51
|
Williams MAC, Mair DB, Lee W, Lee E, Kim DH. Engineering Three-Dimensional Vascularized Cardiac Tissues. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:336-350. [PMID: 33559514 PMCID: PMC9063162 DOI: 10.1089/ten.teb.2020.0343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/08/2021] [Indexed: 12/23/2022]
Abstract
Heart disease is one of the largest burdens to human health worldwide and has very limited therapeutic options. Engineered three-dimensional (3D) vascularized cardiac tissues have shown promise in rescuing cardiac function in diseased hearts and may serve as a whole organ replacement in the future. One of the major obstacles in reconstructing these thick myocardial tissues to a clinically applicable scale is the integration of functional vascular networks capable of providing oxygen and nutrients throughout whole engineered constructs. Without perfusion of oxygen and nutrient flow throughout the entire engineered tissue not only is tissue viability compromised, but also overall tissue functionality is lost. There are many supporting technologies and approaches that have been developed to create vascular networks such as 3D bioprinting, co-culturing hydrogels, and incorporation of soluble angiogenic factors. In this state-of-the-art review, we discuss some of the most current engineered vascular cardiac tissues reported in the literature and future directions in the field. Impact statement The field of cardiac tissue engineering is rapidly evolving and is now closer than ever to having engineered tissue models capable of predicting preclinical responses to therapeutics, modeling diseases, and being used as a means of rescuing cardiac function following injuries to the native myocardium. However, a major obstacle of engineering thick cardiac tissue remains to be the integration of functional vasculature. In this review, we highlight seminal and recently published works that have influenced and pushed the field of cardiac tissue engineering toward achieving vascularized functional tissues.
Collapse
Affiliation(s)
| | - Devin B. Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Wonjae Lee
- Department of Neurosurgery, Stanford School of Medicine, Stanford, California, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
52
|
The Mammary Gland: Basic Structure and Molecular Signaling during Development. Int J Mol Sci 2022; 23:ijms23073883. [PMID: 35409243 PMCID: PMC8998991 DOI: 10.3390/ijms23073883] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023] Open
Abstract
The mammary gland is a compound, branched tubuloalveolar structure and a major characteristic of mammals. The mammary gland has evolved from epidermal apocrine glands, the skin glands as an accessory reproductive organ to support postnatal survival of offspring by producing milk as a source of nutrition. The mammary gland development begins during embryogenesis as a rudimentary structure that grows into an elementary branched ductal tree and is embedded in one end of a larger mammary fat pad at birth. At the onset of ovarian function at puberty, the rudimentary ductal system undergoes dramatic morphogenetic change with ductal elongation and branching. During pregnancy, the alveolar differentiation and tertiary branching are completed, and during lactation, the mature milk-producing glands eventually develop. The early stages of mammary development are hormonal independent, whereas during puberty and pregnancy, mammary gland development is hormonal dependent. We highlight the current understanding of molecular regulators involved during different stages of mammary gland development.
Collapse
|
53
|
Zhan Z, Liu Z, Nan H, Li J, Xie Y, Hu C. Heterogeneous spheroids with tunable interior morphologies by droplet-based microfluidics. Biofabrication 2022; 14. [PMID: 35290971 DOI: 10.1088/1758-5090/ac5e12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/15/2022] [Indexed: 11/11/2022]
Abstract
Heterogeneous spheroids that mimic the complex three-dimensional environment of natural tissues are needed in various biomedical applications. Geometric cues from cellular matrix play invaluable roles in governing cell behavior and phenotype. However, the structural complexity of interior morphologies of spheroids is currently limited due to poor spatial resolution of positioning/orientation of cellular constructs. Here, a coaxial capillary microfluidic device is developed to generate gelatin methacrylate (GelMA) microspheres with tunable dimensions and interior morphologies, such as core-shell, or microspheres with interior undulated wavy, or spiral canals, by manipulating the two-phase flow of hydrogel precursor solution and methylcellulose solution. The formation of diverse and exquisite interior morphologies is caused by the interacting viscous instabilities of the two-phase flow in the microfluidic system, followed by water-in-oil emulsion and photo-initiated polymerization. Polyethylene glycol diacrylate (PEGDA) is incorporated into the GelMA solution to tune the mechanical properties of the fabricated microspheres, and an optimized concentration of PEGDA is confirmed by evaluating the in vitro proliferation and vascularization of human umbilical endothelial cells. Further, a heterogeneous spheroid with spiral blood vessel lumen is constructed to demonstrate the versatility and potential of the proposed droplet-based microfluidic approach for building functional tissue constructs.
Collapse
Affiliation(s)
- Zhen Zhan
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, No 1088, xueyuan Blvd., Xili, Nanshan District, Shenzhen, Guangdong, China, Shenzhen, Guangdong, 518055, CHINA
| | - Zeyang Liu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, No 1088, xueyuan Blvd., Xili, Nanshan District, Shenzhen, Guangdong, China, Shenzhen, Guangdong, 518055, CHINA
| | - Haochen Nan
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, No 1088, xueyuan Blvd., Xili, Nanshan District, Shenzhen, Guangdong, China, Shenzhen, Guangdong, 518055, CHINA
| | - Jianjie Li
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, No 1088, xueyuan Blvd., Xili, Nanshan District, Shenzhen, Guangdong, China, Shenzhen, Guangdong, 518055, CHINA
| | - Yuan Xie
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, No 1088, xueyuan Blvd., Xili, Nanshan District, Shenzhen, Guangdong, China, Shenzhen, Guangdong, 518055, CHINA
| | - Chengzhi Hu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, No 1088, xueyuan Blvd., Xili, Nanshan District, Shenzhen, Guangdong, China, Shenzhen, Guangdong, 518055, CHINA
| |
Collapse
|
54
|
Dynamic miRNA Landscape Links Mammary Gland Development to the Regulation of Milk Protein Expression in Mice. Animals (Basel) 2022; 12:ani12060727. [PMID: 35327124 PMCID: PMC8944794 DOI: 10.3390/ani12060727] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Milk synthesis is vital for maintaining the normal growth of newborn animals. Abnormal mammary gland development leads to a decrease in female productivity and the overall productivity of animal husbandry. This study characterized the dynamic miRNA expression profile during the process of mammary gland development, and identified a novel miRNA regulating expression of β-casein—an important milk protein. The results are valuable for studying mammary gland development, increasing milk production, improving the survival rate of pups, and promoting the development of animal husbandry. Abstract Mammary gland morphology varies considerably between pregnancy and lactation status, e.g., virgin to pregnant and lactation to weaning. Throughout these critical developmental phases, the mammary glands undergo remodeling to accommodate changes in milk production capacity, which is positively correlated with milk protein expression. The purpose of this study was to investigate the microRNA (miRNA) expression profiles in female ICR mice’s mammary glands at the virgin stage (V), day 16 of pregnancy (P16d), day 12 of lactation (L12d), day 1 of forced weaning (FW1d), and day 3 of forced weaning (FW3d), and to identify the miRNAs regulating milk protein gene expression. During the five stages of testing, 852 known miRNAs and 179 novel miRNAs were identified in the mammary glands. Based on their expression patterns, the identified miRNAs were grouped into 12 clusters. The expression pattern of cluster 1 miRNAs was opposite to that of milk protein genes in mammary glands in all five different stages. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that the predicted target genes of cluster 1 miRNAs were related to murine mammary gland development and lactation. Furthermore, fluorescence in situ hybridization (FISH) analysis revealed that the novel-mmu-miR424-5p, which belongs to the cluster 1 miRNAs, was expressed in murine mammary epithelial cells. The dual-luciferase reporter assay revealed that an important milk protein gene—β-casein (CSN2)—was regarded as one of the likely targets for the novel-mmu-miR424-5p. This study analyzed the expression patterns of miRNAs in murine mammary glands throughout five critical developmental stages, and discovered a novel miRNA involved in regulating the expression of CSN2. These findings contribute to an enhanced understanding of the developmental biology of mammary glands, providing guidelines for increasing lactation efficiency and milk quality.
Collapse
|
55
|
Jang H, Kim SH, Koh Y, Yoon KJ. Engineering Brain Organoids: Toward Mature Neural Circuitry with an Intact Cytoarchitecture. Int J Stem Cells 2022; 15:41-59. [PMID: 35220291 PMCID: PMC8889333 DOI: 10.15283/ijsc22004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/19/2022] [Indexed: 11/23/2022] Open
Abstract
The emergence of brain organoids as a model system has been a tremendously exciting development in the field of neuroscience. Brain organoids are a gateway to exploring the intricacies of human-specific neurogenesis that have so far eluded the neuroscience community. Regardless, current culture methods have a long way to go in terms of accuracy and reproducibility. To perfectly mimic the human brain, we need to recapitulate the complex in vivo context of the human fetal brain and achieve mature neural circuitry with an intact cytoarchitecture. In this review, we explore the major challenges facing the current brain organoid systems, potential technical breakthroughs to advance brain organoid techniques up to levels similar to an in vivo human developing brain, and the future prospects of this technology.
