spherical pore arrays direct the formation of stromal networks and facilitate interactions between stroma and hematopoietic-lymphoid cells. However, due to the intricate architecture of these materials, release of expanded cells is restricted and requires mechanical disruption or chemical dissolution of the hydrogel scaffold. One potent biomaterials strategy to release pore-entrapped hematopoietic-lymphoid cells without breaking the scaffolds apart is to transiently increase the dimensions of these materials using stimuli-responsive polymers. Having this mindset, thermoresponsive ICC scaffolds that undergo rapid (<1 min) and substantial (>300%) diameter change over a physiological temperature range (4-37 °C) by using poly(N-isopropylacrylamide) (PNIPAM) with nanogel crosslinkers is developed. For a proof-of-concept study, the stromal niche by creating osteospheroids, aggregates of osteoblasts, and bone chips is first replicated, and subsequently Nalm-6 model hematopoietic-lymphoid cells are introduced. A sixfold increase in cell count is harvested when ICC hydrogel scaffolds are expanded without termination of the established 3D stromal cell culture. It is envisioned that thermoresponsive ICC hydrogel scaffolds will enable for scalable and sustainable ex vivo expansion of hematopoietic-lymphoid cells.
Collapse
Affiliation(s)
- Jun-Goo Kwak
- Molecular and Cellular Biology Graduate Program, Institute for Applied Life Sciences, University of Massachusetts Amherst, Life Sciences Laboratories N560, 240 Thatcher Road, Amherst, MA, 01003, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Molecular and Cellular Biology Graduate Program, Institute for Applied Life Sciences, University of Massachusetts Amherst, Life Sciences Laboratories N567, 240 Thatcher Road, Amherst, MA, 01003, USA
| |
Collapse
|
23
|
Peterson NC, Mahalingaiah PK, Fullerton A, Di Piazza M. Application of microphysiological systems in biopharmaceutical research and development. LAB ON A CHIP 2020; 20:697-708. [PMID: 31967156 DOI: 10.1039/c9lc00962k] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Within the last 10 years, several tissue microphysiological systems (MPS) have been developed and characterized for retention of morphologic characteristics and specific gene/protein expression profiles from their natural in vivo state. Once developed, their utility is typically further tested by comparing responses to known toxic small-molecule pharmaceuticals in efforts to develop strategies for further toxicity testing of compounds under development. More recently, application of this technology in biopharmaceutical (large molecules) development is beginning to be more appreciated. In this review, we describe some of the advances made for tissue-specific MPS and outline the advantages and challenges of applying and further developing MPS technology in preclinical biopharmaceutical research.
Collapse
Affiliation(s)
- Norman C Peterson
- Clinical Pharmacology and Safety Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA.
| | | | | | - Matteo Di Piazza
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Rd, Ridgefield, CT 06877, USA
| |
Collapse
|
24
|
On-chip recapitulation of clinical bone marrow toxicities and patient-specific pathophysiology. Nat Biomed Eng 2020; 4:394-406. [PMID: 31988457 PMCID: PMC7160021 DOI: 10.1038/s41551-019-0495-z] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/22/2019] [Indexed: 12/27/2022]
Abstract
The inaccessibility of living bone marrow hampers the study of its pathophysiology under myelotoxic stress induced by drugs, radiation or genetic mutations. Here, we show that a vascularized human bone-marrow-on-a-chip supports the differentiation and maturation of multiple blood-cell lineages over 4 weeks while improving CD34+ cell maintenance, and that it recapitulates aspects of marrow injury, including myeloerythroid toxicity after clinically relevant exposures to chemotherapeutic drugs and ionizing radiation as well as marrow recovery after drug-induced myelosuppression. The chip comprises a fluidic channel filled with a fibrin gel in which CD34+ cells and bone-marrow-derived stromal cells are co-cultured, a parallel channel lined by human vascular endothelium and perfused with culture medium, and a porous membrane separating the two channels. We also show that bone-marrow chips containing cells from patients with the rare genetic disorder Shwachman–Diamond syndrome reproduced key haematopoietic defects and led to the discovery of a neutrophil-maturation abnormality. As an in vitro model of haematopoietic dysfunction, the bone-marrow-on-a-chip may serve as a human-specific alternative to animal testing for the study of bone-marrow pathophysiology.
Collapse
|
25
|
Ribeiro-Filho AC, Levy D, Ruiz JLM, Mantovani MDC, Bydlowski SP. Traditional and Advanced Cell Cultures in Hematopoietic Stem Cell Studies. Cells 2019; 8:cells8121628. [PMID: 31842488 PMCID: PMC6953118 DOI: 10.3390/cells8121628] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 01/09/2023] Open
Abstract
Hematopoiesis is the main function of bone marrow. Human hematopoietic stem and progenitor cells reside in the bone marrow microenvironment, making it a hotspot for the development of hematopoietic diseases. Numerous alterations that correspond to disease progression have been identified in the bone marrow stem cell niche. Complex interactions between the bone marrow microenvironment and hematopoietic stem cells determine the balance between the proliferation, differentiation and homeostasis of the stem cell compartment. Changes in this tightly regulated network can provoke malignant transformation. However, our understanding of human hematopoiesis and the associated niche biology remains limited due to accessibility to human material and the limits of in vitro culture models. Traditional culture systems for human hematopoietic studies lack microenvironment niches, spatial marrow gradients, and dense cellularity, rendering them incapable of effectively translating marrow physiology ex vivo. This review will discuss the importance of 2D and 3D culture as a physiologically relevant system for understanding normal and abnormal hematopoiesis.
