Journal Pre-proof Towards manufacturing of human organoids
Aswathi Ashok, Deepak Choudhury, Fang Yu, Walter Hunziker PII:
S0734-9750(19)30160-0
DOI:
https://doi.org/10.1016/j.biotechadv.2019.107460
Reference:
JBA 107460
To appear in:
Biotechnology Advances
Received date:
16 April 2019
Revised date:
8 October 2019
Accepted date:
10 October 2019
Please cite this article as: A. Ashok, D. Choudhury, F. Yu, et al., Towards manufacturing of human organoids, Biotechnology Advances (2018), https://doi.org/10.1016/ j.biotechadv.2019.107460
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
© 2018 Published by Elsevier.
Journal Pre-proof
Towards Manufacturing of Human Organoids Aswathi Ashok1,1
[email protected], Deepak Choudhury2,1,*
[email protected]., Fang Yu2
[email protected], Walter Hunziker1,3 1Institute
of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore 2Bio-Manufacturing Programme, Singapore Institute of Manufacturing Technology (SIMTech), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-04, Innovis, Singapore 138634, Singapore 3Department of Physiology, 2 Medical Drive, MD9, National University of Singapore, Singapore 117593, Singapore
ur
na
lP
re
-p
ro
of
author.
Jo
*Corresponing
1
Co-first authors
Journal Pre-proof Abstract Organoids are 3D miniature versions of organs produced from stem cells derived from either patient or healthy individuals in vitro that recapitulate the actual organ. Organoid technology has ensured an alternative to pre-clinical drug testing as well as being currently used for “personalized medicine” to modulate the treatment as they are uniquely identical to each patient’s genetic makeup. Researchers have succeeded in producing different types of organoids and have demonstrated their efficient application in various fields such as disease modeling, pathogenesis, drug screening and regenerative medicine. There are several protocols for
of
fabricating organoids in vitro. In this comprehensive review, we focus on key methods of
ro
producing organoids and manufacturing considerations for each of them while providing insights on the advantages, applications and challenges of these methods. We also discuss pertinent
-p
challenges faced during organoid manufacturing and various bioengineering approaches that can improve the organoid manufacturing process. Organoids size, number and the reproducibility of
re
the fabrication processes are touched upon. The major factors which are involved in organoids
lP
manufacturing like spatio-temporal controls, scaffold designs/types, cell culture parameters and
na
vascularization have been highlighted.
Keywords: Organoids, Organoids manufacturing, Organoids production, Stem cells, Embryoid bodies, 3D cell culture, Air-
Jo
ur
liquid-interface
Journal Pre-proof 1. Introduction Organoid technology has recently emerged as a cutting-edge tool for fundamental research (Lancaster, Madeline A. and Knoblich, Juergen A., 2014; Sato et al., 2009) as well as translational medicine (Bartfeld and Clevers, 2017; Fatehullah et al., 2016; Sato and Clevers, 2013). Animals have been excellent models for research, however, not all the drugs that clear pre-clinical animal trials succeed during clinical trials due to differences in animal and human physiology (Akkerman, 2017; Kamb et al., 2006; Kaushik et al., 2018; van der Worp et al., 2010). Organoids recapitulate the complexity of the actual human organs and could potentially
of
complement animal models in pre-clinical trials (Akkerman, 2017; Kaushik et al., 2018; Weeber et al., 2017). Manufacturing organoids in large scale could benefit a variety of fields including
ro
disease modelling (Huch et al.; McCracken et al., 2014; Weeber et al., 2017), personalized drug
-p
screening (Huang et al., 2015; Skardal et al., 2016), pathogenesis (Huch et al.; Weeber et al., 2017), cancer research (Drost and Clevers, 2018; Drost et al., 2017) and regenerative medicine
re
(Lancaster, M. A. and Knoblich, J. A., 2014; Taguchi et al., 2014; Takebe et al.). Organoids are defined as scaled-down, miniature versions of an organ generated in vitro in a 3D culture system
lP
that can recapitulate its in vivo microenvironment (Kaushik et al., 2018). Recently, the definition of organoid has been narrowed down to self-organizing 3D structures grown from stem cells that
ur
organs (Dutta et al., 2017).
na
mimic the in vivo environment, architecture and multi-lineage differentiation of the original
Organoids could be produced from various sources such as pluripotent stem cells (Lancaster et
Jo
al., 2013; Quadrato et al., 2017) (i.e) embryonic stem cells (Volkner et al., 2016), adult stem cells (Broutier et al., 2016; Sato et al., 2009), patient-derived induced pluripotent stem cells (Crespo et al., 2017; Lancaster et al., 2013), and induced pluripotent stem cells from a donor’s skin fibroblast cells, blood cells or other cell type (Wang et al., 2018). These organoids can then be stored in biobanks for regenerative medicine and fundamental research (Sato and Clevers, 2013). Though both spheroids and organoids are 3D structures made of cells, they are not the same . Spheroids are 3D structures formed under non-adherent conditions from cancer cell lines (Sutherland Rm Fau - McCredie et al.), whereas organoids are 3D cell aggregates that comprises of cells that are more organ specific in lineage that develop from stem cells or progenitor cells in the presence of extracellular matrix in a 3D cell culture medium where cells self-organize recapitulating the in vivo organ-formation process (Lancaster, Madeline A. and Knoblich,
Journal Pre-proof Juergen A., 2014).
