CHAPTER
Cell sources and methods for producing organotypic in vitro human tissue models
2 Patrick J. Hayden
MatTek Corporation, Ashland, MA, United States
Introduction The goal of organ-on-a-chip (OoC) technology is to reproduce key human organ systems using miniaturized in vitro cultures that are equivalent to at least the smallest functional unit of each organ (Dehne et al., 2017; Ronaldson-Bouchard and and Vunjak-Novakovic, 2018; Huh et al., 2011). The organ equivalents incorporated into the chips do not necessarily resemble their in vivo counterparts in a visual sense, but will reproduce the essential functions of the organ, and will ideally also incorporate any relevant physical/mechanical features [e.g., threedimensional (3D) extracellular environment/architecture, stretching, contraction, fluid flow, and shear forces] that contribute to organotypic differentiation and organ functions such as breathing, cardiac beating, and blood flow. OoC platforms should also incorporate sensors or features that allow measurement of relevant functional parameters (e.g., compatibility with imaging devices, sensors for measuring real-time conditions, and ports for removal of media/tissues for downstream analysis). A wide variety of OoC platforms have been developed, ranging from systems that incorporate multiple repetitions of a single organ for highthroughput screening applications to systems that incorporate several interacting organs (Fig. 2.1). The ultimate vision for OoC platforms is to create a human-ona-chip device that will replicate all key organ systems and physiologically relevant interactions of the human body (Fig. 2.2). OoC technologies are expected to facilitate the transition from animal-based models to human-based models, provide faster and more predictive human toxicity assessments, and lead to faster development of effective human therapeutics. A requisite for development and widespread use of human OoC technologies is reliable access to human tissues and cells. This chapter provides a detailed survey of human tissue/cell sources and isolation methods, including methods that use induced pluripotent stem cells (iPSCs) currently available to researchers. State-of-the-art culturing devices and techniques that can be applied to OoC Organ on a Chip. DOI: https://doi.org/10.1016/B978-0-12-817202-5.00002-4 © 2020 Elsevier Inc. All rights reserved.
13
FIGURE 2.1 (A) OrganoPlate 3-lane microfluidic tissue culture device containing 40 independent microfluidic culture chips. (B) TissUse Multi-Organ-Chip platform containing 4 interacting organ models. (A) (Courtesy MIMETAS); (B) Reproduced with permission from Maschmeyer I., Lorenz A.K., Schimek K., Hasenberg T., Ramme A.P., Hu¨bner J., et al., A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents, Lab Chip 15(12), 2015a, 2688 2699. https://doi. org/10.1039/c5lc00392j.
Human tissue/cell sources and isolation methods
FIGURE 2.2 Human-on-a-chip concept: in vitro platform reproducing all key organ tissues and physiological interactions. (Courtesy TissUse, Gmbh)
systems—such as the use of nanopatterned substrates, tunable elastic substrates, air liquid interface cultures, spheroid/organoid culture techniques, and 3D bioprinting—are also described.
Human tissue/cell sources and isolation methods Continuous (immortal) cell lines Human somatic cells are in general capable of only a limited number (40 60) of cell divisions in culture before they become senescent and lose their ability to divide (Hayflick and Moorhead, 1961). However, immortal somatic cell lines with a capacity for unlimited division potential can be obtained by a number of processes. Genetically mutated cells derived from cancerous tissues are a common source for establishing immortal cell lines. In rare cases, cultured normal (noncancer-derived) cells may spontaneously acquire genetic mutations that provide the ability for unlimited growth. Normal cells may also be transformed into immortalized cells by the introduction of viral oncogenes such as EBV, SV40LT, HPV16 E6/E7, and Ad5 E1A (Honegger, 2001; Freshney, 2016). Induction of telomerase activity by transduction of human Telomerase reverse transcriptase (hTERT) into cells can induce immortal transformation while retaining more normal cell phenotypes than viral-induced transformations (Freshney, 2016). The development of methods for establishing and maintaining continuous cell lines in culture marked a revolutionary advancement in biology. Since the establishment of HeLa cells as the first immortal human cell line in 1952
15
16
CHAPTER 2 Cell sources and methods
(Gey et al., 1952), continuous cell lines have become widely used as indispensable and inexpensive tools for basic biological research, chemical metabolism and toxicity tests, and production of biological compounds such as vaccines, antibodies, and therapeutic proteins. Numerous immortal cell lines derived from a wide variety of tissues are now readily available. Key advantages of immortal cell lines are that they are affordable, well characterized, and easy to culture. However, immortal cell lines generally exhibit significant genotypic and phenotypic abnormalities that may limit their ability to reproduce normal cell behavior and may undergo additional genotypic or phenotypic drift with continued long-term passaging. Furthermore, many continuous cell lines have been misidentified or have become contaminated with mycoplasma or other cell lines over time (Geraghty et al., 2014; Lorsch et al., 2014). Authentication of cell lines is now recommended or required for publication or submission of research results to regulatory authorities (Geraghty et al., 2014). Despite their shortcomings, immortal cell lines remain in widespread use and will continue to be important biological tools that may be suitable or advantageous for specific OoC applications. Table 2.1 presents a list of commonly used immortal human cell lines derived from a variety of organs. These cell lines, as well as others, are also available as authenticated and quality-controlled resources from a number of nonprofit repositories (Table 2.2).
Primary cell cultures and early-passage cell lines with finite lifespan Cells obtained directly from fresh tissue are commonly termed as primary cells. Advancements in the development of defined culture media, culture conditions, and matrix requirements have led to an increasing ability to culture many types of normal (nonimmortal) primary cells. With the exception of hematopoiesis-derived cells, which may be cultured as cell suspensions, most primary cells require attachment to a substrate to survive and proliferate. Adherence-dependent primary cell cultures may be initiated by explanting small pieces of tissue into a culture plate with the appropriate medium and allowing cells to migrate and proliferate as monolayer cultures. Alternate methods of initiating primary adherent cell cultures involve mechanical and/or enzymatic disaggregation of tissue to form a cell suspension, following by plating the suspension at a low density onto cell culture plates or flasks. Coating the culture plate with various forms of extracellular matrix material or the presence of an established feeder cell layer may be required to support culture establishment when using either the explant or disaggregation methods (Honegger, 2001; Freshney, 2016). Primary cultures obtained by seeding cells or explanted tissue fragments directly after isolation from fresh tissue will consist of those cells that are capable of attachment and survival under the culturing conditions (e.g., culture medium and extracellular matrix coating). The primary culture will consist of a mixed
Table 2.1 Commonly used continuous (immortal) human cell lines. Cell type
Origin
Name
Reference
Endothelial Hepatic
Liver Liver
SK HEP-1 HepaRG
Epithelial Epithelial Epithelial Keratinocyte Type I pneumocyte Type II pneumocyte Type II pneumocyte Epithelial Epithelial Epithelial Epithelial Epithelial Epithelial Epithelial Glial
Liver Breast Breast Epidermis Lung
HepG2 MCF-7 ZR-75-1 HaCaT hAELVi
Heffelfinger et al. (1992) Parent et al. (2004), Guillouzo et al. (2007), and Takahashi et al. (2015) Diamond et al. (1980) Brooks et al. (1973) Engel et al. (1978) Boukamp et al. (1988) Kuehn et al. (2016)
Lung
A549
Giard et al. (1973)
Lung
NCI-H441
Brower et al. (1986)
Lung Lung Kidney Ovary Colon Colon Cervix Glioma
Reddel et al. (1988) Fogh and Trempe (1975) Graham et al. (1977) Tsuruo et al. (1986) Fogh et al. (1977) Fogh and Trempe (1975) Gey et al. (1952) Balmforth et al. (1986)
Glial Lymphocytic Myeloid Myeloid Myeloid Myeloid
Brain Blood Blood Blood Blood Blood
BEAS-2B Calu-3 HEK-293 A2780 Caco-2 HT-29 HeLa MOG-GCCM U-251 MG EB-3 K562 HL-60 THP-1 U937
Ponten and Macintyre (1968) Epstein and Barr (1964) Andersson et al. (1979) Olsson et al. (1981) Tsuchiya et al. (1980) Sundström and Nilsson (1976)
Adapted from Freshney, R.I. (2016). Culture of Animal Cells: A Manual of Basic Technique and Specialized Applications (7th ed.). NJ: Wiley-Blackwell, with additions.
Table 2.2 Nonprofit immortal cell line repositories. Institution
Headquarters
Website
ATCC
Manassas, VA, United States Australia, Sydney, Australia, Salisbury, Wiltshire, United Kingdom
www.atcc.org
CellBank ECACC
The Leibniz Institute DSMZ: German Collection of Microorganisms and Cell Cultures GmbH (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH) JCRB Cell Bank
Braunschweig, Germany
Ibaraki City, Osaka, Japan
www.cellbankaustralia. com www.pheculturecollections.org.uk/ collections/ecacc.aspx www.dsmz.de
www.cellbank.nibiohn. go.jp
ATCC, American Type Culture Collection; ECACC, The European Collection of Authenticated Cell Cultures; JCRB, Japanese Collection of Research Bioresources.
18
CHAPTER 2 Cell sources and methods
population of cells at various stages of differentiation and, for certain tissue types, tissue-specific stem cells with proliferative potential. Cells that are already committed to terminal differentiation may attach and remain viable but will not proliferate further in culture. The proliferation of tissue-specific stem cells will continue until space in the culture vessel becomes limited, at which point the cells may be harvested and passaged into fresh culture vessels. Cells are typically passaged while still in log-phase growth and before reaching confluence, to avoid loss of proliferative capacity owing to contact inhibition that may occur when the cultures become too densely packed. Adherent cells are generally harvested from culture using enzymatic reagents (trypsin or Accutase) or by manual scraping. Primary cells that have been passaged are thereafter termed primary cell lines. The passaged lines will be enriched in cells that have adapted well to the specific culture conditions and those that retain proliferative capacity, while differentiated/nonproliferative cells will die off. With continued culture and successive passaging, the cell lines will at first continue to adapt and become further enriched in cells with proliferative capacity. However, the cells will eventually exhaust their ability to proliferate and become senescent. If the culture conditions are not specifically tuned to promote only the growth of the desired cell type, the cultures may become overgrown with unwanted cell types such as fibroblasts. Conditions that allow in vitro proliferation of many types of primary human cells [e.g., epithelial cells, stromal cells (fibroblasts, stellate cells, pericytes, astrocytes), and endothelial cells] have been successfully developed. However, certain types of cells from key organ systems (e.g., cardiomyocytes, hepatocytes, neurons, islet cells, and monocytes) can only be maintained for a limited time as primary cultures and do not have significant proliferative capacity in vitro. A number of comprehensive texts on the subject of animal and human cell isolation and cell culture techniques are available (Honegger, 2001; Randell and Fulcher, 2013; Picot, 2005; Mitry and Hughes., 2012). Table 2.3 lists protocols in the literature for the isolation and culture of organ-specific human cell types.