Collapse
Affiliation(s)
- Antonio Carlos Ribeiro-Filho
- Organoid Development Team, Center of Innovation and Translational Medicine (CIMTRA), University of São Paulo School of Medicine, Sao Paulo 05360-130, Brazil; (A.C.R.-F.); (M.d.C.M.)
| | - Débora Levy
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), University of São Paulo School of Medicine, Sao Paulo 05403-900, Brazil;
| | - Jorge Luis Maria Ruiz
- Life and Nature Science Institute, Federal University of Latin American Integration-UNILA, Foz de Iguaçú, PR 858570-901, Brazil;
| | - Marluce da Cunha Mantovani
- Organoid Development Team, Center of Innovation and Translational Medicine (CIMTRA), University of São Paulo School of Medicine, Sao Paulo 05360-130, Brazil; (A.C.R.-F.); (M.d.C.M.)
| | - Sérgio Paulo Bydlowski
- Organoid Development Team, Center of Innovation and Translational Medicine (CIMTRA), University of São Paulo School of Medicine, Sao Paulo 05360-130, Brazil; (A.C.R.-F.); (M.d.C.M.)
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), University of São Paulo School of Medicine, Sao Paulo 05403-900, Brazil;
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, Brazil
- Correspondence:
| |
Collapse
|
26
|
Microphysiological systems in the evaluation of hematotoxicities during drug development. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Lu X, Lodi A, Konopleva M, Tiziani S. Three-Dimensional Leukemia Co-Culture System for In Vitro High-Content Metabolomics Screening. SLAS DISCOVERY 2019; 24:817-828. [PMID: 31345091 DOI: 10.1177/2472555219860446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Metabolomics is increasingly applied to investigate different individuals and time-dependent responses to environmental stimuli. Rapid data acquisition and improved detection limits of direct infusion mass spectrometry (DIMS) are paving the way for applications of metabolomics in preclinical screening, opening new opportunities in drug discovery and personalized medicine. Three-dimensional (3D) cell culture systems, which mimic the in vivo cell microenvironment, are well recognized as tissue and organ substitutes. Here, we investigated cell viability and induction of reactive oxygen species (ROS) in stromal cells cultured in various 3D systems as well as the standard monolayer culture to evaluate which system provides the most favorable growing conditions. The selected 3D system was then tested for use in 3D co-culture of leukemia and stromal cells for DIMS-based high-throughput/high-content metabolic drug screens. The NanobioMatrix-poly(ε-caprolactone) (NBM-PCL) scaffold resulted in the lowest ROS production, supported rapid cell proliferation, and was suitable for the 96- and 384-well plate formats. Doxorubicin treatment in leukemia co-cultured with stromal cells induced some unique metabolic responses that drastically differed from those observed in leukemia cells alone. The DIMS results also showed that the drug-induced metabolic modulations in both normal and cancer cells were weakened by co-culturing even at high treatment doses, thereby demonstrating the value of the 3D co-culture high-content metabolic drug screen. In conclusion, we optimized a high sample throughput method for 3D co-culture with a DIMS-based high-content metabolic drug screen and drug development.
Collapse
Affiliation(s)
- Xiyuan Lu
- 1 Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA.,2 Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Alessia Lodi
- 1 Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA.,2 Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Marina Konopleva
- 3 Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stefano Tiziani
- 1 Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA.,2 Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
28
|
Scala S, Aiuti A. In vivo dynamics of human hematopoietic stem cells: novel concepts and future directions. Blood Adv 2019; 3:1916-1924. [PMID: 31239246 PMCID: PMC6595260 DOI: 10.1182/bloodadvances.2019000039] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/14/2019] [Indexed: 02/06/2023] Open
Abstract
Unveiling the mechanisms and the cellular dynamics at the basis of human hematopoietic homeostasis has been a main focus for the scientific community since the discovery of a pool of multipotent hematopoietic stem cells (HSCs) capable of sustaining the hematopoietic output throughout life and after transplantation. Recently, new works shed light on the (1) differentiation paths, (2) size and replication rate of human HSC population at steady state, and (3) role of the distinct subpopulations comprising the hematopoietic stem and progenitor cell reservoir after transplantation. These papers exploited cutting-edge technologies, including vector integration site clonal tracking, spontaneous mutations, and deep transcriptome profiling. Here we discuss the latest updates in human hematopoietic system biology and in vivo dynamics, highlighting novel concepts and common findings deriving from different approaches and the future directions of these studies. Taken together, this information contributed to partially resolving the complexity of the in vivo HSC behavior and has major implications for HSC transplantation and gene therapy as well as for the development of future therapies.
Collapse
Affiliation(s)
- Serena Scala
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and
- Pediatric Immunohematology and Stem Cell Programme, IRCCS San Raffaele Scientific Institute, Milan, Italy; and
- Medical School, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
29
|
Marino S, Bishop RT, de Ridder D, Delgado-Calle J, Reagan MR. 2D and 3D In Vitro Co-Culture for Cancer and Bone Cell Interaction Studies. Methods Mol Biol 2019; 1914:71-98. [PMID: 30729461 DOI: 10.1007/978-1-4939-8997-3_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Co-culture assays are used to study the mutual interaction between cells in vitro. This chapter describes 2D and 3D co-culture systems used to study cell-cell signaling crosstalk between cancer cells and bone marrow adipocytes, osteoblasts, osteoclasts, and osteocytes. The chapter provides a step-by-step guide to the most commonly used cell culture techniques, functional assays, and gene expression.