Organoids unlike spheroids, have a higher order self organization/self
assembly that is more similar to the in vivo organ formation and hence serve as better in vitro models as they better mimic the complexity of an actual organ (Li, 2019; Merck, 2019). When transplanted into animal models, organoids can further develop and become vascularized (Mansour et al., 2018) The term “spheroid” is sometimes also used to refer to an intermediate stage of organoid growth (Crespo et al., 2017; Freedman et al., 2015; McCracken et al., 2014). Organoids have recently become popular in vitro 3D cell culture model in areas of developmental biology, drug screening, and disease modelling. Arora et al. in 2017 found that
of
13% of spheroids developed into mature intestinal organoids and hypothesized that the key
ro
specification required in spheroid maturation is the morphological feature of the inner mass and diameter (Arora et al., 2017). They demonstrated that spheroids with a diameter greater than 75
-p
μm act as pre-organoids that later mature into mature intestinal organoids (Arora et al., 2017). A study by McCracken et al. showed that only a small fraction of the spheroids matured in
re
organoids (McCracken et al., 2014). Currently, spheroids produced are either scaffold based
lP
(Hydrogels or inserts) / non scaffold based 3D cell aggregates. The study by (Arora et al., 2017) and (McCracken et al., 2014) opens up avenues for organoid research from a manufacturing
na
point of view, where spheroids can be manipulated to develop organoids. With regards to the above mentioned studies, different techniques (Costa et al., 2016) to produce spheroids such as hanging drop method, hanging drop, microfluidic-based assembly, spinner flasks and liquid
ur
overlay method have the potential to be further manipulated to develop organoids in future,
Jo
though currently there isn’t much work on this yet (Bello, 2019, Sep 26)
Organoid models have been developed for a number of organs and include intestinal organoids (Sato et al., 2009; Spence et al., 2011), cerebral organoids (Lancaster, M. A. and Knoblich, J. A., 2014; Lancaster et al., 2013), kidney organoids (Taguchi et al., 2014; Takasato et al., 2016), hepatic organoids (Mitaka, 2002), retinal organoids (Boucherie et al.; DiStefano et al., 2018; Eiraku et al., 2008; Tucker et al., 2014; Volkner et al., 2016), pancreatic organoids (Greggio et al., 2013; Hohwieler et al., 2017; Huang et al., 2015; Li, X. et al., 2014), lung organoids (Bals et al., 2004; Escaffre et al., 2016; Fessart et al., 2013; Nadkarni, R. R. et al., 2016; Pageau et al., 2011; Vaughan et al., 2006), colonic organoids (Crespo et al., 2017),
gastric organoids
Journal Pre-proof (McCracken et al., 2014), cardiac organoids (Nugraha et al., 2019; Stevens et al., 2009), thyroid organoids (Toda et al., 2002), prostate organoids (Gao et al.), saliva-secreting organoids(Ferreira et al., 2019), mammary gland (breast) organoids (Lee et al., 2007; Simian et al., 2001; Weaver et al., 1997), lingual organoids (Hisha et al., 2013), , placenta organoids (Vogt, 2019) and spinal organoids(Research, 2019). In an earlier study, it was observed that cells behave differently in 2D and 3D cultures (Weaver et al., 1997), this was suspected to be one of the main reasons for therapeutic drug failures in clinical trials for drugs tested using 2D models. The identification of Lgr5 gene and its significance as a biomarker for adult stem cells in many tissues marked the
of
beginning of research with 3D cell cultures (Hsu et al., 1998). In 2008, Sasai et al. used the
ro
SFEBq technique to create 3D cerebral cortex tissue from pluripotent stem cells (Eiraku et al., 2008). The significant role of Lgr5 gene to give rise to crypt-villus structures in the absence of a
-p
non-epithelial (mesenchymal) cellular niche and eventually intestinal organoid in vitro was demonstrated by Sato et al. (Sato et al., 2009). Discovery of this intestinal culture system was a
re
major breakthrough in the field of organoids. The study also inferred that organoids developed
lP
from a crypt or a single stem cell (Lgr5+ cells) are indistinguishable (Sato et al., 2009). The starting cell type and system conditions, endogenous and exogenous signals and the physical
na
characteristics of the culture environment were identified as three main characteristic features that influence the successful formation of organoids in vitro (Rossi et al., 2018).
ur
Commercial production of organoids at a large scale still needs to be established, although a few biotech companies are starting to move in that direction (Chakradhar, 2016). This paper
Jo
elaborates on the key methods of producing organoids in vitro that have the potential to be used as standard protocols for mass organoid manufacturing. Before highlighting the methods of organoid production, important clinical applications of organoids are summarized. 2. Clinical applications of organoids Organoids have opened remarkable opportunities in pathogenesis and disease modelling, drug screening and regenerative medicine. These are described in detail below. 2.1 Pathogenesis and Disease models Animals are common models for pathogenesis studies (Campo, 2002; Gold et al., 2006; Lazear et al., 2016). Recent studies have revealed the usage of organoids as an alternative to animal models for disease modelling (Hwang et al., 2016; Lancaster et al., 2013). There are three main
Journal Pre-proof techniques to introduce microbes into organoids to generate disease models: (a) Infecting the dissociated cells before 3D organoid formation (b) microinjecting into the lumen of the organoids and (c) addition of microbes to 2D cultures derived from organoids (Dutta et al., 2017). Disease models can also be created by genome editing in organoids using CRISPR/Cas9 technique to study the genetic basis of diseases. One such example is the creation of genetically modified kidney organoids [produced from human pluripotent stem cells (hPSCs)] to model genetic kidney disease using CRISPR_Cas9 genome editing demonstrated by Freedman et al.(Freedman et al., 2015). There are also patient-derived organoids that model diseases, these
of
have applications in personalized medicine and drug screening (Noordhoek et al., 2016).
ro
Lancaster et al. succeeded in producing miniature cerebral organoids that model microcephaly by using patient-specific iPSCs and RNAi (Lancaster et al., 2013). Cerebral organoids have been
-p
modeled for microcephaly (Lancaster et al., 2013). This was also the first study to report human iPSCs-derived cerebral organoids. Cerebral organoids have also been used as disease models to
re
study numerous other conditions such as seckel syndrome (Gabriel et al., 2016), zika virus
lP
infections (Cugola et al., 2016; Garcez et al., 2016; Lancaster et al., 2013; Qian et al., 2016) and autism spectrum disorders (Mariani et al., 2015). McCracken et al. came up with a robust method
na
for generating human gastric organoids and demonstrated its use as a model for the pathogenesis of Helicobacter pylori infection (McCracken et al., 2014). Retinal organoids modelled for the study of glaucoma (Tucker et al., 2014) and intestinal organoids (Spence et al., 2011) produced
ur
with future prospects of being used in disease modelling to study gastrointestinal diseases
Jo
(Watson et al., 2014). Lung organoids produced using the air-liquid interface culture method were used to model Nipah virus infection (Escaffre et al., 2016). Nadkarni et al. in their review explained lung organoid systems catered for lung development and diseases (Nadkarni, R. R. et al., 2016). Hohwieler et al. made pancreatic organoids directly from human iPSCs and used them as a model for the pathogenesis of cystic fibrosis and other pancreatic disorders (Hohwieler et al., 2017). Strange et al. developed human testicular organoids to study the pathogenesis of zika virus infection (Strange et al., 2018). Organoids from both Pluripotent stem cells (PSCs) (Finkbeiner et al., 2012) (Porotto et al., 2019) as well as adult stem cells (ASC) (Bartfeld et al., 2015; Bartfeld and Clevers, 2015; Castellanos-Gonzalez et al., 2013; Ettayebi et al., 2016) (Zomer-van Ommen et al., 2016) (Yin et al., 2015) also have a great potential use as models for infectious diseases as they enable host-pathogen studies.
Journal Pre-proof Organoids & Cancer research A major attribute of organoids that makes it significant in cancer research is the fact that organoids derived from patient cells mimic the tumor characteristics (Drost and Clevers, 2018). Since organoids are genetically stable they are used to study cancer and tissue homeostasis (Kaushik et al., 2018). Broutier et al. recently developed primary liver cancer organoids for drug screening and disease modelling (Broutier et al., 2017). There are numerous patient-derived organoids that help are extensively used for cancer research (Drost and Clevers, 2018). The following is a list of all the major patient-derived tumour organoids, developed to date used
of
extensively in cancer research: Liver (Broutier et al., 2017), prostate (Gao et al., 2014), Colon
ro
(Sato, Toshiro et al., 2011), Gastrointestinal (Seidlitz et al., 2019; Vlachogiannis, Georgios et al., 2018), Lung (Nadkarni, Rohan R. et al., 2016), Breast (DeRose et al., 2013) and bladder tumour
-p
organoids (Lee et al., 2018) . Recent research found that combining tumour organoids with
re
fibroblast and immune cells resulted in models that enable the study of tumour microenvironment, thereby improving the immune-oncology applications of tumour organoids
lP
(Tuveson and Clevers, 2019). There are a number of well-known organoid cancer biobanks that aid in cancer research and drug discovery such as organoid biobank for breast cancer organoids
na
(Sachs et al., 2018) , colon (van de Wetering et al., 2015) , ovarian (Kopper et al., 2019) and
Jo
2.2.Drug screening
ur
gastric cancer organoids (Yan et al., 2018).