Collapse
Affiliation(s)
- Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Seo Hyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Youmin Koh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
- KAIST-Wonjin Cell Therapy Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| |
Collapse
|
56
|
Mierke CT. Viscoelasticity, Like Forces, Plays a Role in Mechanotransduction. Front Cell Dev Biol 2022; 10:789841. [PMID: 35223831 PMCID: PMC8864183 DOI: 10.3389/fcell.2022.789841] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Viscoelasticity and its alteration in time and space has turned out to act as a key element in fundamental biological processes in living systems, such as morphogenesis and motility. Based on experimental and theoretical findings it can be proposed that viscoelasticity of cells, spheroids and tissues seems to be a collective characteristic that demands macromolecular, intracellular component and intercellular interactions. A major challenge is to couple the alterations in the macroscopic structural or material characteristics of cells, spheroids and tissues, such as cell and tissue phase transitions, to the microscopic interferences of their elements. Therefore, the biophysical technologies need to be improved, advanced and connected to classical biological assays. In this review, the viscoelastic nature of cytoskeletal, extracellular and cellular networks is presented and discussed. Viscoelasticity is conceptualized as a major contributor to cell migration and invasion and it is discussed whether it can serve as a biomarker for the cells' migratory capacity in several biological contexts. It can be hypothesized that the statistical mechanics of intra- and extracellular networks may be applied in the future as a powerful tool to explore quantitatively the biomechanical foundation of viscoelasticity over a broad range of time and length scales. Finally, the importance of the cellular viscoelasticity is illustrated in identifying and characterizing multiple disorders, such as cancer, tissue injuries, acute or chronic inflammations or fibrotic diseases.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
57
|
3D Culture Platform for Enabling Large-Scale Imaging and Control of Cell Distribution into Complex Shapes by Combining 3D Printing with a Cube Device. MICROMACHINES 2022; 13:mi13020156. [PMID: 35208281 PMCID: PMC8875915 DOI: 10.3390/mi13020156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 12/10/2022]
Abstract
While organoid differentiation protocols have been widely developed, local control of initial cell seeding position and imaging of large-scale organoid samples with high resolution remain challenging. 3D bioprinting is an effective method to achieve control of cell positioning, but existing methods mainly rely on the use of synthetic hydrogels that could compromise the native morphogenesis of organoids. To address this problem, we developed a 3D culture platform that combines 3D printing with a cube device to enable an unrestricted range of designs to be formed in biological hydrogels. We demonstrated the formation of channels in collagen hydrogel in the cube device via a molding process using a 3D-printed water-soluble mold. The mold is first placed in uncured hydrogel solution, then easily removed by immersion in water after the gel around it has cured, thus creating a mold-shaped gap in the hydrogel. At the same time, the difficulty in obtaining high-resolution imaging on a large scale can also be solved as the cube device allows us to scan the tissue sample from multiple directions, so that the imaging quality can be enhanced without having to rely on higher-end microscopes. Using this developed technology, we demonstrated (1) mimicking vascular structure by seeding HUVEC on the inner walls of helix-shaped channels in collagen gels, and (2) multi-directional imaging of the vascular structure in the cube device. Thus, this paper describes a concerted method that simultaneously allows for the precise control of cell positioning in hydrogels for organoid morphogenesis, and the imaging of large-sized organoid samples. It is expected that the platform developed here can lead to advancements in organoid technology to generate organoids with more sophisticated structures.
Collapse
|
58
|
Abstract
Engineers point the way toward more complex and homogeneous intestinal organoids.
Collapse
Affiliation(s)
- Tyler R Huycke
- Department of Pharmaceutical Chemistry, University of California San Francisco, CA, USA
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, CA, USA
| |
Collapse
|
59
|
Gjorevski N, Nikolaev M, Brown TE, Mitrofanova O, Brandenberg N, DelRio FW, Yavitt FM, Liberali P, Anseth KS, Lutolf MP. Tissue geometry drives deterministic organoid patterning. Science 2022; 375:eaaw9021. [PMID: 34990240 DOI: 10.1126/science.aaw9021] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Epithelial organoids are stem cell–derived tissues that approximate aspects of real organs, and thus they have potential as powerful tools in basic and translational research. By definition, they self-organize, but the structures formed are often heterogeneous and irreproducible, which limits their use in the lab and clinic. We describe methodologies for spatially and temporally controlling organoid formation, thereby rendering a stochastic process more deterministic. Bioengineered stem cell microenvironments are used to specify the initial geometry of intestinal organoids, which in turn controls their patterning and crypt formation. We leveraged the reproducibility and predictability of the culture to identify the underlying mechanisms of epithelial patterning, which may contribute to reinforcing intestinal regionalization in vivo. By controlling organoid culture, we demonstrate how these structures can be used to answer questions not readily addressable with the standard, more variable, organoid models.
Collapse
Affiliation(s)
- N Gjorevski
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - M Nikolaev
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - T E Brown
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA.,BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - O Mitrofanova
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - N Brandenberg
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - F W DelRio
- Material, Physical, and Chemical Sciences Center, Sandia National Laboratories, Albuquerque, NM 87185, USA
| | - F M Yavitt
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA.,BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - P Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - K S Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA.,BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA
| | - M P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Institute of Chemical Sciences and Engineering, School of Basic Science (SB), EPFL, Lausanne, Switzerland
| |
Collapse
|
60
|
Guided Self-Assembly of ES-Derived Lung Progenitors into Biomimetic Tube Structures That Impact Cell Differentiation. Bioengineering (Basel) 2021; 8:bioengineering8120209. [PMID: 34940362 PMCID: PMC8698605 DOI: 10.3390/bioengineering8120209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 11/25/2022] Open
Abstract
Chemically directed differentiation of pluripotent stem cells (PSCs) into defined cell types is a potent strategy for creating regenerative tissue models and cell therapies. In vitro observations suggest that physical cues can augment directed differentiation. We recently demonstrated that confining human PSC-derived lung progenitor cells in a tube with a diameter that mimics those observed during lung development results in the alteration of cell differentiation towards SOX2−SOX9+ lung cells. Here we set out to assess the robustness of this geometric confinement effect with respect to different culture parameters in order to explore the corresponding changes in cell morphometry and determine the feasibility of using such an approach to enhance directed differentiation protocols. Culture of progenitor cells in polydimethylsiloxane (PDMS) tubes reliably induced self-organization into tube structures and was insensitive to a variety of extracellular matrix coatings. Cellular morphology and differentiation status were found to be sensitive to the diameter of tube cells that were cultured within but not to seeding density. These data suggest that geometric cues impose constraints on cells, homogenize cellular morphology, and influence fate status.
Collapse
|
61
|
Mierke CT. Viscoelasticity Acts as a Marker for Tumor Extracellular Matrix Characteristics. Front Cell Dev Biol 2021; 9:785138. [PMID: 34950661 PMCID: PMC8691700 DOI: 10.3389/fcell.2021.785138] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/23/2021] [Indexed: 12/28/2022] Open
Abstract
Biological materials such as extracellular matrix scaffolds, cancer cells, and tissues are often assumed to respond elastically for simplicity; the viscoelastic response is quite commonly ignored. Extracellular matrix mechanics including the viscoelasticity has turned out to be a key feature of cellular behavior and the entire shape and function of healthy and diseased tissues, such as cancer. The interference of cells with their local microenvironment and the interaction among different cell types relies both on the mechanical phenotype of each involved element. However, there is still not yet clearly understood how viscoelasticity alters the functional phenotype of the tumor extracellular matrix environment. Especially the biophysical technologies are still under ongoing improvement and further development. In addition, the effect of matrix mechanics in the progression of cancer is the subject of discussion. Hence, the topic of this review is especially attractive to collect the existing endeavors to characterize the viscoelastic features of tumor extracellular matrices and to briefly highlight the present frontiers in cancer progression and escape of cancers from therapy. Finally, this review article illustrates the importance of the tumor extracellular matrix mechano-phenotype, including the phenomenon viscoelasticity in identifying, characterizing, and treating specific cancer types.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
62
|
Wang R, Huang F, Wei W, Zhou Y, Ye Z, Yu L, Hu J, Cai C. Programmed Cell Death Ligand 1 Is Enriched in Mammary Stem Cells and Promotes Mammary Development and Regeneration. Front Cell Dev Biol 2021; 9:772669. [PMID: 34805179 PMCID: PMC8602569 DOI: 10.3389/fcell.2021.772669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Programmed cell death ligand 1 (PD-L1) is widely expressed in a variety of human tumors, and inhibition of the PD-L1/PD-1 pathway represents one of the most promising therapy for many types of cancer. However, the physiological function of PD-L1 in tissue development is still unclear, although PD-L1 mRNA is abundant in many tissues. To address this puzzle, we investigated the function of PD-L1 in mammary gland development. Interestingly, we found that PD-L1 is enriched in protein C receptor (Procr)-expressing mammary stem cells (MaSCs), and PD-L1-expressing mammary basal cells (PD-L1+ basal cells) exhibit robust mammary regeneration capacity in transplantation assay. The lineage tracing experiment showed that PD-L1+ cells can differentiate into all lineages of mammary epithelium cells, suggesting that PD-L1+ basal cells have the activities of MaSCs. Furthermore, PD-L1 deficiency significantly impairs mammary development and reduces mammary regeneration capacity of mammary basal cells, suggesting that PD-L1 is not only enriched in MaSCs but also improves activities of MaSCs. In summary, these results demonstrated that PD-L1 is enriched in MaSCs and promotes mammary gland development and regeneration. Mechanistically, our data indicated that PD-L1 expression is induced by continuous activation of Wnt/ß-catenin signaling. In conclusion, these results demonstrated that PD-L1 is a marker of MaSCs, and PD-L1 is essential for mammary development. Our study provides novel insight into the physiological functions of PD-L1 in tissue development.
Collapse
Affiliation(s)
- Ruirui Wang
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Fujing Huang
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Wei Wei
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yu Zhou
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Zi Ye
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Liya Yu
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Cheguo Cai
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China.,Dongguan and Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan, China
| |
Collapse
|
63
|
Cook BL, Chao CJ, Alford PW. Architecture-Dependent Mechano-Adaptation in Single Vascular Smooth Muscle Cells. J Biomech Eng 2021; 143:101002. [PMID: 33972987 DOI: 10.1115/1.4051117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Indexed: 01/03/2023]
Abstract
Arteries grow and remodel following mechanical perturbation. Vascular smooth muscle cells (VSMCs) within the artery undergo hyperplasia, hypertrophy, or change their contractility following sustained changes in loading. Experimental evidence in vivo and in vitro suggests that VSMCs grow and remodel to maintain a constant transmural stress, or "target" stress. This behavior is often described using a stress-dependent finite growth framework. Typically, computational models of arterial growth and remodeling account for VSMC behavior in a constrained mixture formulation that incorporates behavior of each component of the artery. However, these models do not account for differential VSMC architecture observed in situ, which may significantly influence growth and remodeling behavior. Here, we used cellular microbiaxial stretching (CμBS) to characterize how VSMCs with different cytoskeletal architectures respond to a sustained step change in strain. We find that VSMC F-actin architecture becomes more aligned following stretch and retains this alignment after 24 h. Further, we find that VSMC stress magnitude depends on cellular architecture. Qualitatively, however, stress behavior following stretch is consistent across cell architectures-stress increases following stretch and returns to prestretch magnitudes after 24 h. Finally, we formulated an architecture-dependent targeted growth law that accounts for experimentally measured cytoskeletal alignment and attributes stress evolution to individual fiber growth and find that this model robustly captures long-term stress evolution in single VSMCs. These results suggest that VSMC mechano-adaptation depends on cellular architecture, which has implications for growth and remodeling in regions of arteries with differential architecture, such as at bifurcations.