Collapse
Affiliation(s)
- Silvia Marino
- Division Hematology Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Ryan T Bishop
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Daniëlle de Ridder
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Jesus Delgado-Calle
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michaela R Reagan
- Center for Molecular Medicine, Maine Medical Centre Research Institute, Scarborough, ME, USA
| |
Collapse
|
30
|
Polini A, Del Mercato LL, Barra A, Zhang YS, Calabi F, Gigli G. Towards the development of human immune-system-on-a-chip platforms. Drug Discov Today 2019; 24:517-525. [PMID: 30312743 PMCID: PMC6440212 DOI: 10.1016/j.drudis.2018.10.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 01/22/2023]
Abstract
Organ-on-a-chip (OoCs) platforms could revolutionize drug discovery and might ultimately become essential tools for precision therapy. Although many single-organ and interconnected systems have been described, the immune system has been comparatively neglected, despite its pervasive role in the body and the trend towards newer therapeutic products (i.e., complex biologics, nanoparticles, immune checkpoint inhibitors, and engineered T cells) that often cause, or are based on, immune reactions. In this review, we recapitulate some distinctive features of the immune system before reviewing microfluidic devices that mimic lymphoid organs or other organs and/or tissues with an integrated immune system component.
Collapse
Affiliation(s)
- Alessandro Polini
- Dipartimento di Matematica e Fisica E. De Giorgi, University of Salento, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy.
| | - Loretta L Del Mercato
- CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Adriano Barra
- Dipartimento di Matematica e Fisica E. De Giorgi, University of Salento, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; INFN, Sezione di Lecce, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; INdAM (GNFM), Sezione di Lecce, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Franco Calabi
- CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Giuseppe Gigli
- Dipartimento di Matematica e Fisica E. De Giorgi, University of Salento, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| |
Collapse
|
31
|
Biomimetic microspheres for 3D mesenchymal stem cell culture and characterization. Colloids Surf B Biointerfaces 2019; 177:68-76. [PMID: 30711761 DOI: 10.1016/j.colsurfb.2019.01.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 11/21/2022]
Abstract
Stem cells reside in niches, specialized microenvironments that sustain and regulate their fate. Extracellular matrix (ECM), paracrine factors or other cells are key niche regulating elements. As the conventional 2D cell culture lacks these elements, it can alter the properties of naïve stem cells. In this work we designed a novel biomimetic microenvironment for cell culture, consisting of magnetic microspheres, prepared with acrylates and acrylic acid copolymers and functionalized with fibronectin or hyaluronic acid as ECM coatings. To characterize cell proliferation and adhesion, porcine mesenchymal stem cells (MSCs) were grown with the different microspheres. The results showed that the 3D environments presented similar proliferation to the 2D culture and that fibronectin allows cell adhesion, while hyaluronic acid hinders it. In the 3D environments, cells reorganize the microspheres to grow in aggregates, highlighting the advantages of microspheres as 3D environments and allowing the cells to adapt the environment to their requirements.
Collapse
|
32
|
Abstract
This review briefly describes the last decades of experimental work on the thymus. Given the histological complexity of this organ, the multiple embryological origins of its cellular components and its role in carefully regulating T lymphocyte maturation and function, methods to dissect and understand this complexity have been developed through the years. The possibility to study ex vivo the thymus organ function has been achieved by developing Fetal Thymus Organ Cultures (FTOC). Subsequently, the combination of organ disaggregation and reaggregation in vitro represented by Reaggregate Thymus Organ cultures (RTOC) allowed mixing cellular components from different genetic backgrounds. Moreover, RTOC allowed dissecting the different stromal and hematological components to study the interactions between Major Histocompatibility Complex (MHC) molecules and the T-cell receptors during thymocytes selection. In more recent years, prospective isolation of stromal cells and thymocytes at different stages of development made it possible to explore and elucidate the molecular and cellular players in both the developing and adult thymus. Finally, the appearance of novel cell sources such as embryonic stem (ES) cells and more recently induced pluripotent stem (iPS) cells has opened new scenarios in modelling thymus development and regeneration strategies. Most of the work described was carried out in rodents and the current challenge is to develop equivalent or even more informative assays and tools in entirely human model systems.
Collapse
|
33
|
Torisawa YS. Microfluidic Organs-on-Chips to Reconstitute Cellular Microenvironments. Bioanalysis 2019. [DOI: 10.1007/978-981-13-6229-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
34
|
da Conceicao Ribeiro R, Pal D, Ferreira AM, Gentile P, Benning M, Dalgarno K. Reactive jet impingement bioprinting of high cell density gels for bone microtissue fabrication. Biofabrication 2018; 11:015014. [DOI: 10.1088/1758-5090/aaf625] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
35
|
Wang Q, Zhao G, Xing Z, Zhan J, Ma J. Comparative evaluation of the osteogenic capacity of human mesenchymal stem cells from bone marrow and umbilical cord tissue. Exp Ther Med 2018; 17:764-772. [PMID: 30651861 DOI: 10.3892/etm.2018.6975] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been extensively investigated in the field of regenerative medicine. Human bone MSCs (BMSCs) have become a common type of seed cell for bone tissue engineering. However, the viability and cell number of BMSCs are negatively correlated with donor age, and as the extraction process is painful, this method has not been widely used. As human umbilical cord MSCs (UCMSCs) may be harvested inexpensively and inexhaustibly, the present study evaluated and compared the regenerative potential of UCMSCs and BMSCs to determine whether UCMSCs may be used as a novel cell type for bone regeneration. In the present study, the proliferation and osteogenic capacity of BMSCs and UCMSCs was compared in vitro. BMSCs and UCMSCs were respectively combined with biofunctionalized macroporous calcium phosphate cement, and their bone regenerative potentials were determined by investigating their capacity for ectopic bone formation in a nude mouse model as well as their efficacy in a rat model of tibia bone defect. The extent of bone regeneration was examined by X-ray, histological and immunohistochemical analyses. The results revealed that UCMSCs exhibited a good osteogenic differentiation potential, similarly to that of BMSCs, and that UCMSCs were able to contribute to the regeneration of bone and blood vessels. Furthermore, no significant differences were identified between BMSCs and UCMSCs in terms of their bone regenerative effect.