Human tissue sources While protocols for isolation of many organ-specific human cell types have been developed, availability and access to fresh human tissue samples may be a significant limitation for researchers outside of clinical research university or hospital settings. Access to fresh human tissues for research requires informed consent of the tissue donor and institutional review board approval (Pirnay et al., 2015). Even for researchers with access to fresh human tissues, cell isolation protocols require specialized techniques that may be difficult to master and are a time-consuming endeavor that may not be feasible or desirable for many laboratories. As an alternative, a number of vendors offer primary human cells (Table 2.4).
Human tissue/cell sources and isolation methods
Table 2.3 Human organ-specific primary cell isolation protocols. Cell type
Reference
Keratinocytes Melanocytes Breast epithelial cells Oral epithelial cells Olfactory neuroepithelial cells Female reproductive tract epithelium Prostate epithelium Coroid plexus Osteoblasts Chondrocytes Myoblasts Fibroblasts Adipocytes
Randell and Fulcher (2013) and Picot (2005) Picot (2005) and Mitry and Hughes (2012) Randell and Fulcher (2013) Randell and Fulcher (2013) Randell and Fulcher (2013)
Mononuclear phagocytes Peripheral blood natural killer cells Neuronal cells Schwann cells Tracheal/bronchial epithelial cells Alveolar epithelial cells Colon epithelial cells Hepatocytes Kupffer cells Glomerular epithelial cells Renal cortical epithelial cells Parathyroid cells Islets of Langerhans cells Corneal and conjunctiva cells Retinal pigment epithelial cells Fetal gastric epithelial cells
Randell and Fulcher (2013) Randell and Fulcher (2013) Randell and Fulcher (2013) Picot (2005) and Mitry and Hughes (2012) Picot (2005) and Mitry and Hughes (2012) Picot (2005) Picot (2005) and Mitry and Hughes (2012) Picot (2005), Mitry and Hughes (2012), Carswell et al. (2012), Brooks et al. (2017), and Ruiz-Ojeda et al. (2016) Picot (2005) Picot (2005) Picot (2005), Gordon et al. (2013), Peng et al. (2013), and Mains and Patterson (1973) Picot (2005) Randell and Fulcher (2013) and Picot (2005) Picot (2005) and Mitry and Hughes (2012) Picot (2005) Randell and Fulcher (2013), Picot (2005), and Mitry and Hughes (2012) Dixon et al. (2013) and Alabraba et al. (2007) Picot (2005) and Mitry and Hughes (2012) Randell and Fulcher (2013), Picot (2005), Mitry and Hughes (2012), Ichimura et al. (2008), and Valente et al. (2011) Picot (2005) and Mitry and Hughes (2012) Picot (2005) and Mitry and Hughes (2012) Randell and Fulcher (2013) and Picot (2005) Randell and Fulcher, 2013 Picot (2005) and Mitry and Hughes (2012) (Continued)
19
20
CHAPTER 2 Cell sources and methods
Table 2.3 Human organ-specific primary cell isolation protocols. Continued Cell type
Reference
Intestinal crypt and villus cells Ovarian cells Vascular smooth muscle cells Endothelial cells Mesenchymal stem cells Peripheral blood mononuclear cells Dendritic cells Regulatory T-cells
Mitry and Hughes (2012), Chopra et al. (2010), Gjorevski et al. (2016), Tong et al. (2018), and Holmberg et al. (2017) Mitry and Hughes (2012) Mitry and Hughes (2012) Mitry and Hughes (2012) Mitry and Hughes (2012) Mitry and Hughes (2012) Fahrbach et al. (2007) Mitry and Hughes (2012)
Table 2.4 Commercial sources of continuous cell lines and primary human cells. Vendor
Headquarters
Website
AllCells ATCC BioIVT Biopredic International Cell Systems, iXCells Biotechnologies
Emeryville, CA, United States Manassas, VA, United States Westbury, NY, United States Saint Grégoire, France Kirkland, WA, United States San Diego, CA, United States Frederick, MD, United States
www.allcells.com www.atcc.org www.bioivt.com www.biopredic.com www.cell-systems.com www.ixcellsbiotech. com www.lifelinecelltech. com www.lonza.com www.mattek.com www.emdmillipore.com
Lifeline Cell Technology Lonza MatTek Corporation MilliporeSigma Novabiosis PromoCell ScienCell Research Laboratories Sekisui XenoTech, LLC Stemcell Technologies Thermo Fisher Scientific Zen-Bio
Basel, Switzerland Ashland, MA, United States Burlington, MA, United States Morrisville, NC, United States Heidelberg, Germany Carlsbad, CA, United States Kansas City, KS, United States Vancouver, BC, Canada Waltham, MA, United States Durham, NC, United States
ATCC, American Type Culture Collection.
www.novabiosis.com www.promocell.com www.sciencellonline. com www.xenotech.com www.stemcell.com www.thermofisher.com www.zen-bio.com
Methods for producing three-dimensional “organotypic” tissue cultures
Induced pluripotent stem cells A seminal advancement in cell culture and regenerative medicine occurred in 2006 with the development of methods for generating iPSCs from differentiated somatic cells via induced expression of four transcription factors (Takahashi et al., 2007a,b; Yu et al., 2007; Sayed et al., 2016). Because adult cells are used, iPSCs avoid the restrictions and controversy surrounding human embryonic stem cells. Human somatic tissues, fluids, and cell types such as fibroblasts, blood cells, and urine have been used to generate iPSCs. The initial iPSC protocols used retroviral and lentiviral systems to integrate transcription factors into the host genome. Recently developed protocols allow the use of nonintegrating systems, including Sendai virus, episomal reprogramming factors, and microRNAs, to generate iPSCs without integrating the reprograming factors into the genome (Fusaki et al., 2009; Warren et al., 2010). Once generated, iPSCs are theoretically capable of differentiation into any cell type; iPSCs are therefore a valuable source for generation of large numbers of cells that normally do not proliferate in vitro (e.g., cardiomyocytes, hepatocytes, and neuronal cells) (McKernan and Watt, 2013). In addition, iPSCs allow researchers to recreate in vitro models of inherited genetic human diseases and enable the derivation of multiple types of organ models from the same donor (“body-on-a-chip” or “you-on-a-chip” devices). Development of protocols to drive differentiation of iPSCs into various tissue-specific lineages is an area of current intense effort. The growing list of organspecific cell types that have been generated to date from iPSCs includes hepatic, cardiac, neuronal, endothelial (including blood brain barrier), pancreatic, lung, renal, and intestinal cells (Table 2.5). iPSC technology currently requires significant expertise and is a time-consuming process. The science is still developing, and current protocols do not yet reproduce fully differentiated organ-specific cell phenotypes. A growing number of iPSC sources currently exist (McKernan and Watt, 2013; De Sousa et al., 2017; Kim et al., 2017; Ntai et al., 2017). The California Institute for Regenerative Medicine (CIRM) hPSC Repository is the world’s largest, containing iPSCs from over 3000 individuals. The CIRM iPSC lines are produced by nonintegrating episomal reprogramming. Demographic and clinical data are available for a variety of diseases or conditions affecting the brain, heart, lung, liver, and eyes. The Coriell Institute for Medical Research (Camden, NJ, United States) offers dozens of iPSC lines related to Parkinson’s disease, amyotrophic lateral sclerosis, and Huntington’s disease, and supplies iPSCs to other repositories. European-based cell banks include the European Bank for Induced Pluripotent Stem Cells (De Sousa et al., 2017), the Human Induced Pluripotent Stem Cell Initiative, and StemBANCC (Table 2.6).
Methods for producing three-dimensional “organotypic” tissue cultures Traditional cell isolation techniques and culture methods for adherence-dependent cells typically involve submersion cultures of cell monolayers on two-dimensional
21
22
CHAPTER 2 Cell sources and methods
Table 2.5 Cell types and disease models developed from induced pluripotent stem cells to date. Tissue type
References
Hepatic
Meier et al. (2017), Yamashita et al. (2018), Matoba et al. (2018), Wang et al. (2017), and Takayama and Mizuguchi (2017) Sallam et al. (2014), Jiang et al. (2016), Kamdar et al. (2015), Tanaka et al. (2015), Talkhabi et al. (2016), Ronaldson-Bouchard et al. (2018), Matsa et al. (2016), and Sharma et al. (2017) Schwartz et al. (2015), Lancaster et al. (2013), Centeno et al. (2018), Lee et al. (2017), Gabriel and Gopalakrishnan (2017), Klaus et al. (2019), Pamies et al. (2017), and Hofrichter et al. (2017) Bao et al. (2015), Wong et al. (2012), Sone and Nakao (2013), Qian et al. (2017), and Stebbins et al. (2016) Kondo et al. (2018a), Loo et al. (2018), Bose and Sudheer (2016), Yabe et al. (2017), and Kim et al. (2016) Wang et al. (2016) and Wilkinson et al. (2018) Sinagoga et al. (2018), Kondo et al. (2018b), Miura and Suzuki (2018), Takahashi et al. (2018), and Rahmani et al. (2019) Takasato and Little (2017) and Schutgens et al. (2019)
Cardiac
Neuronal
Endothelial (including blood brain barrier) Pancreatic Lung Intestinal
Renal
Table 2.6 Repositories and other sources of induced pluripotent stem cells. Institution
Website
American Type Culture Collection Boston University Center for Regenerative Medicine California Institute for Regenerative Medicine Coriell Institute for Medical Research European Bank for Induced Pluripotent Stem Cells Harvard Stem Cell Institute New York Stem Cell Foundation StemBANCC Tempo Bioscience Human Induced Pluripotent Stem Cell Initiative UK Stem Cell Bank US National Institute of Mental Health US National Institute of Neurological Disorders and Stroke
www.atcc.org www.bu.edu/dbin/stemcells/ iPSC_bank.php www.cirm.ca.gov www.coriell.org www.ebisc.org www.hsci.harvard.edu www.nyscf.org www.stembancc.org www.tempobioscience.com www.hipsci.org www.nibsc.org/ukstemcellbank www.nimhgenetics.org www.nindsgenetics.org
Methods for producing three-dimensional “organotypic” tissue cultures
(2D) plastic substrates. These methods were developed to promote proliferation of cells and generally lead to loss of differentiated cellular functions. Moreover, traditional 2D culture environments lack the important cell cell, cell matrix, 3D architecture, and mechanical cues (e.g., stretch, strain, shear forces, nanotopography, and substrate compliance) that are found in the in vivo environment of the cells and that are essential for functional differentiation (Huh et al., 2011; Schmeichel and Bissell, 2003; Alhaque et al., 2018). Recognition of the inherent limitations of 2D culture environments has motivated efforts to develop 3D cell culture conditions that better replicate in vivo tissue architectures and provide more physiologically relevant “organotypic” in vitro tissue models of normal function and disease. A variety of 3D organotypic tissue culture techniques and models have been developed over the past decades. Several of these techniques are used as key building blocks for producing tissues that may be incorporated into OoC platforms described in this book, which add organ connectivity and mechanical features such as medium flow and shear forces. The remainder of this chapter covers current approaches to producing 3D organotypic tissue models. Table 2.7 provides a list of commercial sources of cultureware, devices, and supplies. Table 2.8 lists the current commercial providers of ready-to-use 3D organotypic tissue models.