Production of patient-derived organoids that closely resemble the original tumors are considered a major breakthrough, as they offer an excellent alternative to pre-clinical drug testing (Weeber et al., 2017). There have been attempts to build a large organoid biobank to facilitate the study of drug development and screening (Weeber et al., 2017). Vlachogiannis et al. showed that drug responses from patient-derived tumor organoids had a high degree of similarity to the in vivo response of the patient in the clinic (Vlachogiannis, G. et al., 2018). Colonic organoids generated from iPSCs of patients with familial adenomatous polyposis was used for drug testing by Crespo et al. (Crespo et al., 2017). Numerous mutations on the cystic fibrosis transmembrane conductance regulator (CFTR) were found to be a leading cause of cystic fibrosis in human
Journal Pre-proof (Dekkers et al., 2013; Weeber et al., 2017).Patient derived intestinal organoids have been used for testing of cystic fibrosis drugs(Noordhoek et al., 2016). 2.3 Regenerative medicine Organoids can potentially be used as an alternative for organ transplantation in the future. Cerebral organoids are miniature versions of brains developed in vitro, used to study the development and function of the brain (Lancaster, M. A. and Knoblich, J. A., 2014). Liver
of
transplants are required to save patients suffering from chronic liver disorders resulting in liver failure (Huch M Fau - Boj et al.). Scientists are trying to use liver organoids as an organ
ro
transplant alternative (Huch M Fau - Boj et al.). Takebe et al. succeeded in generating a vascularized human liver by transplanting liver buds produced in vitro and growing it in mice
-p
(Takebe et al.). Taguchi et al. have successfully achieved transplantation of kidney organoid in
re
adult mice (Taguchi et al., 2014) which, if successful in humans, could one day replace kidney dialysis or renal transplantation in kidney failure patients. Schwank et al demonstrated gene
lP
correction in cystic fibrosis patient-derived organoids using CRISPR/Cas9 (Schwank et al., 2013). All the aforementioned studies showcase the importance of organoids in regenerative
na
medicine, however there many safety and compatibility issues linked with the application of organoids as transplants into humans (Fatehullah et al., 2016). The need for GMP compliant
ur
culture system for manufacturing organoids using scaffolds that are tissue compatible is
Jo
necessary to reduce tissue rejection and organoid incompatibility issues (Dakhore et al., 2018). Types of Organoids (Based on source): Depending on the source, there are two main types of organoids produced (Huch and Koo, 2015)- a) Organoids produced from adult stem cells (ASC Approach): The source of these organoids are adult stem cells/progenitor cells or fragments of isolated primary tissues (Huch and Koo, 2015). This approach for organoid production mainly depends on the tissue repair/renewal principles. Wnt Signalling in adult stem cells have been identified as the key factor that enables the organoids produced from ASC approach expand indefinitely (Maimets et al., 2016). The crypt isolation protocol optimized by Sato et al. in 2009 is mainly used for generating intestinal organoids from primary tissues. To date, only epithelial organoids can be produced from adult stem cells such as
Journal Pre-proof intestinal organoids (Broutier et al., 2016; Ootani et al., 2009; Sato et al., 2009), gastric organoids (Barker et al., 2010; Bartfeld et al., 2015; Li, Xingnan et al., 2014; Stange et al., 2013), liver organoids (Broutier et al., 2016; Huch et al., 2013), pancreatic organoids (Broutier et al., 2016), salivary gland (Maimets et al., 2016; Nanduri et al., 2014), tongue (Hisha et al., 2013) b) Organoids produced from pluripotent stem cells (PSC Approach): The source for these organoids are either embryonic stem cells (ESCs) taken from inner cell mass of blastocysts or induced pluripotent stem cells (iPSCs) produced from somatic cells. This approach mainly involves directed differentiation of pluripotent stem cells (both ESCs as well as iPSCs) to form
of
the respective stem cell progenitor cells. Definitive endoderm layer is what gives rise to digestive
ro
tracts, respiratory tract epithelial lining, lungs, thyroid, pancreas, thymus and liver. The appearance of the primitive streak in the posterior epiblast marks the beginning of the
-p
differentiation process (D'Amour et al., 2005). The ingression of posterior epiblast cells through the primitive streak leads to EMT (epithelial-to-mesenchymal transition) which later either
re
become the mesoderm or definitive ectoderm (D'Amour et al., 2005; Shook and Keller, 2003). It
lP
was found that disruption in WNT or TGFβ signaling pathway prevented the appearance of the primitive streak, mesoderm or definitive endoderm (D'Amour et al., 2005; Haegel et al., 1995;
na
Liu et al., 1999). Using this knowledge, various methods were developed to edit the molecules involved in signaling pathways to direct differentiation of stem cells and form organoids using
ur
Matrigel as the scaffold (D'Amour et al., 2005; McCracken et al., 2014; Spence et al., 2011).
Jo
The growth and development of organoids produced from PSC approach is difficult and they also do not proliferate indefinitely hence researchers try and transfer it to ASCs approach whenever possible (Spence et al., 2011; Volkner et al., 2016). The protocol developed by Spence et al. involves the formation of intestinal organoids by PSC approach. Beginning with the formation of definitive endoderm by directed differentiation, a method developed by D'Amour et al. (D'Amour et al., 2005). Spence et al. used a three-day Activin A protocol (Spence et al., 2011). For hindgut differentiation, the definitive endoderm cells were incubated for 4 days in 2% FBS-DMEM/F12 media containing FGF4 and Wnt3a (Spence et al., 2011). 3D floating spheroids were formed by treatment with growth factors followed by further culturing the intestinal organoids using a crypt isolation protocol as described in (Sato et al., 2009). They further modified the process by manipulating a series of growth factors to re-create the
Journal Pre-proof embryonic intestinal development in vitro (Spence et al., 2011). Similarly generating intestinal organoids from human PSCs involves the formation of definitive endoderm by a directed differentiation method as described by D'Amour et al. in 2005 (Gao et al., 2014). This is followed by floating spheroid formation, which is then embedded into Matrigel leading to the formation of the intestinal organoid using the intestinal culture system demonstrated by Sato et al. in 2009. Yet another successful work is production of retinal organoids by Völkner et al., where after culturing human ESCs and embedding it in Matrigel, a neuroepithelium trisection protocol to retinal organoid (Volkner et al., 2016). All the organoids produced from pluripotent
of
stem cells ( ESCs & iPSCs) are as follows (Huch and Koo, 2015): organoids from endoderm
ro
include intestinal (Spence et al., 2011) ,liver (Takebe et al., 2013), gastric (McCracken et al., 2014), lung (Dye et al., 2015) and thyroid (Antonica et al., 2012) organoids from ectoderm-
-p
cerebral organoids (Lancaster et al., 2013; Muguruma et al., 2015), inner ear (Koehler et al.,
re
2013) and retina organoids (Eiraku et al., 2011; Eiraku et al., 2008; Volkner et al., 2016).