Collapse
Affiliation(s)
- Qian Wang
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Gang Zhao
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Zijun Xing
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Juming Zhan
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| | - Jie Ma
- Institute of Biomedical Science, Tianjin Kang Ting Biological Engineering Co., Ltd., Tianjin 300385, P.R. China
| |
Collapse
|
36
|
Development of solvent-casting particulate leaching (SCPL) polymer scaffolds as improved three-dimensional supports to mimic the bone marrow niche. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 96:153-165. [PMID: 30606521 DOI: 10.1016/j.msec.2018.10.086] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 07/23/2018] [Accepted: 10/27/2018] [Indexed: 12/29/2022]
Abstract
The need for new approaches to investigate ex vivo the causes and effects of tumor and to achieve improved cancer treatments and medical therapies is particularly urgent for malignant pathologies such as lymphomas and leukemias, whose tissue initiator cells interact with the stroma creating a three-dimensional (3D) protective environment that conventional mono- and bi-dimensional (2D) models are not able to simulate realistically. The solvent-casting particulate leaching (SCPL) technique, that is already a standard method to produce polymer-based scaffolds for bone tissue repair, is proposed here to fabricate innovative 3D porous structures to mimic the bone marrow niche in vitro. Two different polymers, namely a rigid polymethyl methacrylate (PMMA) and a flexible polyurethane (PU), were evaluated to the purpose, whereas NaCl, in the form of common salt table, resulted to be an efficient porogen. The adoption of an appropriate polymer-to-salt ratio, experimentally defined as 1:4 for both PMMA and PU, gave place to a rich and interconnected porosity, ranging between 82.1 vol% and 91.3 vol%, and the choice of admixing fine-grained or coarse-grained salt powders allowed to control the final pore size. The mechanical properties under compression load were affected both by the polymer matrix and by the scaffold's architecture, with values of the elastic modulus indicatively varying between 29 kPa and 1283 kPa. Preliminary tests performed with human stromal HS-5 cells co-cultured with leukemic cells allowed us to conclude that stromal cells grown associated to the supports keep their well-known protective and pro-survival effect on cancer cells, indicating that these devices can be very useful to mimic the bone marrow microenvironment and therefore to assess the efficacy of novel therapies in pre-clinical studies.
Collapse
|
37
|
Kiaie N, Aghdam RM, Tafti SHA, Gorabi AM. Stem Cell-Mediated Angiogenesis in Tissue Engineering Constructs. Curr Stem Cell Res Ther 2018; 14:249-258. [PMID: 30394215 DOI: 10.2174/1574888x13666181105145144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/09/2018] [Accepted: 10/31/2018] [Indexed: 11/22/2022]
Abstract
Angiogenesis has always been a concern in the field of tissue engineering. Poor vascularization of engineered constructs is a problem for the clinical success of these structures. Among the various methods employed to induce angiogenesis, stem cells provide a promising tool for the future. The present review aims to present the application of stem cells in the induction of angiogenesis. Additionally, it summarizes recent advancements in stem cell-mediated angiogenesis of different tissue engineering constructs.
Collapse
Affiliation(s)
- Nasim Kiaie
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran.,Department of Tissue Engineering, Amirkabir University of Technology, Tehran 15875, Iran
| | - Rouhollah M Aghdam
- School of Metallurgy & Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Seyed H Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita M Gorabi
- Department of Basic and Clinical Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Iandolo D, Pennacchio FA, Mollo V, Rossi D, Dannhauser D, Cui B, Owens RM, Santoro F. Electron Microscopy for 3D Scaffolds-Cell Biointerface Characterization. ACTA ACUST UNITED AC 2018; 3:e1800103. [PMID: 32627375 DOI: 10.1002/adbi.201800103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/31/2018] [Indexed: 12/18/2022]
Abstract
Cell fate is largely determined by interactions that occur at the interface between cells and their surrounding microenvironment. For this reason, especially in the field of tissue-engineering, there is a growing interest in developing techniques that allow evaluating cell-material interaction at the nanoscale, particularly focusing on cell adhesion processes. While for 2D culturing systems a consolidated series of tools already satisfy this need, in 3D environments, more closely recapitulating complex in vivo structures, there is still a lack of procedures furthering the comprehension of cell-material interactions. Here, the use of scanning electron microscopy coupled with a focused ion beam (SEM/FIB) for the characterization of cell interactions with 3D scaffolds obtained by different fabrication techniques is reported for the first time. The results clearly show the capability of the developed approach to preserve and finely resolve scaffold-cell interfaces highlighting details such as plasma membrane arrangement, extracellular matrix architecture and composition, and cellular structures playing a role in cell adhesion to the surface. It is anticipated that the developed approach will be relevant for the design of efficient cell-instructive platforms in the study of cellular guidance strategies for tissue-engineering applications as well as for in vitro 3D models.