Use of controlled substrate rigidity and nanopatterned culture substrates to enhance morphological and functional differentiation Functional organotypic differentiation depends on the presence of an appropriately structured microenvironment. The mechanical properties of the matrix, such as elasticity and nanotopography, provide fundamental cues that regulate and guide cell migration, morphology, assembly and alignment, signal transduction pathways, and gene transcription (Kim et al., 2013; Park et al., 2012; Kshitiz et al., 2012). Nanopatterned culture surfaces have been designed to provide a cellular microenvironment that mimics the architecture of the native extracellular matrix. Cells can align, elongate, grow, and migrate along the nanopatterned surfaces, leading to more physiologically representative structural and functional phenotypes. For example, iPSC-derived cardiomyocytes cultured on nanopatterned culture surfaces exhibit polarized expression of gap junction proteins such as Cx43 and develop anisotropic cell shape, striated sarcomeres, and tissue-level alignment, as well as achieving enhanced baseline electrophysiology such as faster longitudinal conduction velocity and lower resting membrane potential (Fig. 2.3; Kim et al., 2010). Nanopatterned culture substrates have also been used to enhance the morphological and functional differentiation of vascular smooth muscle cells (Chaterji et al., 2014), endothelial cells (Jeon et al., 2015), osteocytes (You et al., 2010), and skeletal muscle cells (Smith et al., 2016) in vitro. Traditional tissue culture plasticware is much stiffer than the natural extracellular microenvironments found in tissues and organs. The elastic modulus of
23
24
CHAPTER 2 Cell sources and methods
Table 2.7 Commercial suppliers of culture devices and products for organotypic culture models. Company
Headquarters
Products
Website
Accela, Strašnice Advanced BioMatrix
Czech Republic Carlsbad, CA, United States
www.accela.eu www. advancedbiomatrix. com
Corning
Corning, NY, United States
ExCellness Biotech SA
Lausanne, Switzerland
Greiner Bio-One GmbH
Kremsmünster, Austria
Matrigen Life Technologies MatTek Corporation
Brea, CA, United States Ashland, MA, United States
MilliporeSigma
Burlington, MA, United States
NanoSurface Biomedical
Seattle, WA, United States
Rotary bioreactors Extracellular matrix material, scaffolds, CytoSoft elastic modulus plates, bioinks Cell culture plasticware and reagents, microporous membrane inserts, spheroid-forming plates, attachment plates Elastic substrate cultureware and products Cell culture plasticware and reagents, microporous membrane inserts, spheroid-forming plates Softwell elastic substrate plates Glass-bottomed culture dishes, nanopatterned dishes Cell culture plasticware and reagents, microporous membrane inserts, spheroid-forming plates Nanopatterned dishes
Synthecon
Houston, TX, United States Waltham, MA, United States
ThermoFisherScientific
Rotary bioreactors Cell culture plasticware and reagents, microporous membrane inserts, spheroid-forming plates
www.corning.com/ worldwide/en/ products/lifesciences.html
www.excellness. com www.gbo.com/ en_US.html
www.matrigen. com www.mattek.com
www.emdmillipore. com
www. nanosurfacebio. com www.synthecon. com www.thermofisher. com
Methods for producing three-dimensional “organotypic” tissue cultures
Table 2.8 Commercial suppliers of three-dimensional organotypic culture models. Company
Headquarters
Products
Epithelix
Geneva, Switzerland Schlieren, Switzerland Ashland, MA, United States
Lung epithelial models
InSphero MatTek Corporation Organovo
Phenion Episkin StemoniX
San Diego, CA, United States Düsseldorf, Germany Lyon, France Maple Grove, MN, United States
Liver, pancreas, and cancer spheroid models Skin, lung, corneal, intestinal, vaginal, and oral epithelial models; primary human cells Bioprinted liver and kidney models
Skin models Skin, corneal, and other epithelial models Cardiac and brain spheroid models; two-dimensional brain models
www.epithelix. com www.insphero. com www.mattek. com www.organovo. com www.phenion. com www.episkin. com www.stemonix. com
FIGURE 2.3 Alignment of cardiomyocytes on nanopatterned culture well. (Courtesy NanoSurface Biomedical).
typical cell culture plastic is on the order of 1 3 107 kPa, whereas tissues and organs have much lower values, typically 0.2 64 kPa (Wells, 2008; Charrier et al., 2018). Tissue stiffening has been shown to be a major component of aging and fibrotic diseases (Lampi and Reinhart-King, 2018; Asano et al., 2017;
25
26
CHAPTER 2 Cell sources and methods
Liu et al., 2010). Substrate stiffness has also been found to regulate proliferation and differentiation of stem cells (Engler et al., 2006; Gilbert et al., 2010; Smith et al., 2017) and the growth and migration of cancer cells (Tilghman et al., 2010). Polyacrylamide, polydimethylsiloxane, and silicon substrates can be engineered to match the compliance of the in vivo environments of various organs and disease states (Mih et al., 2011; Smith et al., 2017). Several companies offer cultureware that features physiologically compliant substrates, including Matrigen (www. matrigen.com), Advanced BioMatrix (www.advancedbiomatrix.com), and ExCellness Biotech (www.excellness.com).
Use of microporous membrane substrates to prepare cultures with polarized barrier, transport, and differentiated properties Many epithelial and endothelial tissues exist in vivo as polarized sheets or tubes that are bounded on the basolateral side by a basement membrane and have an external (e.g., skin) or luminal (e.g., airways, intestine, kidney tubules, and blood vessels) side that may be exposed to air or liquid. These types of polarized tissues are commonly modeled in vitro by means of a “raft” culture on metal mesh grids (Anacker and Moody, 2012) or on specialized microporous membrane support wells (Fig. 2.4; Adler et al., 1987; Be´rube´ et al., 2010) to create air liquid interface or liquid liquid interface environments. Tissues cultured under these conditions typically form tight junctions between cells at the apical layers and provide selective barrier function. Morphologically, these cultures may range from flattened or simple cuboidal monolayers to multilayered 3D stratified or pseudostratified epithelial structures. Differentiated organotypic functional
FIGURE 2.4 Schematic illustration of air-liquid interface tissue culture using microporous membrane culture insert. (Courtesy MatTek Corp.)
Methods for producing three-dimensional “organotypic” tissue cultures
FIGURE 2.5 Examples of organotypic air-liquid interface human skin, intestine and airway epithelial tissues. (Courtesy MatTek Corp)
attributes of these models include a stratum corneum for epidermal models (Bell et al., 1981; Asselineau et al., 1985; Cannon et al., 1994), mucus-secreting goblet cells for airway (Bolmarcich et al., 2018) or intestine (Ayehunie et al., 2018), brush border for intestine (Ayehunie et al., 2018), and beating cilia for airway (Be´rube´ et al., 2010). Multiple cell types, such as epithelial, stromal, endothelial, and immune cells, may be incorporated together to produce more complex cocultures with additional in vivo like functionality (Fizes¸an et al., 2019; Marescotti et al., 2019). Fig. 2.5 displays representative examples of epidermal, bronchial, and intestinal epithelial tissue models produced on microporous membrane inserts. These organotypic systems provide isolated apical and basolateral compartments that allow realistic in vivo like drug/environmental exposures and are very useful for studies of drug transport and efficacy (Kaluzhny et al., 2018; Ayehunie et al., 2018), toxicology and safety (Hayden et al., 2015; Gordon et al., 2015; Maschmeyer et al., 2015a,b; Peters et al., 2019; Hoppensack et al., 2014), and viral or bacterial infections (Bai et al., 2015; Maldonado-Contreras et al., 2017). Human diseases such as asthma (Bai et al., 2015; Hackett et al., 2011) and psoriasis (Chamcheu et al., 2015) have also been successfully modeled with these organotypic systems. Recently, membranes fabricated from polydimethylsiloxane were used to add in vivo like mechanical stretching features to air liquid interface systems (Fig. 2.6; Novak et al., 2018; Felder et al., 2019).
Techniques for preparing three-dimensional spheroids/organoids with differentiated organotypic functions A variety of methods have been developed for inducing scaffold-free selfassembly of cells into 3D spheroids or organoids that mimic key structural and functional attributes of the original organ (Alhaque et al., 2018; Sato and
27
28
CHAPTER 2 Cell sources and methods
FIGURE 2.6 (A) Air-liquid interface PDMS Lung-on-Chip epithelial tissue allows dynamic medium and air flow, shear stress and stretching. (B) Air-liquid interface Lung-on-Chip platform containing 12 alveolar epithelial tissues featuring membrane with in vivo-like expansion and contraction. (A) Reproduced with permission from Huh D., Matthews B.D., Mammoto A., Montoya-Zavala M., Hsin H.Y. and Ingber D.E., Reconstituting organ-level lung functions on a chip, Science 328 (5986), 2010, 1662 1668. https://doi.org/10.1126/science.1188302; (B) Courtesy Alveolix.
Clevers, 2015). These methods make use of the hanging drop technique, rotary reactor devices, embedding in hydrogel matrices, or nonadherent plates or molds. Organoid culture techniques have been applied to produce models of the liver (Meier et al., 2017; Lee et al., 2013; Bell et al., 2016; Proctor et al., 2017;
Methods for producing three-dimensional “organotypic” tissue cultures
FIGURE 2.6 (Continued).
Messner et al., 2018), pancreas (Bose and Sudheer, 2016; Kim et al., 2016; Zuellig et al., 2014), brain (Schwartz et al., 2015; Lancaster et al., 2013; Lee et al., 2017; Gabriel and Gopalakrishnan, 2017; Klaus et al., 2019; Pamies et al., 2017; Hofrichter et al., 2017), kidney (Schutgens et al., 2019), heart (Beauchamp
29
30
CHAPTER 2 Cell sources and methods
FIGURE 2.7 A process for producing cerebral organoids from hIPSCs using a spinning bioreactor. Reproduced with permission from Lancaster M.A., Renner M., Martin C.-A., Wenzel D., Bicknell L.S., Hurles M.E., et al., Cerebral organoids model human brain development and microcephaly, Nature 501, 2013, 373 379
et al., 2015; Rismani Yazdi et al., 2015), intestine (Miura and Suzuki, 2018; Takahashi et al., 2018; Rahmani et al., 2019; Sachs et al., 2017; Mu´nera and Wells, 2017), and cancer (Schmeichel and Bissell, 2003; Plummer et al., 2019; Herter et al., 2017). Coculture models of the liver have included hepatocytes, stellate cells, and Kupffer cells and are reported to maintain drug metabolism capabilities long-term and to demonstrate in vivo like inflammatory responses (Lee et al., 2013; Messner et al., 2018). Brain organoids have been constructed with glial cells, pericytes, and astrocytes, and demonstrate extensive neurite outgrowth, electrical connectivity, and myelination (Schwartz et al., 2015; Lancaster et al., 2013; Pamies et al., 2017). The size of organoid structures should be limited to diameters of ,500 nm, the limit of oxygen diffusion, to maintain cell viability and avoid cell necrosis from lack of oxygen. An example of cerebral organoid production from human iPSCs is depicted in Fig. 2.7 (Lancaster et al., 2013).