lP
3. Current methods for the generation of organoids This section focuses on the common methodologies employed in the generation of organoids
na
(Figure 1, Table 1). The advantages and disadvantages of each method and considerations for organoids manufacturing are discussed. Understanding of the limitations of these methods for an
ur
organoid generation will help researchers to design more refined and reliable processes for manufacturing organoids at a larger scale. All things considered, the current organoid culture
Jo
systems can be broadly be categorized into four main categories (i) suspension culture system for organoids, (ii) organoid formation using ECM (Matrigel) scaffolds (iii) methods using spinning bioreactors, and (iv) air-liquid interface (ALI) methods. 3D Culture System for Organoids 3.1. Suspension culture system for organoids Morizane et al. developed an easy and efficient monolayer culture protocol to mass-produce kidney organoids within 12 days from nephron progenitor cells (hPSCs to NPSCs differentiation takes 7-9 days) by using 96 well round bottom ultra-low attachment plates and chemically defined compounds (Figure 2). The main advantage of this method is that this protocol can be
Journal Pre-proof used to produce both 2D and 3D kidney organoids. However, for detailed study of disease and phenotype 3D system cultured organoids are recommended (Morizane and Bonventre, 2017). This protocol allows the researchers to examine the cells of the kidney organoid at each stage of differentiation, thereby greatly widening the future possibilities of studying inherited kidney diseases (Morizane and Bonventre, 2017). Daquinag et al. used a magnetic levitation method using nanoparticle assembly to create 3D white adipose tissue growth and development in organoids (Daquinag et al., 2012). Tseng et al. first demonstrated the assembly of 3D co-culture structure of bronchiole using four cell types:
of
fibroblast cells, endothelial cells, epithelial cells and smooth muscle cells using magnetic
ro
levitation (Tseng et al., 2013). Ferreira et al. demonstrated the production of saliva-secreting organoids by a magnetic levitation method and used it to repair salivary gland hypofunction
-p
(Ferreira et al., 2019). A commercially available magnetic levitation based culture system has been used to culture 3D organoids using nanoparticles in 24 well and 96 well bio-assembler
lP
re
plates (Souza et al., 2010)
In magnetic levitation approaches, cells are cultured close to confluence followed by a
na
detachment of cells from the bottom of the culture flask by trypsinization. The cells are counted and added into 24 well or 96 well plates. The cell suspension is diluted if needed. It is then treated with Nanoshuttle-PLTM reagent by adding and dispersing the Nanoshuttle-PLTM reagent
ur
over the cells, thereby introducing biocompatible magnetic nanoparticles to cells. Culture
Jo
Isolation lid is placed on top of the plate along with a magnetic drive assembly lid containing Nanoshuttle-PLTM treated cells. The bio-assembler is then placed in the incubator. Within a very short time the cells get levitated to the air-liquid interface and get concentrated under the magnets leading to the development of 3D cellular structure (Souza et al., 2010).
Advantages of magnetic cell levitation culture system: Prompt cell-cell interactions and development of 3D structures that mirror the in vivo tissue representation. Magnetic Bio-assemblers are compatible with primary and stem cells, different media recipes, standard culturing protocols, and diagnostic techniques.
Journal Pre-proof 3.2 Organoid formation using ECM (Matrigel) scaffolds 3.2.1 Crypt isolation method for organoid production Most of the protocols to produced organoids from adult stem cells involves this method. Clevers and team (Sato et al., 2009) were the first to optimize an efficient culture system to generate intestinal organoids using Matrigel as the extracellular matrix (ECM) from adult Lgr5 stem cells (Figure 3 & Figure 3A). This intestinal organoid culture system is commonly called the RSpondin Method (Fatehullah et al., 2016; van de Wetering et al., 2015). Matrigel is the trade
of
name of the gelatinous protein mix produced by the sarcoma cells of Engelbreth-Holm-Swarm
ro
(EHS) mouse which resembles the ECM surrounding most living cells in vivo, used by researchers as ECM support for the in vitro culturing of cells (Benton et al., 2009; Benton et al.,
-p
2011; Hughes et al.). The development of the intestinal culture system was a major breakthrough
re
in the field of stem cells. Sato et al. demonstrated that a single Lgr5 stem cell present at the tip of the intestinal crypt can self-organize into a crypt-villus structure and form intestinal organoids
lP
without the support of mesenchymal tissue. Crypts are the deep pit that extends inwards into the connective tissue at the margins of the small intestine.
na
In this procedure, isolated crypts from the small intestine of mice are incubated for 30 min at 4℃ in PBS solution with EDTA. Polymerization of crypts with Matrigel is a major step in this After
ur
procedure. Crypts are then mixed with Matrigel and plated in 24 well plates.
polymerization of Matrigel crypt culture medium, Advanced DMEM/F12 from Invitrogen is
Jo
added. The crypt culture medium, Advanced DMEM/F12 contains growth factors R-spondin 1, EGF, and Noggin (Sato et al., 2009). Organoids obtained can be preserved for a long duration by freezing in liquid nitrogen and then storing at -80℃ (Xue and Shah, 2013).
Sato et al. further modified the culture medium in this protocol to produce long-term expanding intestinal epithelial organoids from human colon, adenoma and adenocarcinoma (Sato, T. et al., 2011). The signaling molecules such as Wnt, TGF-⍺, EGF and Notch ligand Dll4 produced by the CD45+ Paneth cells have a significant role in the maintenance of the Lgr5 stem cells, emphasizing the significant role of co-culturing Paneth cells with Lgr5 stem cells for improved organoid production (Sato et al., 2010). In 2016, O’Rourke et al. optimized a protocol adapted
Journal Pre-proof from the crypt isolation protocol of Sato et al., 2009 and used it to isolate, culture and maintain mouse intestinal stem cells (O'Rourke et al., 2016).
Advantages and limitation of the crypt isolation method Most of the method using Matrigel as a scaffold for organoid formation use protocols that are modified versions of crypt isolation organoid culture system by Sato et al. The major advantage of this method is its simplicity (Sato et al., 2009) and it is also an established long-term culture system for organoid culture (Sato et al., 2009). Secondly, this method can be used to produce
of
organoids from multiple sources such as adult tissue (Sato et al., 2009), fetal tissue, ESCs, and
ro
iPSCs (Fatehullah et al., 2016).