Collapse
Affiliation(s)
- Donata Iandolo
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK
| | - Fabrizio A Pennacchio
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125, Italy
| | - Valentina Mollo
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125, Italy
| | - Domenico Rossi
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125, Italy
| | - David Dannhauser
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125, Italy
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, CA, 94305, USA
| | - Roisin M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK
| | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125, Italy
| |
Collapse
|
39
|
Zhang H, Zhou Y, Zhang W, Wang K, Xu L, Ma H, Deng Y. Construction of vascularized tissue-engineered bone with a double-cell sheet complex. Acta Biomater 2018; 77:212-227. [PMID: 30017924 DOI: 10.1016/j.actbio.2018.07.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 06/25/2018] [Accepted: 07/10/2018] [Indexed: 12/14/2022]
Abstract
A double-cell sheet (DCS) complex composed of an osteogenic cell sheet and a vascular endothelial cell sheet with osteogenesis and blood vessel formation potential was developed in this study. The osteogenic and vascular endothelial cell sheets were obtained after induced culture of rabbit adipose-derived mesenchymal stem cells. The osteogenic cell sheet showed positive alizarin red, von Kossa, and alkaline phosphatase (ALP) staining. The vascular endothelial cell sheet exhibited visible W-P bodies in the cells, the expression of CD31 was positive, and a vascular mesh structure was spontaneously formed in a Matrigel matrix. The subcutaneous transplantation results for four groups of DCS and DCS-coral hydroxyapatite (CHA) complexes, and the CHA scaffold group in nude mice revealed mineralization of collagen fibers and vascularization in each group at 12 weeks, but the degrees of mineralization and vascularization showed differences among groups. The pattern involving endothelial cell sheets covered with osteogenic cell sheets, group B, exhibited the best results. In addition, the degree of mineralization of the DCS-CHA complexes was more mature than those of the same group of DCS complexes and the CHA scaffold, and the capillary number was greater than those of the same group of DCS complexes and the CHA scaffold. Therefore, the CHA scaffold strengthened the osteogenesis and blood vessel formation potential of the DCS complexes. Meanwhile, the DCS complexes also promoted the osteogenesis and blood vessel formation potential of the CHA scaffold. This study will provide a basis for building vascularized tissue-engineered bone for bone defect therapy. STATEMENT OF SIGNIFICANCE This study developed a double-cell sheet (DCS) complex composed of an osteogenic cell sheet and a vascular endothelial cell sheet with osteogenesis and blood vessel formation potential. Osteogenic and vascular endothelial cell sheets were obtained after induced culture of rabbit adipose-derived mesenchymal stem cells. The DCS complex and DCS-CHA complex exhibited osteogenic and blood vessel formation potential in vivo. CHA enhanced the osteogenesis and blood vessel formation abilities of the DCS complexes in vivo. Meanwhile, the DCS complexes also promoted the osteogenesis and blood vessel formation potential of the CHA scaffold. Group B of the DCS complexes and DCS-CHA complexes exhibited the best osteogenesis and blood vessel formation abilities.
Collapse
|
40
|
Bray LJ, Secker C, Murekatete B, Sievers J, Binner M, Welzel PB, Werner C. Three-Dimensional In Vitro Hydro- and Cryogel-Based Cell-Culture Models for the Study of Breast-Cancer Metastasis to Bone. Cancers (Basel) 2018; 10:cancers10090292. [PMID: 30150545 PMCID: PMC6162532 DOI: 10.3390/cancers10090292] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022] Open
Abstract
Bone is the most common site for breast-cancer invasion and metastasis, and it causes severe morbidity and mortality. A greater understanding of the mechanisms leading to bone-specific metastasis could improve therapeutic strategies and thus improve patient survival. While three-dimensional in vitro culture models provide valuable tools to investigate distinct heterocellular and environmental interactions, sophisticated organ-specific metastasis models are lacking. Previous models used to investigate breast-to-bone metastasis have relied on 2.5D or singular-scaffold methods, constraining the in situ mimicry of in vitro models. Glycosaminoglycan-based gels have demonstrated outstanding potential for tumor-engineering applications. Here, we developed advanced biphasic in vitro microenvironments that mimic breast-tumor tissue (MCF-7 and MDA-MB-231 in a hydrogel) spatially separated with a mineralized bone construct (human primary osteoblasts in a cryogel). These models allow distinct advantages over former models due to the ability to observe and manipulate cellular migration towards a bone construct. The gels allow for the binding of adhesion-mediating peptides and controlled release of signaling molecules. Moreover, mechanical and architectural properties can be tuned to manipulate cell function. These results demonstrate the utility of these biomimetic microenvironment models to investigate heterotypic cell⁻cell and cell⁻matrix communications in cancer migration to bone.
Collapse
Affiliation(s)
- Laura J Bray
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Australia.
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Australia.
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Queensland University of Technology (QUT), 2 George Street, Brisbane 4001, Australia.
- Translational Research Institute, Mater Research Institute-University of Queensland, 37 Kent Street, Woolloongabba 4102, Australia.
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straβe 6, 01069 Dresden, Germany.
| | - Constanze Secker
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straβe 6, 01069 Dresden, Germany.
| | - Berline Murekatete
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Australia.