Three-dimensional bioprinting The application of 3D printing techniques to biological systems has provided unprecedented opportunities to reproduce heterogeneous tissues with a complex 3D architecture (Xia et al., 2018; Kolesky et al., 2018; Vanderburgh et al., 2017). Cells, growth factors, and matrix materials combined together in so-called “bioinks” are deposited layer by layer to produce the 3D structures. Bioprinting has leveraged inkjet, laser-assisted, and extrusion printing techniques; computeraided design files are used to direct the deposition of the cells and biomaterials
Summary/Outlook
with microscale resolution. Bioinks may be synthetic or biologically derived polymers. Common synthetic polymers used in bioink include modified poly(ethylene glycol), poly(lactic acid), poly(lactic-co-glycolic acid), and poly(e-caprolactone). Bioinks prepared from thermosensitive polymers (e.g., Pluronic F127) that can later be liquefied and removed have been used to create vasculature or tubule-like networks within bioprinted tissues (Herter et al., 2017). Biologically derived polymers commonly used in preparing bioinks include collagens, gelatin, hyaluronic acid, fibrin, alginate, and decellularized extracellular matrices (Herter et al., 2017). These polymers are often chemically modified or combined with ceramic, glass, or hydroxyapatite to provide improved mechanical strength and rheological properties. Bioprinting has been achieved and described for both hard tissues such as bone and cartilage (Kolesky et al., 2018; Mu¨ller et al., 2017; Nguyen et al., 2017) and soft tissues such as skin (Xia et al., 2018), liver (Xia et al., 2018; Ma et al., 2016; Norona et al., 2019), kidney (Xia et al., 2018; King et al., 2017; Homan et al., 2016; Homan et al., 2019; Lin et al., 2019), brain (Xia et al., 2018; Espinosa-Hoyos et al., 2018), lung (Xia et al., 2018), heart (Lind et al., 2017), intestine (Madden et al., 2018), and vasculature (Xia et al., 2018; Kolesky et al., 2016; Zhu et al., 2017). Bioprinting processes enable precise placement of multiple cell types and heterologous material components, thereby providing tremendous promise for replicating functional vasculature, tubules, and glands. Remarkable progress has been made in bioprinting, and techniques continue to develop, along with new bioink materials that are both printable and biocompatible. A variety of companies now offer bioprinting devices, bioinks, and related supplies (Table 2.9).
Summary/Outlook The fields of in vitro cell culture and organotypic model development are experiencing an exciting new era brought about by key advancements. Methods for creating immortal cell lines are well established, and methods for isolation and maintenance of primary human cells have been developed for most tissues and organs. A wide variety of immortal and primary human cells are now readily available to the research community. Use of iPSC technology enables largescale generation of differentiated cell types that were not previously available for research because of proliferation limitations. As the methods for culturing of normal human cells and differentiation of organ-specific cells from iPSCs continue to advance, reliance on immortal cell lines will likely decrease. Key advancements in novel culture substrates and scaffold materials, culturing techniques, spheroid/organoid models, and bioprinting are facilitating rapid progress in the development of in vitro models that replicate complex in vivo like structures and multicellular interactions. Major ongoing needs include
31
32
CHAPTER 2 Cell sources and methods
Table 2.9 Commercial suppliers of bioprinter devices, bioinks, and related services and supplies. Company
Headquarters
Website
3D Bioprinting Solutions Advanced Solutions Life Sciences Allevi
Moscow, Russia Louisville, KY, United States Philadelphia, PA, United States Vancouver, BC, Canada
www.bioprinting.ru www.lifesciences. solutions www.allevi3d.com
Aspect Biosystems Cellbricks CELLINK Cyfuse Biomedical K.K. EnvisionTEC nScrypt Poietis regenHU ROKIT Healthcare
Berlin, Germany Gothenburg, Sweden Tokyo, Japan Dearborn, MI, United States Orlando, FL, United States Pessac, France Villaz-Saint-Pierre, Switzerland Seoul, Korea
www.aspectbiosystems. com www.cellbricks.com www.cellink.com www.cyfusebio.com/en www.envisiontec.com www.nscrypt.com www.poietis.com www.regenhu.com www.rokithealthcare.com
improved protocols for differentiating iPSCs to more faithfully recapitulate organ-specific phenotype and functions, and universal culture media (i.e., blood surrogates) that can support multiple cell and organ types. Continued progress in these areas will lead to better organotypic differentiation, more faithful reproduction of in vivo organ function, and physiologically relevant modeling of human disease. OoC platforms and technologies will leverage advancements in cell and organotypic tissue culture to model organ organ interactions by providing organ connectivity, medium flow (artificial blood), and mechanical forces. These combined technologies are expected to greatly advance our understanding of the biology and interactions of human organs and their functions under normal and disease conditions. Organotypic human models in OoC platforms will also enable rapid in vitro assessment of absorption, distribution, metabolism, excretion, and toxicity of chemicals and drugs, thereby facilitating faster and more efficient development of human therapeutics.
References Adler, K.B., Schwartz, J.E., Whitcutt, M.J., Wu, R., 1987. A new chamber system for maintaining differentiated guinea pig respiratory epithelial cells between air and liquid phases. BioTechniques 5, 462 466.
References
Alabraba, E.B., Curbishley, S.M., Lai, W.K., Wigmore, S.J., Adams, D.H., Afford, S.C., 2007. A new approach to isolation and culture of human Kupffer cells. J. Immunol. Methods 326, 139 144. Alhaque, S., Themis, M., Rashidi, H., 2018. Three-dimensional cell culture: from evolution to revolution. Philos. Trans. R. Soc. London, Ser. B: Biol. Sci. 373 (1750), pii: 20170216. Review. Available from: https://doi.org/10.1098/rstb.2017.0216. Anacker, D., Moody, C., 2012. Generation of organotypic raft cultures from primary human keratinocytes. J. Visualized Exp. 22 (60), pii: 3668. Available from: https://doi. org/10.3791/3668. Andersson, L.C., Nilsson, K., Gahmberg, C.G., 1979. K562—a human erythroleukemic cell line. Int. J. Cancer 23 (2), 143 147. Asano, S., Ito, S., Takahashi, K., Furuya, K., Kondo, M., Sokabe, M., et al., 2017. Matrix stiffness regulates migration of human lung fibroblasts. Physiol. Rep. 5 (9), pii: e13281. Available from: https://doi.org/10.14814/phy2.13281. Asselineau, D., Bernhard, B., Bailly, C., Darmon, M., 1985. Epidermal morphogenesis and induction of the 67 kD keratin polypeptide by culture of human keratinocytes at the liquid-air interface. Exp. Cell Res. 159 (2), 536 539. Ayehunie, S., Landry, T., Stevens, Z., Armento, A., Hayden, P., Klausner, M., 2018. Human primary cell-based organotypic microtissues for modeling small intestinal drug absorption. Pharm. Res. 35 (4), 72. Available from: https://doi.org/10.1007/s11095018-2362-0. Bai, J., Smock, S.L., Jackson Jr, G.R., MacIsaac, K.D., Huang, Y., et al., 2015. Phenotypic responses of differentiated asthmatic human airway epithelial cultures to rhinovirus. PLoS One 10 (2), e0118286. Available from: https://doi.org/10.1371/journal. pone.0118286. eCollection 2015. Balmforth, A.J., Ball, S.G., Freshney, R.I., Graham, D.I., McNamee, H.B., Vaughan, P.F., 1986. D-1 dopaminergic and beta-adrenergic stimulation of adenylate cyclase in a clone derived from the human astrocytoma cell line G-CCM. J. Neurochem. 47 (3), 715 719. Bao, X., Lian, X., Dunn, K.K., Shi, M., Han, T., Qian, T., et al., 2015. Chemically-defined albumin-free differentiation of human pluripotent stem cells to endothelial progenitor cells. Stem Cell Res. 15 (1), 122 129. Available from: https://doi.org/10.1016/j. scr.2015.05.004. Beauchamp, P., Moritz, W., Kelm, J.M., Ullrich, N.D., Agarkova, I., Anson, B.D., et al., 2015. Development and characterization of a scaffold-free 3D spheroid model of induced pluripotent stem cell derived human cardiomyocytes. Tissue Eng., C: Methods 21 (8), 852 861. Available from: https://doi.org/10.1089/ten.TEC.2014.0376. Bell, E., Ehrlich, H.P., Buttle, D.J., Nakatsuji, T., 1981. Living tissue formed in vitro and accepted as skin-equivalent tissue of full thickness. Science 211 (4486), 1052 1054. Bell, C.C., Hendriks, D.F., Moro, S.M., Ellis, E., Walsh, J., Renblom, A., et al., 2016. Characterization of primary human hepatocyte spheroids as a model system for druginduced liver injury, liver function and disease. Sci. Rep. 6, 25187. Available from: https://doi.org/10.1038/srep25187. Be´rube´, K., Pitt, A., Hayden, P., Prytherch, Z., Job, C., 2010. Filter-well technology for advanced three-dimensional cell culture: perspectives for respiratory research. Altern. Lab. Anim. 38 (Suppl 1), 49 65. Bolmarcich, J., Wilbert, S., Jackson, G.R., Oldach, J., Bachelor, M., Kenney, T., et al., 2018. In vitro human airway models for study of goblet cell hyperplasia and mucus
33
34
CHAPTER 2 Cell sources and methods
production: effects of Th2 cytokines, double-stranded RNA, and tobacco smoke. Appl. In Vitro Toxicol. 4 (4), 332. Available from: https://doi.org/10.1089/aivt.2017.0001. Bose, B., Sudheer, P.S., 2016. In vitro differentiation of pluripotent stem cells into functional β islets under 2D and 3D culture conditions and in vivo preclinical validation of 3D islets. Methods Mol. Biol. 1341, 257 284. Available from: https://doi.org/10.1007/ 7651_2015_230. Boukamp, P., Petrussevska, R.T., Breitkreutz, D., Hornung, J., Markham, A., Fusenig, N. E., 1988. Normal keratinization in a spontaneously immortalized aneuploid human keratinocyte cell line. J. Cell Biol. 106 (3), 761 771. Brooks, J.C., Judd, R.L., Easley, C.J., 2017. Culture and sampling of primary adipose tissue in practical microfluidic systems. Methods Mol. Biol. 1566, 185 201. Available from: https://doi.org/10.1007/978-1-4939-6820-6_18. Brooks, S.C., Locke, E.R., Soule, H.D., 1973. Estrogen receptor in a human cell line (MCF-7) from breast carcinoma. J. Biol. Chem. 248 (17), 6251 6253. Brower, M., Carney, D.N., Oie, H.K., Gazdar, A.F., Minna, J.D., 1986. Growth of cell lines and clinical specimens of human non-small cell lung cancer in a serum-free defined medium. Cancer Res. 46, 798 806. Cannon, C.L., Neal, P.J., Southee, J.A., Kubilus, J., Klausner, M., 1994. New epidermal model for dermal irritancy testing. Toxicol. In Vitro 8 (4), 889 891. Carswell, K.A., Lee, M.J., Fried, S.K., 2012. Culture of isolated human adipocytes and isolated adipose tissue. Methods Mol. Biol. 806, 203 214. Centeno, E.G.Z., Cimarosti, H., Bithell, A., 2018. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol. Neurodegener. 13 (1), 27. Available from: https://doi.org/10.1186/s13024-018-0258-4. Chamcheu, J.C., Pal, H.C., Siddiqui, I.A., Adhami, V.M., Ayehunie, S., Boylan, B.T., et al., 2015. Prodifferentiation, anti-inflammatory and antiproliferative effects of delphinidin, a dietary anthocyanidin, in a full-thickness three-dimensional reconstituted human skin model of psoriasis. Skin Pharmacol. Physiol. 28 (4), 177 188. Available from: https://doi.org/10.1159/000368445. Charrier, E.E., Pogoda, K., Wells, R.G., Janmey, P.A., 2018. Control of cell morphology and differentiation by substrates with independently tunable elasticity and viscous dissipation. Nat. Commun. 9 (1), 449. Available from: https://doi.org/10.1038/s41467-01802906-9. Chaterji, S., Kim, P., Choe, S.H., Tsui, J.H., Lam, C.H., Ho, D.S., et al., 2014. Synergistic effects of matrix nanotopography and stiffness on vascular smooth muscle cell function. Tissue Eng., A 20 (15 16), 2115 2126. Available from: https://doi.org/10.1089/ ten.tea.2013.0455. Chopra, D.P., Dombkowski, A.A., Stemmer, P.M., Parker, G.C., 2010. Intestinal epithelial cells in vitro. Stem Cells Dev. 19 (1), 131 142. Available from: https://doi.org/ 10.1089/scd.2009.0109. Dehne, E.M., Hasenberg, T., Marx, U., 2017. The ascendance of microphysiological systems to solve the drug testing dilemma. Future Sci. OA 3 (2), FSO185. Available from: https://doi.org/10.4155/fsoa-2017-0002. De Sousa, P.A., Steeg, R., Wachter, E., Bruce, K., King, J., Hoeve, M., et al., 2017. Rapid establishment of the European Bank for induced Pluripotent Stem Cells (EBiSC)—the Hot Start experience. Stem Cell Res. 20, 105 114. Available from: https://doi.org/ 10.1016/j.scr.2017.03.002.