-p
The primary issue using this method is the difficulty in recapitulating the in vivo signaling and growth factor gradients in the Matrigel matrix. The possible solutions to this limitation are to
re
create a concentration gradient using microfluidic technologies (Fatehullah et al., 2016).
lP
Secondly, since organoids produced using this method depend on Matrigel which is mouse sarcoma produced ECM, these organoids are not compatible with human organ transplantation
na
(Fatehullah et al., 2016).
Jo
ur
3.3. Organoid formation via embryoid bodies formation and agitation using spinning bioreactor The embryoid bodies formation has been mainly used to produce cerebral and retinal organoids (Figure 4). Recreating human brain disorders in animal models have always been difficult. Lancaster et al. tried to generate human cerebral organoid as a disease model for neural disordermicrocephaly and check if the patient-derived cerebral organoid could act as an alternative model for animal models to study neurodevelopmental disorders (Lancaster et al., 2013). Lancaster et al. reprogrammed skin fibroblast cells of a patient with microcephaly using lentiviral delivery of the four major reprogramming factors, Oct 4, Sox2, Klf4 and c-Myc to produce induced pluripotent stem cells (Lancaster et al., 2013). They succeeded in optimizing a cerebral organoid culture system to produce both mouse and human cerebral organoids in vitro from pluripotent stem cells. When pluripotent stem cells form organoids, cells form aggregates
Journal Pre-proof in some cases as an intermediate stage. These 3D cell aggregates of pluripotent stem cells are called embryoid bodies, observed especially in the formation process of cerebral organoids. The human cerebral organoids produced emulate earlier stages of the human brain. Mitomycin C growth inactivated MEFs were used to culture mouse ESCs and passaged using standard protocols. As described by Eiraku et al. the cells were then trypsinized and plated in each well of an ultra-low binding 96 well plate in a differentiation medium(Eiraku et al., 2008). Embryoid bodies formed using this protocol (Koike et al., 2005) were then transferred into neural induction medium on day 5, the neuroectodermal tissues were then embedded in Matrigel droplets on day
of
11 and transferred to the spinning bioreactor after 4 days of stationary growth inside Matrigel
ro
(Lancaster et al., 2013). ESCs of the mouse which, in contrast to those from human divide at a faster rate, were transferred to neural induction medium on day 4, embedded in Matrigel droplets
-p
on day 6, and on day 9 transferred to the spinning bioreactor (Lancaster et al., 2013). Lancaster et al. published a detailed cerebral organoid protocol and discussed the challenges faced during
re
the process (Lancaster, M. A. and Knoblich, J. A., 2014).
lP
Quadrato et al. in 2017 further modified the protocol developed by Lancaster et al. and demonstrated the possibility of developing cerebral organoids with mature features for extended
na
periods by controlling the neuronal activity within the organoids. This can be done using light stimulation of photosensitive cells (Quadrato et al., 2017). Wang et al. developed a protocol
ur
combining both stem cell and organ-on-a-chip technology in 2018 (Wang et al., 2018). Recent studies show improvement in producing mature cerebral organoids (Giandomenico et al., 2018;
Jo
Quadrato et al., 2017). DiStefano et al. used a rotating wall vessel bioreactor to produce 3D retinal organoids (DiStefano et al., 2018). Cerebral organoids (Giandomenico et al., 2018; Lancaster et al., 2013; Quadrato et al., 2017) and retinal organoids (DiStefano et al., 2018) are produced using this method that involves embryoid body formation in the process. Advantages and limitation of the embryoid body method The use of a spinning bioreactor in addition to Matrigel scaffold tackles one of the major challenges in organoid manufacturing, namely optimal nutrient and oxygen supply to the cell in the organoid. This is achieved by the regular supply of media by the spinning bioreactor (Lancaster et al., 2013). Moreover, cerebral organoids (Lancaster et al., 2013) produced using the protocol optimized by Lancaster et al. can be stored up to over 1 year (15 months) in the
Journal Pre-proof spinning bioreactor, though the organoids start to shrink after 6-7 months (Lancaster et al., 2013). The cerebral organoids produced using this protocol not only exhibit the rudimentary mammalian neural development but epitomize human brain development (Lancaster et al., 2013). Furthermore, Lancaster et al. succeeded in modelling these cerebral organoids with the disorder “microcephaly” from patient-derived iPSCs, which is advantageous because this particular disorder has been extremely difficult to model in mice (Lancaster et al., 2013). The major issue using the “embryoid bodies” based approach is the lack of vascularization (Ling et al., 2019). Addition of a spinning bioreactor to the production process to ensure constant
of
supply of media so as to meet the need of nutrient and oxygen of the cells in the organoids has
ro
helped to overcome this problem but only up to an organoid size of approximately 4mm (in diameter) (Lancaster, M. A. and Knoblich, J. A., 2014). The cerebral organoids produced using
-p
this method mimic the initial human brain development but fails to show tissue maturation and
re
complexity as seen in the adult brain (Lancaster et al., 2013). Secondly, the neural and nonneural tissue cross-talk is limited in the organoid as it lacks the surrounding embryonic tissues
lP
(Lancaster, M. A. and Knoblich, J. A., 2014). Organoid size is another limitation in this method. The organoids can only reach up to a size of 4mm in diameter due to the lack of nutrient
na
exchange and limited oxygen exchange at the center of the organoids (Lancaster et al., 2013)
ur
3.4 Air liquid interface (ALI) methods for organoid culture Air liquid interface is a method in which the top layer of the cultured cells is exposed to air, and
Jo
the basal surface is in contact with the liquid medium (Figure 5). The organoids are cultured in a gel and are directly exposed to air instead of submerged in a culture media. Tado et al. first used the air- liquid interface method on thyroid tissue in 2002 (Toda et al., 2002). Inspired by previous work, Ootani et al. developed a protocol to produce intestinal organoids (Ootani et al., 2009). Studies by Fatimah et al. revealed that the air liquid interface method could be used to produce human amnion epithelial cells (HAECs) as it stimulates an early differentiation of HAECs. They demonstrated its potential to be used as a substitute for skin regeneration (Fatimah et al., 2013). In 2014, Takasato et al. optimized a protocol using air liquid interface method to direct the process of embryonic stem cell differentiation into formation of a self-organizing structure including formation of nephron in vitro, by synchronizing the induction of the progenitors for both the collecting duct (UB-Ureteric bud) and nephron (MM-Intermediate
Journal Pre-proof mesoderm-derived metanephric mesenchyme) (Takasato et al., 2014). Takasato et al. generated kidney organoids that matched the first trimester human kidney with each kidney organoid containing about 500 nephrons (Takasato and Little, 2015). A detailed protocol that simultaneously induces all four progenitors (nephron progenitors, endothelial progenitors, ureteric epithelial progenitors and renal interstitial progenitors) to generate human kidney organoids from human pluripotent stem cells was developed in 2016 (Takasato et al., 2016). This is considered to be a major landmark in the research on kidney organoids because previously it was a major challenge to induce all four progenitors simultaneously. Li et al. demonstrated that
of
a single air liquid interface could model several gastrointestinal malignancies from stomach,
ro
colon and pancreas in mesenchymal and primary epithelial organoid culture without the need for any additional growth factors. They also extended their study to successfully produce embryonic
-p
pancreatic organoids within a short period of time (Li, X. et al., 2014). Li et al. optimized a collagen-based ALI long-term culture methodology to expand gastrointestinal epithelium tissue
re
and produce intestinal organoids. Usui et al. produced normal and tumor colorectal tissues from
lP
human patient tissues using ALI organoid culture system (Usui et al., 2018). Takasato et al. used an air liquid interface method to produce kidney organoids (Takasato et al.,
na
2016). Calvin Kuo lab, Stanford University used the ALI method to produce gastric organoids (Li et al., 2016), intestinal organoids (Ootani et al., 2009), and pancreatic organoids. This method has also allowed them to produce cancer organoids. ALI method is currently mainly used to
ur
produce kidney organoid (Rossi et al., 2018), cerebral organoid (Giandomenico et al., 2018),
Jo
lung organoid (Bals et al., 2004; Fessart et al., 2013; Pageau et al., 2011; Vaughan et al., 2006) and skin organoids (Fatimah et al., 2013).