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Australia.
| | - Jana Sievers
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straβe 6, 01069 Dresden, Germany.
| | - Marcus Binner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straβe 6, 01069 Dresden, Germany.
| | - Petra B Welzel
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straβe 6, 01069 Dresden, Germany.
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straβe 6, 01069 Dresden, Germany.
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraβe 105, 01307 Dresden, Germany.
| |
Collapse
|
41
|
Two-Photon Laser Polymerization: From Fundamentals to Biomedical Application in Tissue Engineering and Regenerative Medicine. J Appl Biomater Funct Mater 2018; 10:55-65. [DOI: 10.5301/jabfm.2012.9278] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2012] [Indexed: 12/22/2022] Open
Abstract
Three-dimensional material microstructuring by femtosecond laser-induced two-photon polymerization is emerging as an important tool in biomedicine. During two-photon polymerization, a tightly focused femtosecond laser pulse induces a crosslinking photoreaction in the polymer confined within the focal volume. As a rapid-prototyping technique, two-photon polymerization enables the fabrication of truly arbitrary three-dimensional micro- and nano-structures directly from computer models, with a spatial resolution down to 100 nm. In this review, we discuss the fundamentals, experimental methods, and materials used for two-photon polymerization; in addition, we present some applications of this technology related to microfluidics and to biomaterial scaffolds for tissue engineering and regenerative medicine.
Collapse
|
42
|
Bourgine PE, Klein T, Paczulla AM, Shimizu T, Kunz L, Kokkaliaris KD, Coutu DL, Lengerke C, Skoda R, Schroeder T, Martin I. In vitro biomimetic engineering of a human hematopoietic niche with functional properties. Proc Natl Acad Sci U S A 2018; 115:E5688-E5695. [PMID: 29866839 PMCID: PMC6016789 DOI: 10.1073/pnas.1805440115] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In adults, human hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow (BM) microenvironment. Our understanding of human hematopoiesis and the associated niche biology remains limited, due to human material accessibility and limits of existing in vitro culture models. The establishment of an in vitro BM system would offer an experimentally accessible and tunable platform to study human hematopoiesis. Here, we develop a 3D engineered human BM analog by recapitulating some of the hematopoietic niche elements. This includes a bone-like scaffold, functionalized by human stromal and osteoblastic cells and by the extracellular matrix they deposited during perfusion culture in bioreactors. The resulting tissue exhibited compositional and structural features of human BM while supporting the maintenance of HSPCs. This was associated with a compartmentalization of phenotypes in the bioreactor system, where committed blood cells are released into the liquid phase and HSPCs preferentially reside within the engineered BM tissue, establishing physical interactions with the stromal compartment. Finally, we demonstrate the possibility to perturb HSPCs' behavior within our 3D niches by molecular customization or injury simulation. The developed system enables the design of advanced, tunable in vitro BM proxies for the study of human hematopoiesis.
Collapse
Affiliation(s)
- Paul E Bourgine
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, 4058 Basel, Switzerland
| | - Thibaut Klein
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Anna M Paczulla
- Stem Cells and Hematopoiesis, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Takafumi Shimizu
- Experimental Hematology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Leo Kunz
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, 4058 Basel, Switzerland
| | - Konstantinos D Kokkaliaris
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, 4058 Basel, Switzerland
| | - Daniel L Coutu
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, 4058 Basel, Switzerland
| | - Claudia Lengerke
- Stem Cells and Hematopoiesis, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Radek Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, 4058 Basel, Switzerland;
| | - Ivan Martin
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland;
| |
Collapse
|
43
|
Stem Cells for Osteochondral Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:219-240. [DOI: 10.1007/978-3-319-76735-2_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
44
|
Braham MV, Ahlfeld T, Akkineni AR, Minnema MC, Dhert WJ, Öner FC, Robin C, Lode A, Gelinsky M, Alblas J. Endosteal and Perivascular Subniches in a 3D Bone Marrow Model for Multiple Myeloma. Tissue Eng Part C Methods 2018; 24:300-312. [DOI: 10.1089/ten.tec.2017.0467] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Maaike V.J. Braham
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tilman Ahlfeld
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - A. Rahul Akkineni
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Monique C. Minnema
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Wouter J.A. Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - F. Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Catherine Robin
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
45
|
Kotha S, Sun S, Adams A, Hayes B, Phong KT, Nagao R, Reems JA, Gao D, Torok-Storb B, López JA, Zheng Y. Microvasculature-directed thrombopoiesis in a 3D in vitro marrow microenvironment. PLoS One 2018; 13:e0195082. [PMID: 29617409 PMCID: PMC5884538 DOI: 10.1371/journal.pone.0195082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 03/17/2018] [Indexed: 12/03/2022] Open
Abstract
Vasculature is an interface between the circulation and the hematopoietic tissue providing the means for hundreds of billions of blood cells to enter the circulation every day in a regulated fashion. The precise mechanisms that control the interactions of hematopoietic cells with the vessel wall are largely undefined. Here, we report on the development of an in vitro 3D human marrow vascular microenvironment (VME) to study hematopoietic trafficking and the release of blood cells, specifically platelets. We show that mature megakaryocytes from aspirated marrow as well as megakaryocytes differentiated in culture from CD34+ cells can be embedded in a collagen matrix containing engineered microvessels to create a thrombopoietic VME. These megakaryocytes continue to mature, penetrate the vessel wall, and release platelets into the vessel lumen. This process can be blocked with the addition of antibodies specific for CXCR4, indicating that CXCR4 is required for megakaryocyte migration, though whether it is sufficient is unclear. The 3D marrow VME system shows considerable potential for mechanistic studies defining the role of marrow vasculature in thrombopoiesis. Through a stepwise addition or removal of individual marrow components, this model provides potential to define key pathways responsible for the release of platelets and other blood cells.