References
Diamond, L., Kruszewski, F., Aden, D.P., Knowles, B.B., Baird, W.M., 1980. Metabolic activation of benzo[a]pyrene by a human hepatoma cell line. Carcinogenesis 1 (10), 871 875. Dixon, L.J., Barnes, M., Tang, H., Pritchard, M.T., Nagy, L.E., 2013. Kupffer cells in the liver. Compr. Physiol. 3 (2), 785 797. Available from: https://doi.org/10.1002/cphy. c120026. Engel, L.W., Young, N.A., Tralka, T.S., Lippman, M.E., O’Brien, S.J., Joyce, M.J., 1978. Establishment and characterization of three new continuous cell lines derived from human breast carcinomas. Cancer Res. 38 (10), 3352 3364. Engler, A.J., Sen, S., Sweeney, H.L., Discher, D.E., 2006. Matrix elasticity directs stem cell lineage specification. Cell 126 (4), 677 689. Epstein, M.A., Barr, Y.M., 1964. Cultivation in vitro of human lymphoblasts from Burkitt’s malignant lymphoma. Lancet 1, 252 253. Espinosa-Hoyos, D., Jagielska, A., Homan, K.A., Du, H., Busbee, T., Anderson, D.G., et al., 2018. Engineered 3D-printed artificial axons. Sci. Rep. 8 (1), 478. Available from: https://doi.org/10.1038/s41598-017-18744-6. Fahrbach, K.M., Barry, S.M., Ayehunie, S., Lamore, S., Klausner, M., Hope, T.J., 2007. Activated CD34-derived Langerhans cells mediate transinfection with human immunodeficiency virus. J. Virol. 81 (13), 6858 6868. Felder, M., Trueeb, B., Stucki, A.O., Borcard, S., Stucki, J.D., Schnyder, B., et al., 2019. Impaired wound healing of alveolar lung epithelial cells in a breathing lung-on-a-chip. Front. Bioeng. Biotechnol. 7, 3. Available from: https://doi.org/10.3389/fbioe.2019.00003. Fizes¸an, I., Cambier, S., Moschini, E., Chary, A., Nelissen, I., Ziebel, J., et al., 2019. In vitro exposure of a 3D-tetraculture representative for the alveolar barrier at the airliquid interface to silver particles and nanowires. Part. Fibre Toxicol. 16 (1), 14. Available from: https://doi.org/10.1186/s12989-019-0297-1. Fogh, J., Fogh, J.M., Orfeo, T., 1977. One hundred and twenty-seven cultured human tumor cell lines producing tumors in nude mice. J. Natl. Cancer Inst. 59 (1), 221 226. Fogh, J., Trempe, G., 1975. New human tumor cell lines. In: Fogh, J. (Ed.), Human Tumor Cells In Vitro, 115 141. Plenum Press, New York. Freshney, R.I., 2016. Culture of Animal Cells: A Manual of Basic Technique and Specialized Applications, Seventh ed. Wiley-Blackwell, Hoboken, NJ. Fusaki, N., Ban, H., Nishiyama, A., Saeki, K., Hasegawa, M., 2009. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc. Jpn. Acad., Ser. B: Phys. Biol. Sci. 85, 348 362. Gabriel, E., Gopalakrishnan, J., 2017. Generation of iPSC-derived human brain organoids to model early neurodevelopmental disorders. J. Visualized Exp. 122, e55372. Available from: https://doi.org/10.3791/55372. Geraghty, R.J., Capes-Davis, A., Davis, J.M., Downward, J., Freshney, R.I., Knezevic, I., et al., 2014. Guidelines for the use of cell lines in biomedical research. Br. J. Cancer 111 (6), 1021 1046. Available from: https://doi.org/10.1038/bjc.2014.166. Gey, G.O., Coffman, W.D., Kubicek, M.T., 1952. Tissue culture studies of the proliferative capacity of cervical carcinoma and normal epithelium. Cancer Res. 12, 264 265. Giard, D.J., Aaronson, S.A., Parks, W.P., 1973. In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors. J. Natl. Cancer Inst. 51, 1417 1423.
35
36
CHAPTER 2 Cell sources and methods
Gilbert, P.M., Havenstrite, K.L., Magnusson, K.E., Sacco, A., Leonardi, N.A., Kraft, P., et al., 2010. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 329 (5995), 1078 1081. Available from: https://doi.org/10.1126/ science.1191035. Gjorevski, N., Sachs, N., Manfrin, A., Giger, S., Bragina, M.E., Ordo´n˜ez-Mora´n, P., et al., 2016. Designer matrices for intestinal stem cell and organoid culture. Nature 539 (7630), 560 564. Available from: https://doi.org/10.1038/nature20168. Gordon, J., Amini, S., White, M.K., 2013. General overview of neuronal cell culture. Methods Mol. Biol. 1078, 1 8. Gordon, S., Daneshian, M., Bouwstra, J., Caloni, F., Constant, S., Davies, D.E., et al., 2015. Non-animal models of epithelial barriers (skin, intestine and lung) in research, industrial applications and regulatory toxicology. Altern. Anim. Exp. 32 (4), 327 378. Available from: https://doi.org/10.14573/altex.1510051. Graham, F.L., Smiley, J., Russell, W.C., Nairn, R., 1977. Characteristics of a human cell line transformed by DNA from human adenovirus type 5. J. Gen. Virol. 36 (1), 59 74. Guillouzo, A., Corlu, A., Aninat, C., Glaise, D., Morel, F., Guguen-Guillouzo, C., 2007. The human hepatoma HepaRG cells: a highly differentiated model for studies of liver metabolism and toxicity of xenobiotics. Chem. Biol. Interact. 168 (1), 66 73. Hackett, T.L., Singhera, G.K., Shaheen, F., Hayden, P., Jackson, G.R., Hegele, R.G., et al., 2011. Intrinsic phenotypic differences of asthmatic epithelium and its inflammatory responses to respiratory syncytial virus and air pollution. Am. J. Respir. Cell Mol. Biol. 45 (5), 1090 1100. Available from: https://doi.org/10.1165/rcmb.2011-0031OC. Hayden, P.J., Bachelor, M., Ayehunie, S., Letasiova, S., Kaluzhny, Y., Kandarova, H., 2015. Application of MatTek in vitro reconstructed human skin models for safety, efficacy screening, and basic preclinical research. Appl. In Vitro Toxicol. 1 (3), 226 233. Hayflick, L., Moorhead, P.S., 1961. The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585 621. Heffelfinger, S.C., Hawkins, H.H., Barrish, J., Taylor, L., Darlington, G.J., 1992. SK HEP-1: a human cell line of endothelial origin. In Vitro Cell. Dev. Biol. 28A (2), 136 142. Herter, S., Morra, L., Schlenker, R., Sulcova, J., Fahrni, L., Waldhauer, I., et al., 2017. A novel three-dimensional heterotypic spheroid model for the assessment of the activity of cancer immunotherapy agents. Cancer Immunol. Immunother. 66 (1), 129 140. Available from: https://doi.org/10.1007/s00262-016-1927-1. Hofrichter, M., Nimtz, L., Tigges, J., Kabiri, Y., Schro¨ter, F., Royer-Pokora, B., et al., 2017. Comparative performance analysis of human iPSC-derived and primary neural progenitor cells (NPC) grown as neurospheres in vitro. Stem Cell Res. 25, 72 82. Available from: https://doi.org/10.1016/j.scr.2017.10.013. Holmberg, F.E., Seidelin, J.B., Yin, X., Mead, B.E., Tong, Z., Li, Y., et al., 2017. Culturing human intestinal stem cells for regenerative applications in the treatment of inflammatory bowel disease. EMBO Mol. Med. 9 (5), 558 570. Available from: https://doi.org/10.15252/emmm.201607260. Homan, K.A., Kolesky, D.B., Skylar-Scott, M.A., Herrmann, J., Obuobi, H., Moisan, A., et al., 2016. Bioprinting of 3D convoluted renal proximal tubules on perfusable chips. Sci. Rep. 6, 34845. Available from: https://doi.org/10.1038/srep34845. Homan, K.A., Gupta, N., Kroll, K.T., Kolesky, D.B., Skylar-Scott, M., Miyoshi, T., et al., 2019. Flow-enhanced vascularization and maturation of kidney organoids in vitro.