Advantages and limitations of ALI method The main advantage of the ALI organoids is that they accurately replicate the original organ in terms of physiology, multilineage differentiation and organ structure (Usui et al., 2018). Interfollicular epidermal cells grow on the basement membrane and are exposed to air in vivo, using this knowledge Pruniéras et al. cultivated keratinocytes with a single air liquid interface (Pruniéras et al., 1983). Later it was found that organoids produced using ALI culture method were more precise as it accurately recreates the mode of cell growth that occurs in vivo as this leads to a complete differentiation (Pruniéras et al., 1983; Usui et al., 2018). Cell-stimulant
Journal Pre-proof interactions and cell-cell interactions are better in cells cultured using ALI compared to submerged cell culture systems (Upadhyay and Palmberg, 2018). ALI (Ootani et al., 2009) enables enhanced oxygen transport from both air-collagen interface(top) and semi-permeable membrane interface (bottom) resulting in a higher oxygen concentration within the system compared to the submerged culture system (Sato et al., 2009) which only allows oxygen transport from the top layer of the submerged culture media (DiMarco et al., 2014). A study by DiMarco et al. also showed that oxygen gradient within submerged cultures was 1.5 times higher than that in ALI culture system (DiMarco et al., 2014). This improved oxygenation in the ALI
of
organoid cultures helps preserve the organoids for a longer period of time (Ootani et al., 2009)
ro
and also accelerates self organization in organoids (Sekiya et al., 2019). Culturing cells (keratinocytes) first under submerged conditions for 7 days to form the epidermal layer
-p
(monolayer) followed by culturing the cells in ALI culture system to further promote progressive differentiation has proven to be efficacious even in the process of 3D bioprinting of human skin
re
which resulted in the formation of a multi-layered 3D skin construct (Epidermis, dermis and
lP
stratum corneum) (Lee et al., 2013). Microfluidic device “human lung-on-chip” with integrated ALI culture system provides improved ways to study drug efficacy and toxicity(Huh et al.,
na
2011). ALI method has also been effective in creating patient-derived immunotherapy organoid models that retain its native tumour microenvironment (Neal et al., 2018)
.This is a major
breakthrough as the previous co-culture methods to emulate tumour microenvironment by
ur
combining tumour epithelia with additional cellular components such as tumour infiltrating
Jo
lymphocytes were extremely elusive (Dart, 2019; Neal et al., 2018). Though ALI method has many advantages over other culture systems, it is not without challenges- Despite the successful production of kidney organoids by Takasato et al., they failed to produce kidney organoids that mature to the adult stage with ALI method (Takasato et al., 2016). ALI culture system is used to culture mainly epithelial cells and to develop organoids containing epithelial cells. Use of epithelial cells for culture in ALI culture system is challenging as there are chances of contamination by microbial or fibroblast cells. Also, use of primary epithelial cells from animals for maintaining the culture in ALI system involves sacrifice of a myriad number of animals compared to methods that use an already established cell line (Delgado-Ortega et al., 2014). Recently cerebral organoids (Giandomenico et al., 2018) were developed using ALI method, this opens up the use of this culture system to develop organoids of other cell type as well. However,
Journal Pre-proof currently most of the organoids developed so far using this culture system are those developed from epithelial cell cultures. 3.5 Recent advances in organoid research- 2D Monolayer culture systems of organoids
Cells in 2D culture systems grow by adhering to the base of the culture flask, petri dish or transwells, mostly growing into monolayer cultures (Sun et al., 2006). In 2018, Thorne et al. demonstrated that dissociated 3D organoids (formed using crypt isolation culture system demonstrated by Sato et al. (Sato et al., 2009)) can produce 2D enteroid monolayer (Thorne et
of
al., 2018). They used 96 well plates coated with thin layered Matrigel to optimize a scalable
ro
method to culture 2D enteroid (or organoids recapitulating the small intestine (Foulke-Abel et al., 2016)) monolayers that emulates some of the important features of 3D organoids as well as in
-p
vivo conditions of an organ (Braverman and Yilmaz, 2018; Thorne et al., 2018). 2D Monolayer culture of organoids has a wide variety of applications such as intestinal epithelial
re
barrier models to study the dynamics of host-microbe interactions, as well as molecular
lP
mechanisms involved in differentiation and function of different types of IECs (Intestinal Epithelial cells) (Nakamura, 2019). These monolayer cultures also permit high throughput
na
microscopy for further study of the organoids (Thorne et al., 2018). A similar study by Hee et al. generated 2D monolayer cultures by enzymatic dissociation of 3D porcine intestinal organoids (van der Hee et al., 2018). In this study, 5-day spheroid cultures
ur
were disrupted into a single cell suspension and plated in Matrigel coated culture plates or
Jo
transwells to produce monolayers (van der Hee et al., 2018). This study also demonstrated that cell seeding densities directly affected the capacity to form a monolayer (van der Hee et al., 2018). Furthermore, Liu et al. established a 2D culture system to support self-sustaining of Lgr5 intestinal stem cells monolayer. It was also found that 10μm thick Matrigel greatly supported Lgr5 ISCs (Intestinal Stem Cells) monolayer culture while thicker Matrigel layers (50 μm) supported 3D organoid development (Liu et al., 2018). Though one major limitation of using 2D monolayer cell culture of organoids for studies is the difficulty in passaging (Braverman and Yilmaz, 2018).