Collapse
Affiliation(s)
- Bernard L Cook
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Christina J Chao
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| |
Collapse
|
64
|
Langthasa J, Sarkar P, Narayanan S, Bhagat R, Vadaparty A, Bhat R. Extracellular matrix mediates moruloid-blastuloid morphodynamics in malignant ovarian spheroids. Life Sci Alliance 2021; 4:e202000942. [PMID: 34376568 PMCID: PMC8358442 DOI: 10.26508/lsa.202000942] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer metastasizes into peritoneum through dissemination of transformed epithelia as multicellular spheroids. Harvested from the malignant ascites of patients, spheroids exhibit startling features of organization typical to homeostatic glandular tissues: lumen surrounded by smoothly contoured and adhered epithelia. Herein, we demonstrate that cells of specific ovarian cancer lines in suspension, aggregate into dysmorphic solid "moruloid" clusters that permit intercellular movement, cell penetration, and interspheroidal coalescence. Moruloid clusters subsequently mature into "blastuloid" spheroids with smooth contours, a temporally dynamic lumen and immotile cells. Blastuloid spheroids neither coalesce nor allow cell penetration. Ultrastructural examination reveals a basement membrane-like extracellular matrix coat on the surface of blastuloid, but not moruloid, spheroids. Quantitative proteomics reveals down-regulation in ECM protein Fibronectin-1 associated with the moruloid-blastuloid transition; immunocytochemistry also confirms the relocalization of basement membrane ECM proteins: collagen IV and laminin to the surface of blastuloid spheroids. Fibronectin depletion accelerates, and enzymatic basement membrane debridement impairs, lumen formation, respectively. The regulation by ECM dynamics of the morphogenesis of cancer spheroids potentially influences the progression of the disease.
Collapse
Affiliation(s)
- Jimpi Langthasa
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Purba Sarkar
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Shruthi Narayanan
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Rahul Bhagat
- Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | | | - Ramray Bhat
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
65
|
Saleh J, Mercier B, Xi W. Bioengineering methods for organoid systems. Biol Cell 2021; 113:475-491. [PMID: 34580889 DOI: 10.1111/boc.202000119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 12/23/2022]
Abstract
Organoids have been widely used in fundamental, biomimetic, and therapeutic studies. These multicellular systems form via cell-autonomous self-organization where a cohort of stem cells undergoes in vivo-like proliferation, differentiation, and morphogenesis. They also recapitulate a series of physiological cell organization, complexity and functions that are untouchable by conventional bio-model systems using immortal cell lines. However, the development of organoids is often not easily controlled and their shape and size are yet fully physiological. Recent research has demonstrated that multiple bioengineering tools could be harnessed to control important internal and external cues that dictate stem cell behavior and stem-cell based organoid development. In this review, we introduce the current development of organoid systems and their potentials, as well as their limitations that impede their further utility in research and clinical fields. In comparison to conventional autonomous organoid system, we then review bioengineering approaches that offer improved control over organoid growth and development. We focus on the genetic editing tools that allow the program of build-in responses and phenotypes for organoid systems with enhanced physiological relevance. We also highlight the advances in bioengineering methods to modify cellular external milieus to generate desirable cell composition, 3D micro-architectures, and complex microfluidic systems. We conclude that the emerging biomimetic methods that employ multidisciplinary approaches could prevail in the future development of organoid systems.
Collapse
Affiliation(s)
- Jad Saleh
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Barbara Mercier
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Wang Xi
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| |
Collapse
|
66
|
Carberry BJ, Hergert JE, Yavitt FM, Hernandez JJ, Speckl KF, Bowman CN, McLeod RR, Anseth KS. 3D printing of sacrificial thioester elastomers using digital light processing for templating 3D organoid structures in soft biomatrices. Biofabrication 2021; 13:10.1088/1758-5090/ac1c98. [PMID: 34380115 PMCID: PMC8860055 DOI: 10.1088/1758-5090/ac1c98] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/11/2021] [Indexed: 01/02/2023]
Abstract
Biofabrication allows for the templating of structural features in materials on cellularly-relevant size scales, enabling the generation of tissue-like structures with controlled form and function. This is particularly relevant for growing organoids, where the application of biochemical and biomechanical stimuli can be used to guide the assembly and differentiation of stem cells and form architectures similar to the parent tissue or organ. Recently, ablative laser-scanning techniques was used to create 3D overhang features in collagen hydrogels at size scales of 10-100µm and supported the crypt-villus architecture in intestinal organoids. As a complementary method, providing advantages for high-throughput patterning, we printed thioester functionalized poly(ethylene glycol) (PEG) elastomers using digital light processing (DLP) and created sacrificial, 3D shapes that could be molded into soft (G' < 1000 Pa) hydrogel substrates. Specifically, three-arm 1.3 kDa PEG thiol and three-arm 1.6 kDa PEG norbornene, containing internal thioester groups, were photopolymerized to yield degradable elastomers. When incubated in a solution of 300 mM 2-mercaptoethanol (pH 9.0), 1 mm thick 10 mm diameter elastomer discs degraded in <2 h. Using DLP, arrays of features with critical dimensions of 37 ± 4µm, resolutions of 22 ± 5µm, and overhang structures as small as 50µm, were printed on the order of minutes. These sacrificial thioester molds with physiologically relevant features were cast-molded into Matrigel and subsequently degraded to create patterned void spaces with high fidelity. Intestinal stem cells (ISCs) cultured on the patterned Matrigel matrices formed confluent monolayers that conformed to the underlying pattern. DLP printed sacrificial thioester elastomer constructs provide a robust and rapid method to fabricate arrays of 3D organoid-sized features in soft tissue culture substrates and should enable investigations into the effect of epithelial geometry and spacing on the growth and differentiation of ISCs.
Collapse
Affiliation(s)
- Benjamin J Carberry
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, United States of America
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, United States of America
| | - John E Hergert
- Materials Science and Engineering, University of Colorado, Boulder, CO 80309, United States of America
| | - F Max Yavitt
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, United States of America
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, United States of America
| | - Juan J Hernandez
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, United States of America
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, United States of America
| | - Kelly F Speckl
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, United States of America
| | - Christopher N Bowman
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, United States of America
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, United States of America
- Materials Science and Engineering, University of Colorado, Boulder, CO 80309, United States of America
| | - Robert R McLeod
- Materials Science and Engineering, University of Colorado, Boulder, CO 80309, United States of America
- Department of Electrical, Computer and Energy Engineering, University of Colorado, Boulder, CO 80309, United States of America
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, United States of America
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, United States of America
| |
Collapse
|
67
|
Bouzin M, Zeynali A, Marini M, Sironi L, Scodellaro R, D’Alfonso L, Collini M, Chirico G. Multiphoton Laser Fabrication of Hybrid Photo-Activable Biomaterials. SENSORS 2021; 21:s21175891. [PMID: 34502787 PMCID: PMC8433654 DOI: 10.3390/s21175891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022]
Abstract
The possibility to shape stimulus-responsive optical polymers, especially hydrogels, by means of laser 3D printing and ablation is fostering a new concept of “smart” micro-devices that can be used for imaging, thermal stimulation, energy transducing and sensing. The composition of these polymeric blends is an essential parameter to tune their properties as actuators and/or sensing platforms and to determine the elasto-mechanical characteristics of the printed hydrogel. In light of the increasing demand for micro-devices for nanomedicine and personalized medicine, interest is growing in the combination of composite and hybrid photo-responsive materials and digital micro-/nano-manufacturing. Existing works have exploited multiphoton laser photo-polymerization to obtain fine 3D microstructures in hydrogels in an additive manufacturing approach or exploited laser ablation of preformed hydrogels to carve 3D cavities. Less often, the two approaches have been combined and active nanomaterials have been embedded in the microstructures. The aim of this review is to give a short overview of the most recent and prominent results in the field of multiphoton laser direct writing of biocompatible hydrogels that embed active nanomaterials not interfering with the writing process and endowing the biocompatible microstructures with physically or chemically activable features such as photothermal activity, chemical swelling and chemical sensing.
Collapse
Affiliation(s)
- Margaux Bouzin
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
| | - Amirbahador Zeynali
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
| | - Mario Marini
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
| | - Laura Sironi
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
| | - Riccardo Scodellaro
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
| | - Laura D’Alfonso
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
| | - Maddalena Collini
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
- Institute for Applied Sciences and Intelligent Systems, CNR, 80078 Pozzuoli, Italy
- Correspondence: (M.C.); (G.C.)
| | - Giuseppe Chirico
- Dipartimento di Fisica, Università degli studi di Milano-Bicocca, 20126 Milano, Italy; (M.B.); (A.Z.); (M.M.); (L.S.); (R.S.); (L.D.)
- Institute for Applied Sciences and Intelligent Systems, CNR, 80078 Pozzuoli, Italy
- Correspondence: (M.C.); (G.C.)
| |
Collapse
|
68
|
Engineering a Vascularized Hypoxic Tumor Model for Therapeutic Assessment. Cells 2021; 10:cells10092201. [PMID: 34571851 PMCID: PMC8468635 DOI: 10.3390/cells10092201] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/23/2023] Open
Abstract
Solid tumors in advanced cancer often feature a structurally and functionally abnormal vasculature through tumor angiogenesis, which contributes to cancer progression, metastasis, and therapeutic resistances. Hypoxia is considered a major driver of angiogenesis in tumor microenvironments. However, there remains a lack of in vitro models that recapitulate both the vasculature and hypoxia in the same model with physiological resemblance to the tumor microenvironment, while allowing for high-content spatiotemporal analyses for mechanistic studies and therapeutic evaluations. We have previously constructed a hypoxia microdevice that utilizes the metabolism of cancer cells to generate an oxygen gradient in the cancer cell layer as seen in solid tumor sections. Here, we have engineered a new composite microdevice-microfluidics platform that recapitulates a vascularized hypoxic tumor. Endothelial cells were seeded in a collagen channel formed by viscous fingering, to generate a rounded vascular lumen surrounding a hypoxic tumor section composed of cancer cells embedded in a 3-D hydrogel extracellular matrix. We demonstrated that the new device can be used with microscopy-based high-content analyses to track the vascular phenotypes, morphology, and sprouting into the hypoxic tumor section over a 7-day culture, as well as the response to different cancer/stromal cells. We further evaluated the integrity/leakiness of the vascular lumen in molecular delivery, and the potential of the platform to study the movement/trafficking of therapeutic immune cells. Therefore, our new platform can be used as a model for understanding tumor angiogenesis and therapeutic delivery/efficacy in vascularized hypoxic tumors.