References
Nat. Methods 16 (3), 255 262Epub 2019 Feb 11. Available from: https://doi.org/ 10.1038/s41592-019-0325-y. Honegger, P., 2001. Overview of cell and tissue culture techniques. Curr. Protoc. Pharmacol. 4, 12.1.1 12.1.12. Available from: https://doi.org/10.1002/0471141755. ph1201s04. Hoppensack, A., Kazanecki, C.C., Colter, D., Gosiewska, A., Schanz, J., Walles, H., et al., 2014. A human in vitro model that mimics the renal proximal tubule. Tissue Eng., C: Methods 20 (7), 599 609Epub 2014 Jan 3. Available from: https://doi.org/10.1089/ten. TEC.2013.0446. Huh, D., Hamilton, G.A., Ingber, D.E., 2011. From 3D cell culture to organs-on-chips. Trends Cell Biol. 21 (12), 745 754Epub 2011 Oct 25. Review. Available from: https://doi.org/10.1016/j.tcb.2011.09.005. Ichimura, T., Asseldonk, E.J., Humphreys, B.D., Gunaratnam, L., Duffield, J.S., Bonventre, J., 2008. V. Kidney injury molecule-1 is a phosphatidylserine receptor that confers a phagocytic phenotype on epithelial cells. J. Clin. Invest. 118 (5), 1657 1668. Available from: https://doi.org/10.1172/JCI34487. Jeon, H., Tsui, J.H., Jang, S.I., Lee, J.H., Park, S., Mun, K., et al., 2015. Combined effects of substrate topography and stiffness on endothelial cytokine and chemokine secretion. ACS Appl. Mater. Interfaces 7 (8), 4525 4532. Available from: https://doi.org/ 10.1021/acsami.5b00554. Jiang, W., Lan, F., Zhang, H., 2016. Human induced pluripotent stem cells for inherited cardiovascular diseases modeling. Curr. Stem Cell Res. Ther. 11 (7), 533 541. Kaluzhny, Y., Kinuthia, M.W., Truong, T., Lapointe, A.M., Hayden, P., Klausner, M., 2018. New human organotypic corneal tissue model for ophthalmic drug delivery studies. Invest. Ophthalmol. Visual Sci. 59 (7), 2880 2898. Available from: https://doi.org/ 10.1167/iovs.18-23944. Kamdar, F., Klaassen Kamdar, A., Koyano-Nakagawa, N., Garry, M.G., Garry, D.J., 2015. Cardiomyopathy in a dish: using human inducible pluripotent stem cells to model inherited cardiomyopathies. J. Card. Failure 21 (9), 761 770. Available from: https:// doi.org/10.1016/j.cardfail.2015.04.010. Kim, D.H., Lipke, E.A., Kim, P., Cheong, R., Thompson, S., Delannoy, M., et al., 2010. Nanoscale cues regulate the structure and function of macroscopic cardiac tissue constructs. Proc. Natl. Acad. Sci. U.S.A. 107 (2), 565 570Published online 2009 Dec 16. Available from: https://doi.org/10.1073/pnas.0906504107. Kim, H.N., Jiao, A., Hwang, N.S., Kim, M.S., Kang, D.H., Kim, D.H., et al., 2013. Nanotopography-guided tissue engineering and regenerative medicine. Adv. Drug Delivery Rev. 65 (4), 536 558. Available from: https://doi.org/10.1016/j.addr.2012.07.014. Kim, J.H., Kurtz, A., Yuan, B.Z., Zeng, F., Lomax, G., Loring, J.F., et al., 2017. Report of the International Stem Cell Banking Initiative Workshop Activity: current hurdles and progress in seed-stock banking of human pluripotent stem cells. Stem Cells Transl. Med. 6 (11), 1956 1962. Available from: https://doi.org/10.1002/sctm.17-0144. Kim, Y., Kim, H., Ko, U.H., Oh, Y., Lim, A., Sohn, J.W., et al., 2016. Islet-like organoids derived from human pluripotent stem cells efficiently function in the glucose responsiveness in vitro and in vivo. Sci. Rep. 6, 35145. Available from: https://doi.org/ 10.1038/srep35145. King, S.M., Higgins, J.W., Nino, C.R., Smith, T.R., Paffenroth, E.H., Fairbairn, C.E., et al., 2017. 3D Proximal tubule tissues recapitulate key aspects of renal physiology to enable
37
38
CHAPTER 2 Cell sources and methods
nephrotoxicity testing. Front. Physiol. 8, 123. Available from: https://doi.org/10.3389/ fphys.2017.00123. Klaus, J., Kanton, S., Kyrousi, C., Ayo-Martin, A.C., Di Giaimo, R., Riesenberg, S., et al., 2019. Altered neuronal migratory trajectories in human cerebral organoids derived from individuals with neuronal heterotopia. Nat. Med. 25, 561 568. Available from: https://doi.org/10.1038/s41591-019-0371-0. Kolesky, D.B., Homan, K.A., Skylar-Scott, M., Lewis, J.A., 2018. In vitro human tissues via multi-material 3-D bioprinting. Altern. Lab. Anim. 46 (4), 209 215. Kolesky, D.B., Homan, K.A., Skylar-Scott, M.A., Lewis, J.A., 2016. Three-dimensional bioprinting of thick vascularized tissues. Proc. Natl. Acad. Sci. U.S.A. 113 (12), 3179 3184. Available from: https://doi.org/10.1073/pnas.1521342113. Kondo, Y., Toyoda, T., Inagaki, N., Osafune, K., 2018a. iPSC technology-based regenerative therapy for diabetes. J. Diabetes Invest. 9 (2), 234 243. Available from: https:// doi.org/10.1111/jdi.12702. Kondo, S., Mizuno, S., Hashita, T., Iwao, T., Matsunaga, T., 2018b. Using human iPS cellderived enterocytes as novel in vitro model for the evaluation of human intestinal mucosal damage. Inflammation Res. 67 (11 12), 975 984. Available from: https:// doi.org/10.1007/s00011-018-1193-0. Kshitiz, Park, J., Kim, P., Helen, W., Engler, A.J., Levchenko, A., et al., 2012. Control of stem cell fate and function by engineering physical microenvironments. Integr. Biol. 4 (9), 1008 1018. Kuehn, A., Kletting, S., de Souza Carvalho-Wodarz, C., Repnik, U., Griffiths, G., Fischer, U., et al., 2016. Human alveolar epithelial cells expressing tight junctions to model the air-blood barrier. Altern. Anim. Exp. 33 (3), 251 260. Available from: https://doi.org/ 10.14573/altex.1511131. Lampi, M.C., Reinhart-King, C.A., 2018. Targeting extracellular matrix stiffness to attenuate disease: from molecular mechanisms to clinical trials. Sci. Transl. Med. 10 (422), eaao0475. Available from: https://doi.org/10.1126/scitranslmed.aao0475. Lancaster, M.A., Renner, M., Martin, C.-A., Wenzel, D., Bicknell, L.S., Hurles, M.E., et al., 2013. Cerebral organoids model human brain development and microcephaly. Nature 501, 373 379. Lee, C.T., Bendriem, R.M., Wu, W.W., Shen, R.F., 2017. 3D brain Organoids derived from pluripotent stem cells: promising experimental models for brain development and neurodegenerative disorders. J. Biomed. Sci. 24 (1), 59. Available from: https://doi.org/ 10.1186/s12929-017-0362-8. Lee, S.A., No da, Y., Kang, E., Ju, J., Kim, D.S., Lee, S.H., 2013. Spheroid-based threedimensional liver-on-a-chip to investigate hepatocyte-hepatic stellate cell interactions and flow effects. Lab Chip 13 (18), 3529 3537. Available from: https://doi.org/ 10.1039/c3lc50197c. Lin, N.Y.C., Homan, K.A., Robinson, S.S., Kolesky, D.B., Duarte, N., Moisan, A., et al., 2019. Renal reabsorption in 3D vascularized proximal tubule models. Proc. Natl. Acad. Sci. U.S.A. 116 (12), 5399 5404. Available from: https://doi.org/10.1073/ pnas.1815208116. Lind, J.U., Busbee, T.A., Valentine, A.D., Pasqualini, F.S., Yuan, H., Yadid, M., et al., 2017. Instrumented cardiac microphysiological devices via multimaterial threedimensional printing. Nat. Mater. 16 (3), 303 308. Available from: https://doi.org/ 10.1038/nmat4782.
References
Liu, F., Mih, J.D., Shea, B.S., Kho, A.T., Sharif, A.S., Tager, A.M., et al., 2010. Feedback amplification of fibrosis through matrix stiffening and COX-2 suppression. J. Cell Biol. 190 (4), 693 706. Available from: https://doi.org/10.1083/jcb.201004082. Loo, L.S.W., Lau, H.H., Jasmen, J.B., Lim, C.S., Teo, A.K.K., 2018. An arduous journey from human pluripotent stem cells to functional pancreatic β cells. Diabetes Obes. Metab. 20 (1), 3 13. Available from: https://doi.org/10.1111/dom.12996. Lorsch, J.R., Collins, F.S., Lippincott-Schwartz, J., 2014. Cell biology. Fixing problems with cell lines. Science 346 (6216), 1452 1453. Available from: https://doi.org/ 10.1126/science.1259110. Ma, X., Qu, X., Zhu, W., Li, Y.S., Yuan, S., Zhang, H., et al., 2016. Deterministically patterned biomimetic human iPSC-derived hepatic model via rapid 3D bioprinting. Proc. Natl. Acad. Sci. U.S.A. 113 (8), 2206 2211. Available from: https://doi.org/10.1073/ pnas.1524510113. Madden, L.R., Nguyen, T.V., Garcia-Mojica, S., Shah, V., Le, A.V., Peier, A., et al., 2018. Bioprinted 3D primary human intestinal tissues model aspects of native physiology and ADME/Tox functions. iScience 2, 156 167. Available from: https://doi.org/10.1016/j. isci.2018.03.015. Mains, R.E., Patterson, P.H., 1973. Primary cultures of dissociated sympathetic neurons. I. Establishment of long-term growth in culture and studies of differentiated properties. J. Cell Biol. 59 (2 Pt 1), 329 345. Maldonado-Contreras, A., Birtley, J.R., Boll, E., Zhao, Y., Mumy, K.L., Toscano, J., et al., 2017. Shigella depends on SepA to destabilize the intestinal epithelial integrity via cofilin activation. Gut Microbes 8 (6), 544 560. Available from: https://doi.org/ 10.1080/19490976.2017.1339006. Marescotti, D., Serchi, T., Luettich, K., Xiang, Y., Moschini, E., Talikka, M., et al., 2019. How complex should an in vitro model be? Evaluation of complex 3D alveolar model with transcriptomic data and computational biological network models. Altern. Anim. Exp. 2019, [Epub ahead of print]. Available from: https://doi.org/10.14573/ altex.1811221. Maschmeyer, I., Lorenz, A.K., Schimek, K., Hasenberg, T., Ramme, A.P., Hu¨bner, J., et al., 2015a. A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents. Lab Chip 15 (12), 2688 2699. Available from: https://doi.org/10.1039/c5lc00392j. Maschmeyer, I., Hasenberg, T., Jaenicke, A., Lindner, M., Lorenz, A.K., Zech, J., et al., 2015b. Chip-based human liver-intestine and liver-skin co-cultures—a first step toward systemic repeated dose substance testing in vitro. Eur. J. Pharm. Biopharm. 95 (Pt A), 77 87. Available from: https://doi.org/10.1016/j.ejpb.2015.03.002. Matoba, N., Yamashita, T., Takayama, K., Sakurai, F., Mizuguchi, H., 2018. Optimal human iPS cell culture method for efficient hepatic differentiation. Differentiation 104, 13 21. Available from: https://doi.org/10.1016/j.diff.2018.09.005. Matsa, E., Burridge, P.W., Yu, K.H., Ahrens, J.H., Termglinchan, V., Wu, H., et al., 2016. Transcriptome profiling of patient-specific human iPSC-cardiomyocytes predicts individual drug safety and efficacy responses in vitro. Cell Stem Cell 19 (3), 311 325. Available from: https://doi.org/10.1016/j.stem.2016.07.006. McKernan, R., Watt, F.M., 2013. What is the point of large-scale collections of human induced pluripotent stem cells? Nat. Biotechnol. 31, 875 877. Erratum. Nature Biotechnology 31, 1148. Available from: https://doi.org/10.1038/nbt.2710.