Journal Pre-proof 4. Challenges in organoids manufacturing and possible answers Organoids are a thriving topic of research with numerous clinical and pre-clinical applications, with the scientific community exploring ways to overcome issues that currently limit their full potential. One of the biggest challenges to organoid manufacturing is its limited reproducibility, which possesses a major hurdle if organoids are to be used for toxicity testing or another high throughput testing in future (Kelava and Lancaster; Lancaster et al., 2013). Organoid production scalability, batch to batch variation, cellular composition and architecture of organoids are all essential factors that affect the reproducibility of data obtained with organoids (Nugraha et al.,
of
2019). In a recent study, Krefft et al. succeeded in generating a standard procedure to produce
sample to sample variability (Krefft et al., 2018).
ro
reproducible forebrain type cerebral organoids from human iPSCs which demonstrated improved
-p
Another important limitation is non-vascularization (Kaushik et al., 2018). Issues of
re
vascularization could be potentially solved by either using a spinning bioreactor or by coculturing with endothelial cells (Kaushik et al., 2018). Insufficient supply of nutrient and oxygen
lP
exchange to the center of the organoids is the reason why most healthy tissues are at the periphery of the organoid, affecting the overall tissue patterning(Kelava and Lancaster; Lancaster
na
et al., 2013). To improve the tissue vasculature, Allenby et al. fabricated ceramic hollow fiber constructs in 2018 (Allenby et al., 2018). Mansour et al. remarkably succeeded in producing
ur
vascularized human brain organoids in the mouse brain in vivo (Mansour et al., 2018). This was a major breakthrough as it eliminates most of the challenges faced due to limited vascularization
Jo
of organoids (Mansour et al., 2018). This technique of cerebral organoids transplantation into mouse brain method definitely eliminates some of the problems/limitations faced by the in vitro culture systems (Lancaster, 2018), but faces ethical issues. A third major hurdle for manufacturing of organoids is tissue maturation (Kaushik et al., 2018). In most cases, early stages of development and organization in the organoids are well developed, but the tissues fail to evolve into a functionally developed and mature organ (Kaushik et al., 2018). Albeit, studies show that intestinal organoid produces Lgr5 stem cells that indicate further maturation (Sato et al., 2009). Giandomenico et al. have successfully optimized an efficient protocol for long-term production of mature cerebral organoids using ALI (Giandomenico et al.,
Journal Pre-proof 2018). In the case of generation of cardiac organoids, the role of maturity of the cardiomyocytes cells used in the co-culture is yet to be discovered (Nugraha et al., 2019). The absence of continuous anterior-posterior or dorsal-ventral axis that guide and support proper organoid directionality which results in the heterogeneity of organoids produced is another confounding issue, especially for cerebral organoid, (Kelava and Lancaster). Various parts of the cerebral organoid are randomly arranged when compared to the actual brain organ (Bagley et al., 2017). Knoblich and his team have been working on solving this limitation via two approaches: (a) by combining bioengineering techniques along with organoids to rectify the positioning of
of
dorsal-ventral axis and anterior-posterior axis of the brain using biocompatible bioengineered
ro
fibers (PLGA fibers) poly (lactic-co-glycolic acid) and (b) by producing each distinct regions of the brain separately in vitro and recapitulating the exact order in the actual brain. Bagley et al.
-p
succeeded in producing the dorsal-ventral axis of organoids by fusing organoid identities of
re
dorsal and ventral forebrain, emphasizing on the significance of such fused cerebral organoid cultures in modeling complex interactions between different parts of the brain (Bagley et al.,
lP
2017). The most recent studies showed successful assembling of brain organoids by rectifying the dorsal-ventral axis and posterior-anterior axis positioning. These so-called “assembloids” are
na
considered to be the next generation brain organoids (Paşca, 2019). Biological complexity is another challenge for organoid manufacturing. Tumors in our body
ur
display complex genetic heterogeneity. Tumoroids are created in vitro to resemble the original
Jo
tumors, although the extent to which the tumoroids hold the complexity and possible changes of the original tumors is still unclear (Dutta et al., 2017). Organoids produced using rigid extracellular matrices (ECM) are limited in their usage in drug screening as the ECM can hamper drug penetration (Fatehullah et al., 2016). One of the major limitations of organoids is the lack of blood cells, stroma and immune cells (Drost and Clevers, 2018). Bioengineering techniques are being used to incorporate additional cellular elements to overcome this problem (Yin et al., 2016). Yet another success in research is the immunotherapy models (patient-derived organoid models) that retain the tumour microenvironment produced by ALI method. This could open up great possibilities for in vitro modelling in personalised immunotherapy for cancer research (Dart, 2019).
Journal Pre-proof Organoids produced using Matrigel (which is a mouse sarcoma cell product) as the extracellular matrix was observed to cause compatibility issues in humans restricting the use of organoids in human organ transplantation (Fatehullah et al., 2016). Currently, research focusing on the development of a better defined ECM that abide by human regulations of organ transplantations are being carried out (Fatehullah et al., 2016). Furthermore, organoid production relying on embedding the stem cells into Matrigel restricts the surface to mass ratio, thereby limiting the manufacturing of organoids in a large scale (Lu et al., 2017). Lu et al. demonstrated a new capsule-based organoid production method that overcomes this limitation and produces
of
organoids at a large scale with a better cell recovery (Lu et al., 2017). These capsules are
ro
produced using a two fluidic electrostatic co-spraying method and the core of the capsule consists of Matrigel that supports the growth of the organoids.
-p
Size of organoids is a crucial factor that restricts organoid technology from reaching its full potential and limiting their manufacturing. The maximum distance that oxygen and nutrients can
re
diffuse inside the organoid determines the size of the organoids in culture (Akkerman, 2017). As
lP
the size of the organoids increases, an oxygen gradient is created within the organoids which ultimately leads to limited availability of oxygen to the center of organoids causing death in the
na
central part of organoid after it reaches a particular size (Skardal et al., 2016) thereby limiting the production of organoids of larger sizes (Akkerman, 2017). Mathematical models of cerebral organoids shows that 1.43mm is the maximum attainable size of cerebral organoids (Akkerman,
ur
2017), although researchers have succeeded in generating cerebral organoids with a size up o
Jo
4mm, with some cell death been observed at the center of the organoids (Akkerman, 2017; Lancaster, M. A. and Knoblich, J. A., 2014; Lancaster et al., 2013; McMurtrey, 2016). Moreover, Takasato et al. managed in generating larger sized kidney organoids (up to 6mm in diameter) by using an ALI method. (Takasato et al., 2016). These are exceptional cases with an average organoid size of 3mm commonly being obtained (Akkerman, 2017; Eicher et al., 2018). Recently, a very intriguing research carried out by Dr Alysson Muotri and team, where they developed a pea sized Neanderthal brain organoid. These Neanderthal organoids unlike its human organoid counterpart differs in size , appearance and neuronal activity (Cohen, 2018). In order to consider large-scale manufacturing of organoids, the standardization of size may be crucial (Eicher et al., 2018). Organoid imaging is an additional issue faced by researchers which
Journal Pre-proof are being overcome with deeper imaging and high throughput screening imaging techniques especially in the case of cardiac organoids (Nugraha et al., 2019).