Collapse
|
69
|
Abstract
Micropatterning encompasses a set of methods aimed at precisely controlling the spatial distribution of molecules onto the surface of materials. Biologists have borrowed the idea and adapted these methods, originally developed for electronics, to impose physical constraints on biological systems with the aim of addressing fundamental questions across biological scales from molecules to multicellular systems. Here, I approach this topic from a developmental biologist's perspective focusing specifically on how and why micropatterning has gained in popularity within the developmental biology community in recent years. Overall, this Primer provides a concise overview of how micropatterns are used to study developmental processes and emphasises how micropatterns are a useful addition to the developmental biologist's toolbox.
Collapse
Affiliation(s)
- Guillaume Blin
- Institute for Regeneration and Repair, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| |
Collapse
|
70
|
Siriwardane ML, Derosa K, Collins G, Pfister BJ. Engineering Fiber-Based Nervous Tissue Constructs for Axon Regeneration. Cells Tissues Organs 2021; 210:105-117. [PMID: 34198287 DOI: 10.1159/000515549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 03/02/2021] [Indexed: 11/19/2022] Open
Abstract
Biomaterial-based scaffolds used in nerve conduits including channels for confining regenerating axons and 3-dimensional (3D) gels as substrates for growth have made improvements in models of nerve repair. Many biomaterial strategies, however, continue to fall short of autologous nerve grafts, which remain the current gold standard in repairing severe nerve lesions (<20 mm). Intraluminal nerve conduit fibers have also shown considerable promise in directing regenerating axons in vitro and in vivo and have gained increasing interest for nerve repair. It is unknown, however, how growing axons respond to a fiber when encountered in a 3D environment. In this study, we considered a construct consisting of a compliant collagen hydrogel matrix and a fiber component to assess contact-guided axon growth. We investigated preferential axon outgrowth on synthetic and natural polymer fibers by utilizing small-diameter microfibers of poly-L-lactic acid and type I collagen representing 2 different fiber stiffnesses. We found that axons growing freely in a 3D hydrogel culture preferentially attach, turn and follow fibers with outgrowth rates and distances that far exceed outgrowth in a hydrogel alone. Wet-spun type I collagen from rat tail tendon performed the best, associated with highly aligned and accelerated outgrowth. This study also evaluated the response of dorsal root ganglion neurons from adult rats to provide data more relevant to axon regenerative potential in nerve repair. We found that ECM treatments on fibers enhanced the regeneration of adult axons indicating that both the physical and biochemical presentation of the fibers are essential for enhancing axon guidance and growth.
Collapse
Affiliation(s)
- Mevan L Siriwardane
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Kathleen Derosa
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - George Collins
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Bryan J Pfister
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
71
|
Mechanical compartmentalization of the intestinal organoid enables crypt folding and collective cell migration. Nat Cell Biol 2021; 23:745-757. [PMID: 34155382 PMCID: PMC7611697 DOI: 10.1038/s41556-021-00699-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 05/12/2021] [Indexed: 02/06/2023]
Abstract
Intestinal organoids capture essential features of the intestinal epithelium such as crypt folding, cellular compartmentalization and collective movements. Each of these processes and their coordination require patterned forces that are at present unknown. Here we map three-dimensional cellular forces in mouse intestinal organoids grown on soft hydrogels. We show that these organoids exhibit a non-monotonic stress distribution that defines mechanical and functional compartments. The stem cell compartment pushes the extracellular matrix and folds through apical constriction, whereas the transit amplifying zone pulls the extracellular matrix and elongates through basal constriction. The size of the stem cell compartment depends on the extracellular-matrix stiffness and endogenous cellular forces. Computational modelling reveals that crypt shape and force distribution rely on cell surface tensions following cortical actomyosin density. Finally, cells are pulled out of the crypt along a gradient of increasing tension. Our study unveils how patterned forces enable compartmentalization, folding and collective migration in the intestinal epithelium.
Collapse
|
72
|
Geuens T, Ruiter FAA, Schumacher A, Morgan FLC, Rademakers T, Wiersma LE, van den Berg CW, Rabelink TJ, Baker MB, LaPointe VLS. Thiol-ene cross-linked alginate hydrogel encapsulation modulates the extracellular matrix of kidney organoids by reducing abnormal type 1a1 collagen deposition. Biomaterials 2021; 275:120976. [PMID: 34198162 DOI: 10.1016/j.biomaterials.2021.120976] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/10/2021] [Accepted: 06/13/2021] [Indexed: 02/06/2023]
Abstract
Differentiated kidney organoids from induced pluripotent stem cells hold promise as a treatment for patients with kidney diseases. Before these organoids can be translated to the clinic, shortcomings regarding their cellular and extracellular compositions, and their developmental plateau need to be overcome. We performed a proteomic analysis on kidney organoids cultured for a prolonged culture time and we found a specific change in the extracellular matrix composition with increased expression of types 1a1, 2 and 6a1 collagen. Such an excessive accumulation of specific collagen types is a hallmark of renal fibrosis that causes a life-threatening pathological condition by compromising key functions of the human kidney. Here we hypothesized the need for a three-dimensional environment to grow the kidney organoids, which could better mimic the in vivo surroundings of the developing kidney than standard culture on an air-liquid interface. Encapsulating organoids for four days in a soft, thiol-ene cross-linked alginate hydrogel resulted in decreased type 1a1 collagen expression. Furthermore, the encapsulation did not result in any changes of organoid structural morphology. Using a biomaterial to modulate collagen expression allows for a prolonged kidney organoid culture in vitro and a reduction of abnormal type 1a1 collagen expression bringing kidney organoids closer to clinical application.
Collapse
Affiliation(s)
- Thomas Geuens
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, the Netherlands
| | - Floor A A Ruiter
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, the Netherlands; MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Maastricht, the Netherlands
| | - Anika Schumacher
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, the Netherlands
| | - Francis L C Morgan
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Maastricht, the Netherlands
| | - Timo Rademakers
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, the Netherlands
| | - Loes E Wiersma
- Department of Internal Medicine - Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Cathelijne W van den Berg
- Department of Internal Medicine - Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine - Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthew B Baker
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Maastricht, the Netherlands.
| | - Vanessa L S LaPointe
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
73
|
Ren G, Zheng X, Sharma V, Letson J, Nestor-Kalinoski AL, Furuta S. Loss of Nitric Oxide Induces Fibrogenic Response in Organotypic 3D Co-Culture of Mammary Epithelia and Fibroblasts-An Indicator for Breast Carcinogenesis. Cancers (Basel) 2021; 13:cancers13112815. [PMID: 34198735 PMCID: PMC8201212 DOI: 10.3390/cancers13112815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Fibrosis, which is often caused by chronic diseases and environmental substances, is closely associated with cancer. Thus, the development of a robust method allowing for deep studies of the linkage between fibrosis and cancer is essential. Here, we tested whether our novel three-dimensional (3D) co-culture of breast epithelia and fibroblasts would be a suitable model for that purpose. We compared the phenotypic effects of L-NAME, an inhibitor of nitric oxide (NO) production, on 3D mono- and co-cultures. We previously reported that prolonged NO depletion with L-NAME caused fibrosis and tumorigenesis in mouse mammary glands. Such in vivo effects of L-NAME were well recapitulated in 3D co-cultures, but not in 3D mono-cultures of epithelia and fibroblasts. These results support not only the essential roles of the presence of the stroma in cancer development, but also the utility of this co-culture in studying the causal relationship between fibrosis and cancer. Abstract Excessive myofibroblast activation, which leads to dysregulated collagen deposition and the stiffening of the extracellular matrix (ECM), plays pivotal roles in cancer initiation and progression. Cumulative evidence attests to the cancer-causing effects of a number of fibrogenic factors found in the environment, diseases and drugs. While identifying such factors largely depends on epidemiological studies, it would be of great importance to develop a robust in vitro method to demonstrate the causal relationship between fibrosis and cancer. Here, we tested whether our recently developed organotypic three-dimensional (3D) co-culture would be suitable for that purpose. This co-culture system utilizes the discontinuous ECM to separately culture mammary epithelia and fibroblasts in the discrete matrices to model the complexity of the mammary gland. We observed that pharmaceutical deprivation of nitric oxide (NO) in 3D co-cultures induced myofibroblast differentiation of the stroma as well as the occurrence of epithelial–mesenchymal transition (EMT) of the parenchyma. Such in vitro response to NO deprivation was unique to co-cultures and closely mimicked the phenotype of NO-depleted mammary glands exhibiting stromal desmoplasia and precancerous lesions undergoing EMT. These results suggest that this novel 3D co-culture system could be utilized in the deep mechanistic studies of the linkage between fibrosis and cancer.
Collapse
Affiliation(s)
- Gang Ren
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Xunzhen Zheng
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Vandana Sharma
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Joshua Letson
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Andrea L. Nestor-Kalinoski
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA;
| | - Saori Furuta
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
- Correspondence:
| |
Collapse
|
74
|
Finot L, Chanat E, Dessauge F. Mammary gland 3D cell culture systems in farm animals. Vet Res 2021; 52:78. [PMID: 34078471 PMCID: PMC8173821 DOI: 10.1186/s13567-021-00947-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/27/2021] [Indexed: 11/17/2022] Open
Abstract
In vivo study of tissue or organ biology in mammals is very complex and progress is slowed by poor accessibility of samples and ethical concerns. Fortunately, however, advances in stem cell identification and culture have made it possible to derive in vitro 3D “tissues” called organoids, these three-dimensional structures partly or fully mimicking the in vivo functioning of organs. The mammary gland produces milk, the source of nutrition for newborn mammals. Milk is synthesized and secreted by the differentiated polarized mammary epithelial cells of the gland. Reconstructing in vitro a mammary-like structure mimicking the functional tissue represents a major challenge in mammary gland biology, especially for farm animals for which specific agronomic questions arise. This would greatly facilitate the study of mammary gland development, milk secretion processes and pathological effects of viral or bacterial infections at the cellular level, all with the objective of improving milk production at the animal level. With this aim, various 3D cell culture models have been developed such as mammospheres and, more recently, efforts to develop organoids in vitro have been considerable. Researchers are now starting to draw inspiration from other fields, such as bioengineering, to generate organoids that would be more physiologically relevant. In this chapter, we will discuss 3D cell culture systems as organoids and their relevance for agronomic research.