39
40
CHAPTER 2 Cell sources and methods
Meier, F., Freyer, N., Brzeszczynska, J., Kno¨spel, F., Armstrong, L., Lako, M., et al., 2017. Hepatic differentiation of human iPSCs in different 3D models: a comparative study. Int. J. Mol. Med. 40 (6), 1759 1771. Available from: https://doi.org/10.3892/ijmm.2017.3190. Messner, S., Fredriksson, L., Lauschke, V.M., Roessger, K., Escher, C., Bober, M., et al., 2018. Transcriptomic, proteomic, and functional long-term characterization of multicellular three-dimensional human liver microtissues. Appl. In Vitro Toxicol. 4. Available from: https://doi.org/10.1089/aivt.2017.0022. Mih, J.D., Sharif, A.S., Liu, F., Marinkovic, A., Symer, M.M., Tschumperlin, D.J., 2011. A multiwell platform for studying stiffness-dependent cell biology. PLoS One 6 (5), e19929. Available from: https://doi.org/10.1371/journal.pone.0019929. Mitry, R., Hughes, R. (Eds.), 2012. Human Cell Culture Protocols. Humana Press, NY. Miura, S., Suzuki, A., 2018. Brief summary of the current protocols for generating intestinal organoids. Dev. Growth Differ. 60 (6), 387 392. Available from: https://doi.org/ 10.1111/dgd.12559. ¨ ztu¨rk, E., Arlov, Ø., Gatenholm, P., Zenobi-Wong, M., 2017. Alginate Mu¨ller, M., O sulfate-nanocellulose bioinks for cartilage bioprinting applications. Ann. Biomed. Eng. 45 (1), 210 223. Available from: https://doi.org/10.1007/s10439-016-1704-5. Mu´nera, J.O., Wells, J.M., 2017. Generation of gastrointestinal organoids from human pluripotent stem cells. Methods Mol. Biol. 1597, 167 177. Available from: https://doi. org/10.1007/978-1-4939-6949-4_12. Nguyen, D., Ha¨gg, D.A., Forsman, A., Ekholm, J., Nimkingratana, P., Brantsing, C., et al., 2017. Cartilage tissue engineering by the 3D bioprinting of iPS cells in a nanocellulose/alginate bioink. Sci. Rep. 7 (1), 658. Available from: https://doi.org/10.1038/ s41598-017-00690-y. Norona, L.M., Nguyen, D.G., Gerber, D.A., Presnell, S.C., Mosedale, M., Watkins, P.B., 2019. Bioprinted liver provides early insight into the role of Kupffer cells in TGF-β1 and methotrexate-induced fibrogenesis. PLoS One 14 (1), e0208958. Available from: https://doi.org/10.1371/journal.pone.0208958. Novak, R., Didier, M., Calamari, E., Ng, C.F., Choe, Y., Clauson, S.L., et al., 2018. Scalable fabrication of stretchable, dual channel, microfluidic organ chips. J. Visualized Exp. (140), . Available from: https://doi.org/10.3791/58151. Ntai, A., Baronchelli, S., La Spada, A., Moles, A., Guffanti, A., De Blasio, P., et al., 2017. A Review of research-grade human induced pluripotent stem cells qualification and biobanking processes. Biopreserv. Biobanking 15 (4), 384 392. Available from: https://doi.org/10.1089/bio.2016.0097. Olsson, I., Olofsson, T., Mauritzon, N., 1981. Characterization of mononuclear blood cellderived differentiation inducing factors for the human promyelocytic leukemia cell line HL-60. J. Natl. Cancer Inst. 67 (6), 1225 1230. Pamies, D., Barreras, P., Block, K., Makri, G., Kumar, A., Wiersma, D., et al., 2017. A human brain microphysiological system derived from induced pluripotent stem cells to study neurological diseases and toxicity. Altern. Anim. Exp. 34 (3), 362 376. Available from: https://doi.org/10.14573/altex.1609122. Parent, R., Marion, M.J., Furio, L., Tre´po, C., Petit, M.A., 2004. Origin and characterization of a human bipotent liver progenitor cell line. Gastroenterology 126 (4), 1147 1156. Park, J., Kim, H.N., Kim, D.H., Levchenko, A., Suh, K.Y., 2012. Quantitative analysis of the combined effect of substrate rigidity and topographic guidance on cell morphology.
References
IEEE Trans. Nanobiosci. 11 (1), 28 36. Available from: https://doi.org/10.1109/ TNB.2011.2165728. Peng, Y., Xiong, W.C., Mei, L., 2013. Culture of dissociated hippocampal neurons. In: Zhou, R., Mei, L. (Eds.), Neural Development: Methods and Protocols, Vol. 1018. Humana Press, Totowa, NJ, Methods in Molecular Medicine. Peters, M.F., Landry, T., Pin, C., Maratea, K., Dick, C., Wagoner, M.P., et al., 2019. Human 3D gastrointestinal microtissue barrier function as a predictor of drug-induced diarrhea. Toxicol. Sci. 168 (1), 3 17. Available from: https://doi.org/10.1093/toxsci/ kfy268. Picot, J., 2005. Second ed. Human Cell Culture Protocols, vol. 107. Humana Press, NJ, Methods in Molecular Medicine. Pirnay, J.P., Baudoux, E., Cornu, O., Delforge, A., Delloye, C., Guns, J., et al., 2015. Access to human tissues for research and product development: from EU regulation to alarming legal developments in Belgium. EMBO Rep. 16 (5), 557 562. Available from: https://doi.org/10.15252/embr.201540070. Plummer, S., Wallace, S., Ball, G., Lloyd, R., Schiapparelli, P., Quin˜ones-Hinojosa, A., et al., 2019. A Human iPSC-derived 3D platform using primary brain cancer cells to study drug development and personalized medicine. Sci. Rep. 9, 1407. Available from: https://doi.org/10.1038/s41598-018-38130-0. Ponten, J., Macintyre, E.H., 1968. Long term culture of normal and neoplastic human glia. Acta Pathol. Microbiol. Scand. 74, 465 486. Proctor, W.R., Foster, A.J., Vogt, J., Summers, C., Middleton, B., Pillings, M.A., et al., 2017. Utility of spherical human liver microtissues for prediction of clinical druginduced liver injury. Arch. Toxicol. 91 (8), 2849 2863. Available from: https://doi. org/10.1007/s00204-017-2002-1. Qian, T., Maguire, S.E., Canfield, S.G., Bao, X., Olson, W.R., Shusta, E.V., et al., 2017. Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial cells. Sci. Adv. 3 (11), e1701679. Available from: https://doi.org/10.1126/ sciadv.1701679. Rahmani, S., Breyner, N.M., Su, H.M., Verdu, E.F., Didar, T.F., 2019. Intestinal organoids: a new paradigm for engineering intestinal epithelium in vitro. Biomaterials 194, 195 214. Available from: https://doi.org/10.1016/j.biomaterials.2018.12.006. Second ed. Randell, S.H., Fulcher, M. (Eds.), 2013. Epithelial Cell Culture Protocols, vol. 945. Humana Press, NJ, Methods in Molecular Medicine. Reddel, R.R., Ke, Y., Gerwin, B.I., McMenamin, M.G., Lechner, J.F., Su, R.T., et al., 1988. Transformation of human bronchial epithelial cells by infection with SV40 or adenovirus-12 SV40 hybrid virus, or transfection via strontium phosphate coprecipitation with a plasmid containing SV40 early region genes. Cancer Res. 48 (7), 1904 1909. Rismani Yazdi, S., Shadmani, A., Bu¨rgel, S.C., Misun, P.M., Hierlemann, A., Frey, O., 2015. Adding the “heart” to hanging drop networks for microphysiological multi-tissue experiments. Lab Chip 15 (21), 4138 4147. Available from: https://doi.org/10.1039/ c5lc01000d. Ronaldson-Bouchard, K., Ma, S.P., Yeager, K., Chen, T., Song, L., Sirabella, D., et al., 2018. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556 (7700), 239 243. Available from: https://doi.org/10.1038/s41586-0180016-3.