Collapse
Affiliation(s)
- Surya Kotha
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Sijie Sun
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- Bloodworks Research Institute, Seattle, Washington, United States of America
| | - Amie Adams
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Brian Hayes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kiet T. Phong
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Ryan Nagao
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Jo-Anna Reems
- Bloodworks Research Institute, Seattle, Washington, United States of America
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
| | - Beverly Torok-Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - José A. López
- Bloodworks Research Institute, Seattle, Washington, United States of America
- Department of Medicine (Hematology), University of Washington, Seattle, Washington, United States of America
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
46
|
Karimpoor M, Yebra-Fernandez E, Parhizkar M, Orlu M, Craig D, Khorashad JS, Edirisinghe M. Alginate foam-based three-dimensional culture to investigate drug sensitivity in primary leukaemia cells. J R Soc Interface 2018; 15:20170928. [PMID: 29695605 PMCID: PMC5938583 DOI: 10.1098/rsif.2017.0928] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/28/2018] [Indexed: 01/14/2023] Open
Abstract
The development of assays for evaluating the sensitivity of leukaemia cells to anti-cancer agents is becoming an important aspect of personalized medicine. Conventional cell cultures lack the three-dimensional (3D) structure of the bone marrow (BM), the extracellular matrix and stromal components which are crucial for the growth and survival of leukaemia stem cells. To accurately predict the sensitivity of the leukaemia cells in an in vitro assay a culturing system containing the essential components of BM is required. In this study, we developed a porous calcium alginate foam-based scaffold to be used for 3D culture. The new 3D culture was shown to be cell compatible as it supported the proliferation of both normal haematopoietic and leukaemia cells. Our cell differential assay for myeloid markers showed that the porous foam-based 3D culture enhanced myeloid differentiation in both leukaemia and normal haematopoietic cells compared to two-dimensional culture. The foam-based scaffold reduced the sensitivity of the leukaemia cells to the tested antileukaemia agents in K562 and HL60 leukaemia cell line model and also primary myeloid leukaemia cells. This observation supports the application of calcium alginate foams as scaffold components of the 3D cultures for investigation of sensitivity to antileukaemia agents in primary myeloid cells.
Collapse
Affiliation(s)
- Mahroo Karimpoor
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
- Centre for Haematology, Department of Medicine, Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Eva Yebra-Fernandez
- Molecular Pathology, North West London Pathology, Hammersmith Hospital, London, W12 0HS, UK
| | - Maryam Parhizkar
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Mine Orlu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Duncan Craig
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Jamshid S Khorashad
- Centre for Haematology, Department of Medicine, Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Molecular Pathology, North West London Pathology, Hammersmith Hospital, London, W12 0HS, UK
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| |
Collapse
|
47
|
Geraili A, Jafari P, Hassani MS, Araghi BH, Mohammadi MH, Ghafari AM, Tamrin SH, Modarres HP, Kolahchi AR, Ahadian S, Sanati-Nezhad A. Controlling Differentiation of Stem Cells for Developing Personalized Organ-on-Chip Platforms. Adv Healthc Mater 2018; 7. [PMID: 28910516 DOI: 10.1002/adhm.201700426] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/01/2017] [Indexed: 01/09/2023]
Abstract
Organ-on-chip (OOC) platforms have attracted attentions of pharmaceutical companies as powerful tools for screening of existing drugs and development of new drug candidates. OOCs have primarily used human cell lines or primary cells to develop biomimetic tissue models. However, the ability of human stem cells in unlimited self-renewal and differentiation into multiple lineages has made them attractive for OOCs. The microfluidic technology has enabled precise control of stem cell differentiation using soluble factors, biophysical cues, and electromagnetic signals. This study discusses different tissue- and organ-on-chip platforms (i.e., skin, brain, blood-brain barrier, bone marrow, heart, liver, lung, tumor, and vascular), with an emphasis on the critical role of stem cells in the synthesis of complex tissues. This study further recaps the design, fabrication, high-throughput performance, and improved functionality of stem-cell-based OOCs, technical challenges, obstacles against implementing their potential applications, and future perspectives related to different experimental platforms.