Collapse
Affiliation(s)
- Laurence Finot
- PEGASE, INRAE, Institut Agro, 35590, Saint Gilles, France.
| | - Eric Chanat
- PEGASE, INRAE, Institut Agro, 35590, Saint Gilles, France
| | | |
Collapse
|
75
|
Inchanalkar S, Balasubramanian N. Adhesion-growth factor crosstalk regulates AURKB activation and ERK signalling in re-adherent fibroblasts. J Biosci 2021. [DOI: 10.1007/s12038-021-00164-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
76
|
Nerger BA, Jaslove JM, Elashal HE, Mao S, Košmrlj A, Link AJ, Nelson CM. Local accumulation of extracellular matrix regulates global morphogenetic patterning in the developing mammary gland. Curr Biol 2021; 31:1903-1917.e6. [PMID: 33705716 PMCID: PMC8119325 DOI: 10.1016/j.cub.2021.02.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/23/2020] [Accepted: 02/08/2021] [Indexed: 01/19/2023]
Abstract
The tree-like pattern of the mammary epithelium is formed during puberty through a process known as branching morphogenesis. Although mammary epithelial branching is stochastic and generates an epithelial tree with a random pattern of branches, the global orientation of the developing epithelium is predictably biased along the long axis of the gland. Here, we combine analysis of pubertal mouse mammary glands, a three-dimensional (3D)-printed engineered tissue model, and computational models of morphogenesis to investigate the origin and the dynamics of the global bias in epithelial orientation during pubertal mammary development. Confocal microscopy analysis revealed that a global bias emerges in the absence of pre-aligned networks of type I collagen in the fat pad and is maintained throughout pubertal development until the widespread formation of lateral branches. Using branching and annihilating random walk simulations, we found that the angle of bifurcation of terminal end buds (TEBs) dictates both the dynamics and the extent of the global bias in epithelial orientation. Our experimental and computational data demonstrate that a local increase in stiffness from the accumulation of extracellular matrix, which constrains the angle of bifurcation of TEBs, is sufficient to pattern the global orientation of the developing mammary epithelium. These data reveal that local mechanical properties regulate the global pattern of mammary epithelial branching and may provide new insight into the global patterning of other branched epithelia.
Collapse
Affiliation(s)
- Bryan A Nerger
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Jacob M Jaslove
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Hader E Elashal
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Sheng Mao
- Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing 100871, China; Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Andrej Košmrlj
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - A James Link
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
77
|
Pramanik D, Jolly MK, Bhat R. Matrix adhesion and remodeling diversifies modes of cancer invasion across spatial scales. J Theor Biol 2021; 524:110733. [PMID: 33933478 DOI: 10.1016/j.jtbi.2021.110733] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022]
Abstract
The metastasis of malignant epithelial tumors begins with the egress of transformed cells from the confines of their basement membrane (BM) to their surrounding collagen-rich stroma. Invasion can be morphologically diverse: when breast cancer cells are separately cultured within BM-like matrix, collagen I (Coll I), or a combination of both, they exhibit collective-, dispersed mesenchymal-, and a mixed collective-dispersed (multimodal)- invasion, respectively. In this paper, we asked how distinct these invasive modes are with respect to the cellular and microenvironmental cues that drive them. A rigorous computational exploration of invasion was performed within an experimentally motivated Cellular Potts-based modeling environment. The model comprised of adhesive interactions between cancer cells, BM- and Coll I-like extracellular matrix (ECM), and reaction-diffusion-based remodeling of ECM. The model outputs were parameters cognate to dispersed- and collective- invasion. A clustering analysis of the output distribution curated through a careful examination of subsumed phenotypes suggested at least four distinct invasive states: dispersed, papillary-collective, bulk-collective, and multimodal, in addition to an indolent/non-invasive state. Mapping input values to specific output clusters suggested that each of these invasive states are specified by distinct input signatures of proliferation, adhesion and ECM remodeling. In addition, specific input perturbations allowed transitions between the clusters and revealed the variation in the robustness between the invasive states. Our systems-level approach proffers quantitative insights into how the diversity in ECM microenvironments may steer invasion into diverse phenotypic modes during early dissemination of breast cancer and contributes to tumor heterogeneity.
Collapse
Affiliation(s)
- D Pramanik
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India; Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - M K Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - R Bhat
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
78
|
Leggett SE, Hruska AM, Guo M, Wong IY. The epithelial-mesenchymal transition and the cytoskeleton in bioengineered systems. Cell Commun Signal 2021; 19:32. [PMID: 33691719 PMCID: PMC7945251 DOI: 10.1186/s12964-021-00713-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/26/2021] [Indexed: 01/04/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is intrinsically linked to alterations of the intracellular cytoskeleton and the extracellular matrix. After EMT, cells acquire an elongated morphology with front/back polarity, which can be attributed to actin-driven protrusion formation as well as the gain of vimentin expression. Consequently, cells can deform and remodel the surrounding matrix in order to facilitate local invasion. In this review, we highlight recent bioengineering approaches to elucidate EMT and functional changes in the cytoskeleton. First, we review transitions between multicellular clusters and dispersed individuals on planar surfaces, which often exhibit coordinated behaviors driven by leader cells and EMT. Second, we consider the functional role of vimentin, which can be probed at subcellular length scales and within confined spaces. Third, we discuss the role of topographical patterning and EMT via a contact guidance like mechanism. Finally, we address how multicellular clusters disorganize and disseminate in 3D matrix. These new technologies enable controlled physical microenvironments and higher-resolution spatiotemporal measurements of EMT at the single cell level. In closing, we consider future directions for the field and outstanding questions regarding EMT and the cytoskeleton for human cancer progression. Video Abstract.
Collapse
Affiliation(s)
- Susan E Leggett
- Department of Chemical and Biological Engineering, Princeton University, William St, Princeton, NJ, 08544, USA
| | - Alex M Hruska
- School of Engineering, Center for Biomedical Engineering, and Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA
| | - Ming Guo
- Department of Mechanical Engineering, MIT, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Ian Y Wong
- School of Engineering, Center for Biomedical Engineering, and Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA.
| |
Collapse
|
79
|
Kulwatno J, Gong X, DeVaux R, Herschkowitz JI, Mills KL. An Organotypic Mammary Duct Model Capturing Matrix Mechanics-Dependent Ductal Carcinoma In Situ Progression. Tissue Eng Part A 2021; 27:454-466. [PMID: 33397202 DOI: 10.1089/ten.tea.2020.0239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a precancerous stage breast cancer, where abnormal cells are contained within the duct, but have not invaded into the surrounding tissue. However, only 30-40% of DCIS cases are likely to progress into an invasive ductal carcinoma (IDC), while the remainder are innocuous. Since little is known about what contributes to the transition from DCIS to IDC, clinicians and patients tend to opt for treatment, leading to concerns of overdiagnosis and overtreatment. In vitro models are currently being used to probe how DCIS transitions into IDC, but many models do not take into consideration the macroscopic tissue architecture and the biomechanical properties of the microenvironment. In this study, we modeled an organotypic mammary duct as a channel molded in a collagen matrix and lined with basement membrane. By adjusting the concentration of collagen (4 and 8 mg/mL), we modulated the stiffness and morphological properties of the matrix and examined how an assortment of breast cells, including the isogenic MCF10 series that spans the range from healthy to aggressive, behaved within our model. We observed distinct characteristics of breast cancer progression such as hyperplasia and invasion. Normal mammary epithelial cells (MCF10A) formed a single-cell layer on the lumen surface, whereas the most aggressive (MCF10CA1) were several cell layers thick. The model captured collagen concentration-dependent protrusive behaviors by the MCF10A and MCF10CA1 cells, as well as a known invasive cell line (MDA-MB-231). The MCF10A and MCF10CA1 cells extended protrusions into the lower collagen concentration matrix, while the MDA-MB-231 cells fully invaded matrices of either collagen concentration but to a greater distance in the higher collagen concentration matrix. Our results show that the model can recapitulate different stages of breast cancer progression and that the MCF10 series is adaptable to physiologically relevant in vitro studies, demonstrating the potential of both the model and cell lines to elucidate key factors that may contribute to understanding the transition from DCIS to IDC. Impact statement The success of early preventative measures for breast cancer has left patients susceptible to overdiagnosis and overtreatment. Limited knowledge of factors driving an invasive transition has inspired the development of in vitro models that accurately capture this phenomenon. However, current models tend to neglect the macroscopic architecture and biomechanical properties of the mammary duct. In this study, we introduce an organotypic model that recapitulates the cylindrical geometry of the tissue and the altered stroma seen in tumor microenvironments. Our model was able to capture distinct features associated with breast cancer progression, demonstrating its potential to uncover novel insights into disease progression.