41
42
CHAPTER 2 Cell sources and methods
Ronaldson-Bouchard, K., Vunjak-Novakovic, G., 2018. Organs-on-a-chip: A fast track for engineered human tissues in drug development. Cell Stem Cell 22 (3), 310 324. Available from: https://doi.org/10.1016/j.stem.2018.02.011. Ruiz-Ojeda, F.J., Rupe´rez, A.I., Gomez-Llorente, C., Gil, A., Aguilera, C.M., 2016. Cell models and their application for studying adipogenic differentiation in relation to obesity: a review. Int. J. Mol. Sci. 17 (7), 1040. Available from: https://doi.org/10.3390/ ijms17071040. Sachs, N., Tsukamoto, Y., Kujala, P., Peters, P.J., Clevers, H., 2017. Intestinal epithelial organoids fuse to form self-organizing tubes in floating collagen gels. Development 144 (6), 1107 1112. Available from: https://doi.org/10.1242/dev.143933. Sallam, K., Kodo, K., Wu, J.C., 2014. Modeling inherited cardiac disorders. Circ. J. 78 (4), 784 794. Sato, T., Clevers, H., 2015. SnapShot: growing organoids from stem cells. Cell 161 (7), 1700 1700.e1. Available from: https://doi.org/10.1016/j.cell.2015.06.028. Sayed, N., Liu, C., Wu, J.C., 2016. Translation of human-induced pluripotent stem cells: From clinical trial in a dish to precision medicine. J. Am. Coll. Cardiol. 67 (18), 2161 2176. Available from: https://doi.org/10.1016/j.jacc.2016.01.083. Schmeichel, K.L., Bissell, M.J., 2003. Modeling tissue-specific signaling and organ function in three dimensions. J. Cell Sci. 116 (Pt 12), 2377 2388. Schutgens, F., Rookmaaker, M.B., Margaritis, T., Rios, A., Ammerlaan, C., Jansen, J., et al., 2019. Tubuloids derived from human adult kidney and urine for personalized disease modeling. Nat. Biotechnol. 37 (3), 303 313. Available from: https://doi.org/ 10.1038/s41587-019-0048-8. Schwartz, M.P., Hou, Z., Propson, N.E., Zhang, J., Engstrom, C.J., Santos Costa, V., et al., 2015. Human pluripotent stem cell-derived neural constructs for predicting neural toxicity. Proc. Natl. Acad. Sci. U.S.A. 112, 12516 12521. Sharma, A., Burridge, P.W., McKeithan, W.L., Serrano, R., Shukla, P., Sayed, N., et al., 2017. High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells. Sci. Transl. Med. 9 (377), eaaf2584. Available from: https://doi.org/10.1126/scitranslmed.aaf2584. Sinagoga, K.L., McCauley, H.A., Mu´nera, J.O., Reynolds, N.A., Enriquez, J.R., Watson, C., et al., 2018. Deriving functional human enteroendocrine cells from pluripotent stem cells. Development 145 (19), dev165795. Available from: https://doi.org/10.1242/ dev.165795. Smith, A.S.T., Davis, J., Lee, G., Mack, D.L., Kim, D.H., 2016. Muscular dystrophy in a dish: engineered human skeletal muscle mimetics for disease modeling and drug discovery. Drug Discov. Today 21 (9), 1387 1398. Available from: https://doi.org/ 10.1016/j.drudis.2016.04.013. Smith, Q., Chan, X.Y., Carmo, A.M., Trempel, M., Saunders, M., Gerecht, S., 2017. Compliant substratum guides endothelial commitment from human pluripotent stem cells. Sci. Adv. 3 (5), e1602883. Available from: https://doi.org/10.1126/sciadv.1602883. Sone, M., Nakao, K., 2013. Vascular research using human pluripotent stem cells and humoral factors. Endocr. J. 60 (4), 397 402. Stebbins, M.J., Wilson, H.K., Canfield, S.G., Qian, T., Palecek, S.P., Shusta, E.V., 2016. Differentiation and characterization of human pluripotent stem cell-derived brain microvascular endothelial cells. Methods 101, 93 102. Available from: https://doi.org/ 10.1016/j.ymeth.2015.10.016.
References
Sundstro¨m, C., Nilsson, K., 1976. Establishment and characterization of a human histiocytic lymphoma cell line (U-937). Int. J. Cancer 17 (5), 565 577. Takahashi, K., Okita, K., Nakagawa, M., Yamanaka, S., 2007a. Induction of pluripotent stem cells from fibroblast cultures. Nat. Protoc. 2, 3081 3089. Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., et al., 2007b. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861 872. Takahashi, Y., Hori, Y., Yamamoto, T., Urashima, T., Ohara, Y., Tanaka, H., 2015. 3D spheroid cultures improve the metabolic gene expression profiles of HepaRG cells. Biosci. Rep. 35 (3), e00208. Available from: https://doi.org/10.1042/ BSR20150034. Takahashi, Y., Sato, S., Kurashima, Y., Yamamoto, T., Kurokawa, S., Yuki, Y., et al., 2018. A refined culture system for human induced pluripotent stem cell-derived intestinal epithelial organoids. Stem Cell Rep. 10 (1), 314 328. Available from: https://doi. org/10.1016/j.stemcr.2017.11.004. Takasato, M., Little, M.H., 2017. Making a kidney organoid using the directed differentiation of human pluripotent stem cells. Methods Mol. Biol. 1597, 195 206. Available from: https://doi.org/10.1007/978-1-4939-6949-4_14. Takayama, K., Mizuguchi, H., 2017. Generation of human pluripotent stem cell-derived hepatocyte-like cells for drug toxicity screening. Drug Metab. Pharmacokinet. 32 (1), 12 20. Available from: https://doi.org/10.1016/j.dmpk.2016.10.408. Talkhabi, M., Aghdami, N., Baharvand, H., 2016. Human cardiomyocyte generation from pluripotent stem cells: a state-of-art. Life Sci. 145, 98 113. Available from: https://doi. org/10.1016/j.lfs.2015.12.023. Tanaka, A., Yuasa, S., Node, K., Fukuda, K., 2015. Cardiovascular disease modeling using patient-specific induced pluripotent stem cells. Int. J. Mol. Sci. 16 (8), 18894 18922. Available from: https://doi.org/10.3390/ijms160818894. Tilghman, R.W., Cowan, C.R., Mih, J.D., Koryakina, Y., Gioeli, D., Slack-Davis, J.K., et al., 2010. Matrix rigidity regulates cancer cell growth and cellular phenotype. PLoS One 5 (9), e12905. Available from: https://doi.org/10.1371/journal. pone.0012905. Tong, Z., Martyn, K., Yang, A., Yin, X., Mead, B.E., Joshi, N., et al., 2018. Towards a defined ECM and small molecule based monolayer culture system for the expansion of mouse and human intestinal stem cells. Biomaterials 154, 60 73. Available from: https://doi.org/10.1016/j.biomaterials.2017.10.038. Tsuchiya, S., Yamabe, M., Yamaguchi, Y., Kobayashi, Y., Konno, T., Tada, K., 1980. Establishment and characterization of a human acute monocytic leukemia cell line (THP-1). Int. J. Cancer 26 (2), 171 176. Tsuruo, T., Hamilton, T.C., Louie, K.G., Behrens, B.C., Young, R.C., Ozols, R.F., 1986. Collateral susceptibility of adriamycin-, melphalan- and cisplatin-resistant human ovarian tumor cells to bleomycin. Jpn. J. Cancer Res. 77, 941 945. Valente, M.J., Henrique, R., Costa, V.L., Jero´nimo, C., Carvalho, F., Bastos, M.L., et al., 2011. A rapid and simple procedure for the establishment of human normal and cancer renal primary cell cultures from surgical specimens. PLoS One 6 (5), e19337. Available from: https://doi.org/10.1371/journal.pone.0019337. Vanderburgh, J., Sterling, J.A., Guelcher, S.A., 2017. 3D printing of tissue engineered constructs for in vitro modeling of disease progression and drug screening. Ann.
43
44
CHAPTER 2 Cell sources and methods
Biomed. Eng. 45 (1), 164 179. Available from: https://doi.org/10.1007/s10439-0161640-4. Wang, C., Hei, F., Ju, Z., Yu, J., Yang, S., Chen, M., 2016. Differentiation of urinederived human induced pluripotent stem cells to alveolar type II epithelial cells. Cell. Reprogram. 18 (1), 30 36. Available from: https://doi.org/10.1089/ cell.2015.0015. Wang, Y., Alhaque, S., Cameron, K., Meseguer-Ripolles, J., Lucendo-Villarin, B., Rashidi, H., et al., 2017. Defined and scalable generation of hepatocyte-like cells from human pluripotent stem cells. J. Visualized Exp. (121), . Available from: https://doi.org/10.3791/55355. Warren, L., Manos, P.D., Ahfeldt, T., Loh, Y.H., Li, H., Lau, F., et al., 2010. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7, 618 630. Wells, R.G., 2008. The role of matrix stiffness in regulating cell behavior. Hepatology 47 (4), 1394 1400. Available from: https://doi.org/10.1002/hep.22193. Wilkinson, D.C., Mellody, M., Meneses, L.K., Hope, A.C., Dunn, B., Gomperts, B.N., 2018. Development of a three-dimensional bioengineering technology to generate lung tissue for personalized disease modeling. Curr. Protoc. Stem Cell Biol. 46 (1), e56. Available from: https://doi.org/10.1002/cpsc.56. Wong, W.T., Huang, N.F., Botham, C.M., Sayed, N., Cooke, J.P., 2012. Endothelial cells derived from nuclear reprogramming. Circ. Res. 111 (10), 1363 1375. Available from: https://doi.org/10.1161/CIRCRESAHA.111.247213. Xia, Z., Jin, S., Ye, K., 2018. Tissue and organ 3D bioprinting. SLAS Technol. 23 (4), 301 314. Available from: https://doi.org/10.1177/2472630318760515. Yamashita, T., Takayama, K., Sakurai, F., Mizuguchi, H., 2018. Billion-scale production of hepatocyte-like cells from human induced pluripotent stem cells. Biochem. Biophys. Res. Commun. 496 (4), 1269 1275. Available from: https://doi.org/10.1016/j. bbrc.2018.01.186. Yabe, S.G., Fukuda, S., Takeda, F., Nashiro, K., Shimoda, M., Okochi, H., 2017. Efficient generation of functional pancreatic β-cells from human induced pluripotent stem cells. J. Diabetes 9 (2), 168 179. Available from: https://doi.org/10.1111/ 1753-0407.12400. You, M.-H., Kwak, M.K., Kim, D.-H., Kim, K., Levchenko, A., Kim, D.-Y., et al., 2010. Synergistically enhanced osteogenic differentiation of human mesenchymal stem cells by culture on nanostructured surfaces with induction media. Biomacromolecules 11 (7), 1856 1862. Available from: https://doi.org/10.1021/bm100374n. Yu, J., Vodyanik, M.A., Smuga-Otto, K., Antosiewicz-Bourget, J., Frane, J.L., Tian, S., et al., 2007. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917 1920. Zhu, W., Qu, X., Zhu, J., Ma, X., Patel, S., Liu, J., et al., 2017. Direct 3D bioprinting of prevascularized tissue constructs with complex microarchitecture. Biomaterials 124, 106 115. Available from: https://doi.org/10.1016/j.biomaterials.2017.01.042. Zuellig, R.A., Cavallari, G., Gerber, P., Tschopp, O., Spinas, G.A., Moritz, W., et al., 2014. Improved physiological properties of gravity-enforced reassembled rat and human pancreatic pseudo-islets. J. Tissue Eng. Regener. Med. 11, 109 120. Available from: https://doi.org/10.1002/term.1891.
Further reading
Further reading Huh, D., Matthews, B.D., Mammoto, A., Montoya-Zavala, M., Hsin, H.Y., Ingber, D.E., 2010. Reconstituting organ-level lung functions on a chip. Science 328 (5986), 1662 1668. Available from: https://doi.org/10.1126/science.1188302. Shi, Y., Inoue, H., Wu, J.C., Yamanaka, S., 2017. Induced pluripotent stem cell technology: a decade of progress. Nat. Rev. Drug Discov. 16 (2), 115 130. Available from: https://doi.org/10.1038/nrd.2016.245.
45