Collapse
Affiliation(s)
- Armin Geraili
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
| | - Parya Jafari
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
- Department of Electrical Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohsen Sheikh Hassani
- Department of Systems and Computer Engineering; Carleton University; 1125 Colonel By Drive Ottawa K1S 5B6 ON Canada
| | - Behnaz Heidary Araghi
- Department of Materials Science and Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Mohammad Ghafari
- Department of Stem Cells and Developmental Biology; Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology; Tehran 16635-148 Iran
| | - Sara Hasanpour Tamrin
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
- Center for Bioengineering Research and Education; Biomedical Engineering Program; University of Calgary; Calgary T2N 1N4 AB Canada
| |
Collapse
|
48
|
Ahadian S, Civitarese R, Bannerman D, Mohammadi MH, Lu R, Wang E, Davenport-Huyer L, Lai B, Zhang B, Zhao Y, Mandla S, Korolj A, Radisic M. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. Adv Healthc Mater 2018; 7. [PMID: 29034591 DOI: 10.1002/adhm.201700506] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/15/2017] [Indexed: 12/11/2022]
Abstract
Significant advances in biomaterials, stem cell biology, and microscale technologies have enabled the fabrication of biologically relevant tissues and organs. Such tissues and organs, referred to as organ-on-a-chip (OOC) platforms, have emerged as a powerful tool in tissue analysis and disease modeling for biological and pharmacological applications. A variety of biomaterials are used in tissue fabrication providing multiple biological, structural, and mechanical cues in the regulation of cell behavior and tissue morphogenesis. Cells derived from humans enable the fabrication of personalized OOC platforms. Microscale technologies are specifically helpful in providing physiological microenvironments for tissues and organs. In this review, biomaterials, cells, and microscale technologies are described as essential components to construct OOC platforms. The latest developments in OOC platforms (e.g., liver, skeletal muscle, cardiac, cancer, lung, skin, bone, and brain) are then discussed as functional tools in simulating human physiology and metabolism. Future perspectives and major challenges in the development of OOC platforms toward accelerating clinical studies of drug discovery are finally highlighted.
Collapse
Affiliation(s)
- Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Robert Civitarese
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Dawn Bannerman
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Rick Lu
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Erika Wang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Locke Davenport-Huyer
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Ben Lai
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Boyang Zhang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Serena Mandla
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| |
Collapse
|
49
|
Sieber S, Wirth L, Cavak N, Koenigsmark M, Marx U, Lauster R, Rosowski M. Bone marrow-on-a-chip: Long-term culture of human haematopoietic stem cells in a three-dimensional microfluidic environment. J Tissue Eng Regen Med 2017; 12:479-489. [PMID: 28658717 DOI: 10.1002/term.2507] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 06/13/2017] [Accepted: 06/16/2017] [Indexed: 12/17/2022]
Abstract
Multipotent haematopoietic stem and progenitor cells (HSPCs) are the source for all blood cell types. The bone marrow stem cell niche in which the HSPCs are maintained is known to be vital for their maintenance. Unfortunately, to date, no in vitro model exists that accurately mimics the aspects of the bone marrow niche and simultaneously allows the long-term culture of HSPCs. In this study, a novel three-dimensional coculture model is presented, based on a hydroxyapatite coated zirconium oxide scaffold, comprising of human mesenchymal stromal cells (MSCs) and cord blood derived HSPCs, enabling successful HSPC culture for a time span of 28 days within the microfluidic multiorgan chip. The HSPCs were found to stay in their primitive state (CD34+ CD38- ) and capable of granulocyte, erythrocyte, macrophage, megakaryocyte colony formation. Furthermore, a microenvironment was formed bearing molecular and structural similarity to the in vivo bone marrow niche containing extracellular matrix and signalling molecules known to play an important role in HSPC homeostasis. Here, a novel human in vitro bone marrow model is presented for the first time, capable of long-term culture of primitive HSPCs in a microfluidic environment.
Collapse
Affiliation(s)
- Stefan Sieber
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charitè Campus Virchow Klinikum, Berlin, Germany
| | - Lorenz Wirth
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | - Nino Cavak
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | - Marielle Koenigsmark
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | | | - Roland Lauster
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | - Mark Rosowski
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| |
Collapse
|
50
|
Hoch AI, Duhr R, Di Maggio N, Mehrkens A, Jakob M, Wendt D. Expansion of Bone Marrow Mesenchymal Stromal Cells in Perfused 3D Ceramic Scaffolds Enhances In Vivo Bone Formation. Biotechnol J 2017; 12. [PMID: 28881093 DOI: 10.1002/biot.201700071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/31/2017] [Indexed: 01/05/2023]
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSC), when expanded directly within 3D ceramic scaffolds in perfusion bioreactors, more reproducibly form bone when implanted in vivo as compared to conventional expansion on 2D polystyrene dishes/flasks. Since the bioreactor-based expansion on 3D ceramic scaffolds encompasses multiple aspects that are inherently different from expansion on 2D polystyrene, we aimed to decouple the effects of specific parameters among these two model systems. We assessed the effects of the: 1) 3D scaffold vs. 2D surface; 2) ceramic vs. polystyrene materials; and 3) BMSC niche established within the ceramic pores during in vitro culture, on subsequent in vivo bone formation. While BMSC expanded on 3D polystyrene scaffolds in the bioreactor could maintain their in vivo osteogenic potential, results were similar as BMSC expanded in monolayer on 2D polystyrene, suggesting little influence of the scaffold 3D environment. Bone formation was most reproducible when BMSC are expanded on 3D ceramic, highlighting the influence of the ceramic substrate. The presence of a pre-formed niche within the scaffold pores had negligible effects on the in vivo bone formation. The results of this study allow a greater understanding of the parameters required for perfusion bioreactor-based manufacturing of osteogenic grafts for clinical applications.
Collapse
Affiliation(s)
- Allison I Hoch
- Department of Biomedicine, University of Basel, University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.,University of California, Davis, CA, USA
| | - Ralph Duhr
- Department of Biomedicine, University of Basel, University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Nunzia Di Maggio
- Department of Biomedicine, University of Basel, University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Arne Mehrkens
- Department of Biomedicine, University of Basel, University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.,Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marcel Jakob
- Department of Biomedicine, University of Basel, University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.,Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - David Wendt
- Department of Biomedicine, University of Basel, University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.,Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, University Hospital Basel, Basel, Switzerland
| |
Collapse
|