Collapse
Affiliation(s)
- Jonathan Kulwatno
- Department of Biomedical Engineering, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA.,Center for Biotechnology and Interdisciplinary Studies, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Xiangyu Gong
- Center for Biotechnology and Interdisciplinary Studies, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Rebecca DeVaux
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Albany, New York, USA
| | - Jason I Herschkowitz
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Albany, New York, USA
| | - Kristen L Mills
- Center for Biotechnology and Interdisciplinary Studies, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| |
Collapse
|
80
|
Chhetri A, Rispoli JV, Lelièvre SA. 3D Cell Culture for the Study of Microenvironment-Mediated Mechanostimuli to the Cell Nucleus: An Important Step for Cancer Research. Front Mol Biosci 2021; 8:628386. [PMID: 33644116 PMCID: PMC7902798 DOI: 10.3389/fmolb.2021.628386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/11/2021] [Indexed: 11/21/2022] Open
Abstract
The discovery that the stiffness of the tumor microenvironment (TME) changes during cancer progression motivated the development of cell culture involving extracellular mechanostimuli, with the intent of identifying mechanotransduction mechanisms that influence cell phenotypes. Collagen I is a main extracellular matrix (ECM) component used to study mechanotransduction in three-dimensional (3D) cell culture. There are also models with interstitial fluid stress that have been mostly focusing on the migration of invasive cells. We argue that a major step for the culture of tumors is to integrate increased ECM stiffness and fluid movement characteristic of the TME. Mechanotransduction is based on the principles of tensegrity and dynamic reciprocity, which requires measuring not only biochemical changes, but also physical changes in cytoplasmic and nuclear compartments. Most techniques available for cellular rheology were developed for a 2D, flat cell culture world, hence hampering studies requiring proper cellular architecture that, itself, depends on 3D tissue organization. New and adapted measuring techniques for 3D cell culture will be worthwhile to study the apparent increase in physical plasticity of cancer cells with disease progression. Finally, evidence of the physical heterogeneity of the TME, in terms of ECM composition and stiffness and of fluid flow, calls for the investigation of its impact on the cellular heterogeneity proposed to control tumor phenotypes. Reproducing, measuring and controlling TME heterogeneity should stimulate collaborative efforts between biologists and engineers. Studying cancers in well-tuned 3D cell culture platforms is paramount to bring mechanomedicine into the realm of oncology.
Collapse
Affiliation(s)
- Apekshya Chhetri
- Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Joseph V Rispoli
- Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Sophie A Lelièvre
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States.,Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
81
|
Fu J, Warmflash A, Lutolf MP. Stem-cell-based embryo models for fundamental research and translation. NATURE MATERIALS 2021; 20:132-144. [PMID: 33199861 PMCID: PMC7855549 DOI: 10.1038/s41563-020-00829-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/14/2020] [Indexed: 06/01/2023]
Abstract
Despite its importance, understanding the early phases of human development has been limited by availability of human samples. The recent emergence of stem-cell-derived embryo models, a new field aiming to use stem cells to construct in vitro models to recapitulate snapshots of the development of the mammalian conceptus, opens up exciting opportunities to promote fundamental understanding of human development and advance reproductive and regenerative medicine. This Review provides a summary of the current knowledge of early mammalian development, using mouse and human conceptuses as models, and emphasizes their similarities and critical differences. We then highlight existing embryo models that mimic different aspects of mouse and human development. We further discuss bioengineering tools used for controlling multicellular interactions and self-organization critical for the development of these models. We conclude with a discussion of the important next steps and exciting future opportunities of stem-cell-derived embryo models for fundamental discovery and translation.
Collapse
Affiliation(s)
- Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, TX, USA.
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
- Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
82
|
Gao J, Yang C, Li J, Liu S, Ao Z, Han D. Interfacial Curvature as a Potential Index for Prognosis of Colon Adenocarcinoma. Adv Biol (Weinh) 2021; 5:e1900277. [PMID: 33729697 DOI: 10.1002/adbi.201900277] [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: 11/23/2019] [Revised: 12/14/2020] [Indexed: 11/07/2022]
Abstract
Tumor invasion and metastasis are complex interfacial mechanical processes between the tumor and its surrounding tissue, with the interfacial curvature of tumor playing an important role in cancer progression. In this study, the potential role of interfacial curvature in the prognosis of patients with colon adenocarcinoma is investigated. The front edge interfacial curvature of adenocarcinoma from biopsies of patients in different tumor, lymph node, and metastasis (TNM) stages are calculated and compared, and prognosis assessment is conducted using Kaplan-Meier and Cox proportional hazards regression analyses. Results reveal that patients with larger interfacial curvature of adenocarcinoma are more likely to belong to higher TNM stages. Concomitantly, in the same TNM stage, patients with increased adenocarcinoma interfacial curvature show worse prognosis with higher recurrence and lower survival rates. Besides, interfacial curvature is an independent prognostic factor for cause-specific survival and relapse-free survival among all selected patients. Mechanical models of colon adenocarcinoma invasion and metastasis are established to better understand the close association between interfacial curvature and tumor progression. The results together with hematoxylin and eosin staining indicate that metastasis in stages T3N0M0 and T3N1M0 may be linked to large interfacial curvatures. Therefore, interfacial curvature may serve as a potential index for predicting prognosis in patients with colon adenocarcinoma.
Collapse
Affiliation(s)
- Jingwei Gao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chongqing Yang
- Pathology Department, Beijing Hospital, Beijing, 100730, P. R. China
| | - Jianjun Li
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Sidi Liu
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhuo Ao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Dong Han
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
83
|
Saini H, Nikkhah M. Fabrication Method of a High-Density Co-Culture Tumor-Stroma Platform to Study Cancer Progression. Methods Mol Biol 2021; 2258:241-255. [PMID: 33340365 DOI: 10.1007/978-1-0716-1174-6_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer has now been established as one of the most common chronic diseases due to high mortality rate. The early stage of non-invasive tumors can now be successfully treated leading to have high survival rates; however, the late stage invasive and metastatic tumors still suffer from poor treatment outcomes. Among multiple contributing factors, the role of tumor microenvironment and its complexities has been well recognized in cancer progression. Stromal cells including cancer-associated fibroblasts (CAFs), endothelial cells, adipocytes, immune cells as well as extracellular matrix (ECM) continuously interact with malignant cells and regulate various hallmarks of cancer including tumor growth, invasion, and intravasation. To better understand the role of the interaction between tumor cells and their surrounding microenvironment, numerous model systems ranging from two-dimensional (2D) assays to 3D hydrogels and in vivo murine xenografts have been utilized. While each one of these model systems exhibit certain advantages in studying biological facets of tumor progression, they are often limited to perform well-controlled mechanistic studies due to various factors including lack of tumor-stroma organotypic organization and presence of confounding biochemical and biophysical factors within the tumor microenvironment. In this regard, in the past few years, 3D in vitro microengineered model systems are becoming instrumental to precisely mimic the complexities of the native tumor microenvironment to conduct fundamental and well-designed studies for multiple purposes ranging from biological discovery to therapeutic screening. These model systems include microfluidics, micro-patterned features, and 3D organoids. In this chapter, we will outline the fabrication strategy of our microengineered 3D co-culture tumor-stromal model which comprises high-density array of tumor seeded microwells surrounded by stromal cells, such as CAFs encapsulated within collagen-based hydrogel. The developed platform provides excellent spatial organization of tumor and stromal entities with designated initial architecture and cellular positioning, therefore enabling to study the specific role of cell-cell and cell-ECM interaction on tumor proliferation/expansion, cancer cell migration as well as stromal activation. The developed platform is compatible with standard biological assays enabling gene and protein expression analyses across different types of cancer and co-culture of tumor and stromal cells.
Collapse
Affiliation(s)
- Harpinder Saini
- Harrington Department of Bioengineering, School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, USA
| | - Mehdi Nikkhah
- Harrington Department of Bioengineering, School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, USA.
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
84
|
Abstract
Organoids are in vitro miniaturized and simplified model systems of organs that have gained enormous interest for modelling tissue development and disease, and for personalized medicine, drug screening and cell therapy. Despite considerable success in culturing physiologically relevant organoids, challenges remain to achieve real-life applications. In particular, the high variability of self-organizing growth and restricted experimental and analytical access hamper the translatability of organoid systems. In this Review, we argue that many limitations of traditional organoid culture can be addressed by engineering approaches at all levels of organoid systems. We investigate cell surface and genetic engineering approaches, and discuss stem cell niche engineering based on the design of matrices that allow spatiotemporal control of organoid growth and shape-guided morphogenesis. We examine how microfluidic approaches and lessons learnt from organs-on-a-chip enable the integration of mechano-physiological parameters and increase accessibility of organoids to improve functional readouts. Applying engineering principles to organoids increases reproducibility and provides experimental control, which will, ultimately, be required to enable clinical translation.
Collapse
Affiliation(s)
- Moritz Hofer
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthias P. Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
85
|
Muncie JM, Ayad NME, Lakins JN, Xue X, Fu J, Weaver VM. Mechanical Tension Promotes Formation of Gastrulation-like Nodes and Patterns Mesoderm Specification in Human Embryonic Stem Cells. Dev Cell 2020; 55:679-694.e11. [PMID: 33207224 PMCID: PMC7755684 DOI: 10.1016/j.devcel.2020.10.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/20/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022]
Abstract
Embryogenesis is directed by morphogens that induce differentiation within a defined tissue geometry. Tissue organization is mediated by cell-cell and cell-extracellular matrix (ECM) adhesions and is modulated by cell tension and tissue-level forces. Whether cell tension regulates development by modifying morphogen signaling is less clear. Human embryonic stem cells (hESCs) exhibit an intrinsic capacity for self-organization, which motivates their use as a tractable model of early human embryogenesis. We engineered patterned substrates that recapitulate the biophysical properties of the early embryo and mediate the self-organization of "gastrulation-like" nodes in cultured hESCs. Tissue geometries that generated local nodes of high cell-adhesion tension directed the spatial patterning of the BMP4-dependent "gastrulation-like" phenotype by enhancing phosphorylation and junctional release of β-catenin to promote Wnt signaling and mesoderm specification. Furthermore, direct force application via mechanical stretching promoted BMP-dependent mesoderm specification, confirming that tissue-level forces can directly regulate cell fate specification in early human development.
Collapse
Affiliation(s)
- Jonathon M Muncie
- Graduate Program in Bioengineering, University of California, San Francisco and University of California Berkeley, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nadia M E Ayad
- Graduate Program in Bioengineering, University of California, San Francisco and University of California Berkeley, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Johnathon N Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
86
|
Tran R, Hoesli CA, Moraes C. Accessible dynamic micropatterns in monolayer cultures via modified desktop xurography. Biofabrication 2020; 13. [PMID: 33238251 DOI: 10.1088/1758-5090/abce0b] [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: 08/11/2020] [Accepted: 11/25/2020] [Indexed: 11/12/2022]
Abstract
Micropatterned cell cultures provide an important tool to understand dynamic biological processes, but often require specialized equipment and expertise. Here we present subtractive bioscribing (SuBscribe), a readily accessible and inexpensive technique to generate dynamic micropatterns in biomaterial monolayers on-the-fly. We first describe our modifications to a commercially available desktop xurographer and demonstrate the utility and limits of this system in creating micropatterned cultures by mechanically scribing patterns into a brittle, non-adhesive biomaterial layer. Patterns are sufficiently small to influence cell morphology and orientation and can be extended to pattern large areas with complex reproducible shapes. We also demonstrate the use of this system as a dynamic patterning tool for cocultures. Finally, we use this technique to explore and improve upon the well-established epithelial scratch assay, and demonstrate that robotic control of the scratching tool can be used to create custom-shaped wounds in epithelial monolayers, and that the scribing direction leaves trace remnants of matrix molecules that may significantly affect conventional implementations of this common assay.
Collapse
Affiliation(s)
- Raymond Tran
- Department of Chemical Engineering, McGill University, 3610 University Street, Montreal, Quebec, H4X1N3, CANADA
| | - Corinne Annette Hoesli
- Department of Chemical Engineering, McGill University, 3610 University Street, Montreal, Quebec, H4X 1N3, CANADA
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University, 3610 University Street, Rm 3A, Montreal, Quebec, H4X1N3, CANADA
| |
Collapse
|
87
|
Hayward KL, Kouthouridis S, Zhang B. Organ-on-a-Chip Systems for Modeling Pathological Tissue Morphogenesis Associated with Fibrosis and Cancer. ACS Biomater Sci Eng 2020; 7:2900-2925. [PMID: 34275294 DOI: 10.1021/acsbiomaterials.0c01089] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tissue building does not occur exclusively during development. Even after a whole body is built from a single cell, tissue building can occur to repair and regenerate tissues of the adult body. This confers resilience and enhanced survival to multicellular organisms. However, this resiliency comes at a cost, as the potential for misdirected tissue building creates vulnerability to organ deformation and dysfunction-the hallmarks of disease. Pathological tissue morphogenesis is associated with fibrosis and cancer, which are the leading causes of morbidity and mortality worldwide. Despite being the priority of research for decades, scientific understanding of these diseases is limited and existing therapies underdeliver the desired benefits to patient outcomes. This can largely be attributed to the use of two-dimensional cell culture and animal models that insufficiently recapitulate human disease. Through the synergistic union of biological principles and engineering technology, organ-on-a-chip systems represent a powerful new approach to modeling pathological tissue morphogenesis, one with the potential to yield better insights into disease mechanisms and improved therapies that offer better patient outcomes. This Review will discuss organ-on-a-chip systems that model pathological tissue morphogenesis associated with (1) fibrosis in the context of injury-induced tissue repair and aging and (2) cancer.
Collapse
Affiliation(s)
- Kristen L Hayward
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Sonya Kouthouridis
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
88
|
Micek HM, Visetsouk MR, Masters KS, Kreeger PK. Engineering the Extracellular Matrix to Model the Evolving Tumor Microenvironment. iScience 2020; 23:101742. [PMID: 33225247 PMCID: PMC7666341 DOI: 10.1016/j.isci.2020.101742] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical evidence supports a role for the extracellular matrix (ECM) in cancer risk and prognosis across multiple tumor types, and numerous studies have demonstrated that individual ECM components impact key hallmarks of tumor progression (e.g., proliferation, migration, angiogenesis). However, the ECM is a complex network of fibrillar proteins, glycoproteins, and proteoglycans that undergoes dramatic changes in composition and organization during tumor development. In this review, we will highlight how engineering approaches can be used to examine the impact of changes in tissue architecture, ECM composition (i.e., identity and levels of individual ECM components), and cellular- and tissue-level mechanics on tumor progression. In addition, we will discuss recently developed methods to model the ECM that have not yet been applied to the study of cancer.
Collapse
Affiliation(s)
- Hannah M. Micek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mike R. Visetsouk
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kristyn S. Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Pamela K. Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
89
|
Tian M, Qi Y, Zhang X, Wu Z, Chen J, Chen F, Guan W, Zhang S. Regulation of the JAK2-STAT5 Pathway by Signaling Molecules in the Mammary Gland. Front Cell Dev Biol 2020; 8:604896. [PMID: 33282878 PMCID: PMC7705115 DOI: 10.3389/fcell.2020.604896] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022] Open
Abstract
Janus kinase 2 (JAK2) and signal transducers and activators of transcription 5 (STAT5) are involved in the proliferation, differentiation, and survival of mammary gland epithelial cells. Dysregulation of JAK2-STAT5 activity invariably leads to mammary gland developmental defects and/or diseases, including breast cancer. Proper functioning of the JAK2-STAT5 signaling pathway relies on crosstalk with other signaling pathways (synergistically or antagonistically), which leads to normal biological performance. This review highlights recent progress regarding the critical components of the JAK2-STAT5 pathway and its crosstalk with G-protein coupled receptor (GPCR) signaling, PI3K-Akt signaling, growth factors, inflammatory cytokines, hormone receptors, and cell adhesion.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yingao Qi
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoli Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhihui Wu
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| |
Collapse
|
90
|
Ort C, Lee W, Kalashnikov N, Moraes C. Disentangling the fibrous microenvironment: designer culture models for improved drug discovery. Expert Opin Drug Discov 2020; 16:159-171. [PMID: 32988224 DOI: 10.1080/17460441.2020.1822815] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Standard high-throughput screening (HTS) assays rarely identify clinically viable 'hits', likely because cells do not experience physiologically realistic culture conditions. The biophysical nature of the extracellular matrix has emerged as a critical driver of cell function and response and recreating these factors could be critically important in streamlining the drug discovery pipeline. AREAS COVERED The authors review recent design strategies to understand and manipulate biophysical features of three-dimensional fibrous tissues. The effects of architectural parameters of the extracellular matrix and their resulting mechanical behaviors are deconstructed; and their individual and combined impact on cell behavior is examined. The authors then illustrate the potential impact of these physical features on designing next-generation platforms to identify drugs effective against breast cancer. EXPERT OPINION Progression toward increased culture complexity must be balanced against the demanding technical requirements for high-throughput screening; and strategies to identify the minimal set of microenvironmental parameters needed to recreate disease-relevant responses must be specifically tailored to the disease stage and organ system being studied. Although challenging, this can be achieved through integrative and multidisciplinary technologies that span microfabrication, cell biology, and tissue engineering.
Collapse
Affiliation(s)
- Carley Ort
- Department of Chemical Engineering, McGill University , Montreal, Canada
| | - Wontae Lee
- Department of Chemical Engineering, McGill University , Montreal, Canada
| | - Nikita Kalashnikov
- Department of Chemical Engineering, McGill University , Montreal, Canada
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University , Montreal, Canada.,Department of Biomedical Engineering, McGill University , Montreal, Canada.,Rosalind & Morris Goodman Cancer Research Center, McGill University , Montreal, Canada
| |
Collapse
|
91
|
Yu SM, Li B, Amblard F, Granick S, Cho YK. Adaptive architecture and mechanoresponse of epithelial cells on a torus. Biomaterials 2020; 265:120420. [PMID: 33007611 DOI: 10.1016/j.biomaterials.2020.120420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/20/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022]
Abstract
Curvature is a geometric feature widely observed in the epithelia and critical to the performance of fundamental biological functions. Understanding curvature-related biophysical phenomena remains challenging partly owing to the difficulty of quantitatively tuning and measuring curvatures of interfacing individual cells. In this study, we prepared confluent wild-type Madin-Darby canine kidney cells on a torus structure presenting positive, zero, and negative Gaussian curvatures with a tubule diameter of 2-7 cells and quantified the mechanobiological characteristics of individual cells. Cells on the torus surface exhibited topological sensing ability both as an individual cell and collective cell organization. Both cell bodies and nuclei, adapted on the torus, exhibited local Gaussian curvature-dependent preferential orientation. The cells on the torus demonstrated significant adjustment in the nuclear area and exhibited asymmetric nuclear position depending on the local Gaussian curvature. Moreover, cells on top of the torus, where local Gaussian curvature is near zero, exhibited more sensitive morphological adaptations than the nuclei depending on the Gaussian curvature gradient. Furthermore, the spatial heterogeneity of intermediate filament proteins related to mechanoresponsive expression of the cell body and nucleus, vimentin, keratin and lamin A, revealed local Gaussian curvature as a key factor of cellular adaptation on curved surfaces.
Collapse
Affiliation(s)
- S-M Yu
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - B Li
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - F Amblard
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - S Granick
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Y-K Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
92
|
The not-so-sweet side of sugar: Influence of the microenvironment on the processes that unleash cancer. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165960. [PMID: 32919034 DOI: 10.1016/j.bbadis.2020.165960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022]
Abstract
The role of "aerobic glycolysis" in cancer has been examined often in the past. Results from those studies, most of which were performed on two dimensional conditions (2D, tissue culture plastic), demonstrate that aerobic glycolysis occurs as a consequence of oncogenic events. These oncogenic events often drive malignant cell growth and survival. Although 2D based experiments are useful in elucidating the molecular mechanisms of oncogenesis, they fail to take contributions of the extracellular microenvironment into account. Indeed we, and others, have shown that the cellular microenvironment is essential in regulating processes that induce and/or suppress the malignant phenotype/properties. This regulation between the cell and its microenvironment is both dynamic and reciprocal and involves the integration of cellular signaling networks in the right context. Therefore, given our previous demonstration of the effect of the microenvironment including tissue architecture and media composition on gene expression and the integration of signaling events observed in three-dimension (3D), we hypothesized that glucose uptake and metabolism must also be essential components of the tissue's signal "integration plan" - that is, if uptake and metabolism of glucose were hyperactivated, the canonical oncogenic pathways should also be similarly activated. This hypothesis, if proven true, suggests that direct inhibition of glucose metabolism in cancer cells should either suppress or revert the malignant phenotype in 3D. Here, we review the up-to-date progress that has been made towards understanding the role that glucose metabolism plays in oncogenesis and re-establishing basally polarized acini in malignant human breast cells.
Collapse
|
93
|
Zhang Y, Tang C, Span PN, Rowan AE, Aalders TW, Schalken JA, Adema GJ, Kouwer PHJ, Zegers MMP, Ansems M. Polyisocyanide Hydrogels as a Tunable Platform for Mammary Gland Organoid Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001797. [PMID: 32999851 PMCID: PMC7509700 DOI: 10.1002/advs.202001797] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Indexed: 05/20/2023]
Abstract
In the last decade, organoid technology has developed as a primary research tool in basic biological and clinical research. The reliance on poorly defined animal-derived extracellular matrix, however, severely limits its application in regenerative and translational medicine. Here, a well-defined, synthetic biomimetic matrix based on polyisocyanide (PIC) hydrogels that support efficient and reproducible formation of mammary gland organoids (MGOs) in vitro is presented. Only decorated with the adhesive peptide RGD for cell binding, PIC hydrogels allow MGO formation from mammary fragments or from purified single mammary epithelial cells. The cystic organoids maintain their capacity to branch for over two months, which is a fundamental and complex feature during mammary gland development. It is found that small variations in the 3D matrix give rise to large changes in the MGO: the ratio of the main cell types in the MGO is controlled by the cell-gel interactions via the cell binding peptide density, whereas gel stiffness controls colony formation efficiency, which is indicative of the progenitor density. Simple hydrogel modifications will allow for future introduction and customization of new biophysical and biochemical parameters, making the PIC platform an ideal matrix for in depth studies into organ development and for application in disease models.
Collapse
Affiliation(s)
- Ying Zhang
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 135NijmegenAJ 6525The Netherlands
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Chunling Tang
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Paul N. Span
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Alan E. Rowan
- Australian Institute for Bioengineering and Nanotechnology (AIBN)The University of QueenslandBrisbaneQLD4072Australia
| | - Tilly W. Aalders
- Experimental UrologyRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Jack A. Schalken
- Experimental UrologyRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Gosse J. Adema
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| | - Paul H. J. Kouwer
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 135NijmegenAJ 6525The Netherlands
| | - Mirjam M. P. Zegers
- Department of Cell BiologyRadboud Institute for Molecular SciencesRadboud University Medical CenterGeert Grooteplein 28NijmegenGA6525The Netherlands
| | - Marleen Ansems
- Radiotherapy & OncoImmunology LaboratoryRadboud University Medical CenterGeert Grooteplein 32NijmegenGA6525The Netherlands
| |
Collapse
|
94
|
Pasturel A, Strale P, Studer V. Tailoring Common Hydrogels into 3D Cell Culture Templates. Adv Healthc Mater 2020; 9:e2000519. [PMID: 32743980 DOI: 10.1002/adhm.202000519] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/26/2020] [Indexed: 12/13/2022]
Abstract
Physiologically relevant cell-based models require engineered microenvironments which recapitulate the topographical, biochemical, and mechanical properties encountered in vivo. In this context, hydrogels are the materials of choice. Here a light-based toolbox is able to craft such microniches out of common place materials. Extensive use of benzophenone photoinitiators and their interaction with oxygen achieves this. First, the oxygen inhibition of radicals is harnessed to photoprint hydrogel topographies. Then the chemical properties of benzophenone are exploited to crosslink and functionalize native hydrogels lacking photosensitive moieties. At last, photoscission is introduced: an oxygen-driven, benzophenone-enabled reaction that photoliquefies Matrigel and other common gels. Using these tools, soft hydrogel templates are tailored for cells to grow or self-organize into standardized structures. The described workflow emerges as an effective microniche manufacturing toolset for 3D cell culture.
Collapse
Affiliation(s)
- Aurélien Pasturel
- Interdisciplinary Institute for Neuroscience University of Bordeaux CNRS UMR 5297 Bordeaux F‐33000 France
- Alvéole 30 rue de Campo Formio Paris F‐75013 France
| | | | - Vincent Studer
- Interdisciplinary Institute for Neuroscience University of Bordeaux CNRS UMR 5297 Bordeaux F‐33000 France
| |
Collapse
|
95
|
Steinway SN, Saleh J, Koo BK, Delacour D, Kim DH. Human Microphysiological Models of Intestinal Tissue and Gut Microbiome. Front Bioeng Biotechnol 2020; 8:725. [PMID: 32850690 PMCID: PMC7411353 DOI: 10.3389/fbioe.2020.00725] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract is a complex system responsible for nutrient absorption, digestion, secretion, and elimination of waste products that also hosts immune surveillance, the intestinal microbiome, and interfaces with the nervous system. Traditional in vitro systems cannot harness the architectural and functional complexity of the GI tract. Recent advances in organoid engineering, microfluidic organs-on-a-chip technology, and microfabrication allows us to create better in vitro models of human organs/tissues. These micro-physiological systems could integrate the numerous cell types involved in GI development and physiology, including intestinal epithelium, endothelium (vascular), nerve cells, immune cells, and their interplay/cooperativity with the microbiome. In this review, we report recent progress in developing micro-physiological models of the GI systems. We also discuss how these models could be used to study normal intestinal physiology such as nutrient absorption, digestion, and secretion as well as GI infection, inflammation, cancer, and metabolism.
Collapse
Affiliation(s)
- Steven N. Steinway
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jad Saleh
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology, Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Delphine Delacour
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Deok-Ho Kim
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
96
|
Rens EG, Zeegers MT, Rabbers I, Szabó A, Merks RMH. Autocrine inhibition of cell motility can drive epithelial branching morphogenesis in the absence of growth. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190386. [PMID: 32713299 DOI: 10.1098/rstb.2019.0386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epithelial branching morphogenesis drives the development of organs such as the lung, salivary gland, kidney and the mammary gland. It involves cell proliferation, cell differentiation and cell migration. An elaborate network of chemical and mechanical signals between the epithelium and the surrounding mesenchymal tissues regulates the formation and growth of branching organs. Surprisingly, when cultured in isolation from mesenchymal tissues, many epithelial tissues retain the ability to exhibit branching morphogenesis even in the absence of proliferation. In this work, we propose a simple, experimentally plausible mechanism that can drive branching morphogenesis in the absence of proliferation and cross-talk with the surrounding mesenchymal tissue. The assumptions of our mathematical model derive from in vitro observations of the behaviour of mammary epithelial cells. These data show that autocrine secretion of the growth factor TGF[Formula: see text]1 inhibits the formation of cell protrusions, leading to curvature-dependent inhibition of sprouting. Our hybrid cellular Potts and partial-differential equation model correctly reproduces the experimentally observed tissue-geometry-dependent determination of the sites of branching, and it suffices for the formation of self-avoiding branching structures in the absence and also in the presence of cell proliferation. This article is part of the theme issue 'Multi-scale analysis and modelling of collective migration in biological systems'.
Collapse
Affiliation(s)
- Elisabeth G Rens
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Mathé T Zeegers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Iraes Rabbers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - András Szabó
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Roeland M H Merks
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|
97
|
Abstract
Organoids form through self-organization processes in which initially homogeneous populations of stem cells spontaneously break symmetry and undergo in-vivo-like pattern formation and morphogenesis, though the processes controlling this are poorly characterized. While these in vitro self-organized tissues far exceed the microscopic and functional complexity obtained by current tissue engineering technologies, they are non-physiological in shape and size and have limited function and lifespan. Here, we discuss how engineering efforts for guiding stem-cell-based development at multiple stages can form the basis for the assembly of highly complex and rationally designed self-organizing multicellular systems with increased robustness and physiological relevance.
Collapse
|
98
|
Mechanical Adaptations of Epithelial Cells on Various Protruded Convex Geometries. Cells 2020; 9:cells9061434. [PMID: 32527037 PMCID: PMC7349491 DOI: 10.3390/cells9061434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
The shape of epithelial tissue supports physiological functions of organs such as intestinal villi and corneal epithelium. Despite the mounting evidence showing the importance of geometry in tissue microenvironments, the current understanding on how it affects biophysical behaviors of cells is still elusive. Here, we cultured cells on various protruded convex structure such as triangle, square, and circle shape fabricated using two-photon laser lithography and quantitatively analyzed individual cells. Morphological data indicates that epithelial cells can sense the sharpness of the corner by showing the characteristic cell alignments, which was caused by actin contractility. Cell area was mainly influenced by surface convexity, and Rho-activation increased cell area on circle shape. Moreover, we found that intermediate filaments, vimentin, and cytokeratin 8/18, play important roles in growth and adaptation of epithelial cells by enhancing expression level on convex structure depending on the shape. In addition, microtubule building blocks, α-tubulin, was also responded on geometric structure, which indicates that intermediate filaments and microtubule can cooperatively secure mechanical stability of epithelial cells on convex surface. Altogether, the current study will expand our understanding of mechanical adaptations of cells on out-of-plane geometry.
Collapse
|
99
|
Abstract
PURPOSE OF REVIEW Engineering functional organs starting from stem or progenitor cells holds promise to address the urgent need for organ transplants. However, to date, the development of complex organ structures remains an open challenge. RECENT FINDINGS Among multiple approaches to organ regeneration that are being investigated, two main directions can be identified, namely the patterned deposition of cells to impose specific structures, using bioprinting technologies, and (ii) the spontaneous development of organoids, according to principles of self-organization. In this review, we shortly describe the advantages and limitations of these paradigms and we discuss how they can synergize their positive features to better control and robustly develop organs from stem cells, toward organogenesis by design. SUMMARY The outlined possibilities to bring together tools and concepts of bioprinting and self-organization will be relevant not only to generate implantable organs, but also to dissect fundamental mechanisms of organogenesis and to test therapeutic strategies in modeled pathological settings.
Collapse
|
100
|
Morgan MM, Schuler LA, Ciciliano JC, Johnson BP, Alarid ET, Beebe DJ. Modeling chemical effects on breast cancer: the importance of the microenvironment in vitro. Integr Biol (Camb) 2020; 12:21-33. [PMID: 32118264 PMCID: PMC7060306 DOI: 10.1093/intbio/zyaa002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/18/2019] [Accepted: 02/01/2020] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that our ability to predict chemical effects on breast cancer is limited by a lack of physiologically relevant in vitro models; the typical in vitro breast cancer model consists of the cancer cell and excludes the mammary microenvironment. As the effects of the microenvironment on cancer cell behavior becomes more understood, researchers have called for the integration of the microenvironment into in vitro chemical testing systems. However, given the complexity of the microenvironment and the variety of platforms to choose from, identifying the essential parameters to include in a chemical testing platform is challenging. This review discusses the need for more complex in vitro breast cancer models and outlines different approaches used to model breast cancer in vitro. We provide examples of the microenvironment modulating breast cancer cell responses to chemicals and discuss strategies to help pinpoint what components should be included in a model.
Collapse
Affiliation(s)
- Molly M Morgan
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jordan C Ciciliano
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Elaine T Alarid
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|