5. Technologies to improve organoids manufacturing Combining bioengineering tools with 3D cell culture techniques definitely help in improving the size of the organoids formed and eliminates a number of challenges currently faced (SaglamMetiner et al., 2019). Factors to be considered to improve manufacturing of organoids are as follows (Figure 6): (a) scaffolds (b) regulating culture parameters such as oxygen distribution,
of
cell-cell interaction, pH level etc. (c) spatiotemporal control of signaling and (d) efficient
ro
vascularization.
-p
Scaffolds: Most of the organoid culture system uses scaffolds made of Matrigel. But as described earlier Matrigel is incompatible with humans hence hinders with transplantation into humans.
re
Alternative functional scaffolds made of biomolecules such as basic fibroblast growth factors
lP
(BFGFs), vascular endothelial growth factors (VEGFs) and platelet-derived growth factors (PDGFs) have improved the vascular networks of organoids produced (Yin et al., 2016). Defined
na
ECM which include alternatives such as biomimetic scaffolds made from both natural and synthetic polymers (polyethylene glycol and polyacrylamide) is being developed to overcome the issues caused due to Matrigel in organoid research (Fatehullah et al., 2016; Yin et al., 2016).
ur
Regulating culture parameters: Sensors and devices that cause minimal disturbances to the in
Jo
vivo cell environment and the natural processes can enable precise monitoring of these cell parameters (Yin et al., 2016), for example by direct in vivo measurements of oxygen tension using two-photon phosphorescence lifetime microscopy (Spencer et al.). Control over cell-cell interactions was made possible by using silicone substrate microscale devices (Hui and Bhatia, 2007). Microfluidic organs-on-a-a-chip also allows precise control and measurements on these important cell parameters, but they do have their own limitations (Bhatia and Ingber, 2014; Yin et al., 2016). Spatio-temporal control: The difficulty in recreating in vitro scaffolds that replicate the spatialtemporal signals to cells is the main cause of the observed difference between the organogenesis in vivo and in vitro (Yin et al., 2016). Recent advances envision ways to overcome this limitation by using light-mediated patterning to manipulate the ECM (Yin et al., 2016). Use of a more
Journal Pre-proof defined ECM as an alternative to Matrigel in organoid culture is crucial for improved spatiotemporal cues (Yin et al., 2016) and tissue regeneration applications. Efficient vascularization: Researchers have focused on several different bioengineering approaches to gain more control over the production of organoids and their behavior (Yin et al., 2016). For the proper development of functional organoids, in vitro vascularization is very important in order to provide a sufficient supply of nutrients and oxygen (Yin et al., 2016). One approach to achieve efficient vascularization is to seed the endothelial cells within the system to allow for neo-angiogenesis to occur. Alternatively, scaffold-based methods in which 3D
of
microfluidic devices are used to allow uniform flow and mass transfer could be engineered. Yet
ro
another bioengineering approach could take advantage of bioprinting to seed the channels with vascular cells (Kang et al., 2015; Zhang et al., 2016). Bioprinting could be used to generate
-p
complex multicellular structures by layering different cell types along with hydrogel-based bioinks in a 3D bioprinter (Choudhury et al., 2018b; Kang et al., 2015). Recent studies showed
re
improved kidney organoid vascularization with the application of fluid flow across the surface of
lP
partially differentiated kidney organoids attached to extracellular matrix housed within a 3D printed millifluidic chip (Allison, 2019; Homan et al., 2019). Later studies showcasing how this
na
technique could help study polycystic kidney diseases was demonstrated (Boettner, 2019, Feb 11). Lancaster et al. demonstrated the use of a spinning bioreactor to provide improved nutrient and oxygen supply during organoid manufacturing (Lancaster et al., 2013). Using cellular
ur
memory devices that are a key development in the field of synthetic biology can help store all the
Jo
transient responses of the organoids for a longer period (Purcell and Lu, 2014). Allenby et al. fabricated an efficient manufacturing tool based on ceramic hollow fiber constructs that when incorporated into 3D tissue models facilitate the separation of mononuclear cells( MNCs) and red blood cells ( RBCs) and improve tissue vasculature (Allenby et al., 2018). Merge of organoid technology & CRISPR Cas9 genome editing: Drost et al. demonstrated the deletion of key DNA repair genes of human colon organoids using CRISPR/Cas9 technology to study the origin of the characteristic mutations of cancer (Drost et al., 2017). Organoids have been developed from prostate cancer patients as in vitro prostate cancer models (Gao et al.). Attempts were made to generate cardiac organoids by combining CRISPR/Cas 9 genome editing tool and hiPSCs in order to correct the mutations that cause heart diseases and model heart diseases (Nugraha et al., 2019). To study the significance of
Journal Pre-proof hyperproliferation of colorectal cells and the pathogenesis of colorectal cancer, Crespo et al. demonstrated the idea of “disease in a dish” by generating colonic organoids using iPSCs derived from patients with familial adenomatous polyposis (Crespo et al., 2017). O’Rourke et al. succeeded in producing colorectal cancer models for future genetic and pre-clinical studies through the orthotopic engraftment of colon organoids (O'Rourke et al., 2017). The merge of organoid technology and CRISPR-Cas 9 genome editing has been a remarkable milestone in the field of cancer research as well as study of other disease and definitely adds value to research on organoids (Driehuis and Clevers, 2017; Lokody, 2013; Matano et al., 2015; Nie and Hashino,
ro
of
2017; Schwank et al., 2013; Wang et al., 2017) (Kim et al., 2019).
6. Conclusion
-p
In this review, we have deliberated on the main methodologies to fabricate organoids. Each of the methods has their own advantages and limitations. We have discussed some key protocols
re
involved in the production of organoids and broadly classified them into four main categories (i)
lP
suspension culture system (ii) Systems using ECM (Matrigel) scaffolds, (iii) methods using spinning bioreactors and (iv) Air liquid interface (ALI) methods. ALI (3D culture system) can be
na
integrated with various bioengineering technique such as bioprinting (Choudhury et al., 2018a) and microfabricated devices such as microfluidic organ-on-chip devices (Yu and Choudhury, 2019; Yu et al., 2019). Microfluidic organoid-on-a-chip platforms can ensure better nutrient and
ur
gaseous exchange enabling mimicking of 3D tissue architecture and physiology. Although to
Jo
increase the critical size of the organoids, greater efforts are needed for ensuring organoids vascularization; either via generating artificial blood vessels or spontaneously generating vessels by stimulating the angiogenic pathways. A more interdisciplinary approach with efforts from both biologists and bioengineers may solve most of the current limitations, making way towards manufacturing of organoids (Huh et al., 2011; Kimura et al., 2018). In future, there is a possibility that all of the methods mentioned in the paper could be used interchangeably at various stages regardless of the source to obtain the optimal organoids. Meanwhile, a non-profit organization called HUB (Hubrecht Organoid Technology) has initiated and established “Living Biobank”. It was created with an aim to collect all well-characterized organoids generated from patient tissues. Such a centralized system of organoids “Living Biobank” will be beneficial for scientific research, personalized medicine, and drug
Journal Pre-proof development. At present, organoids produced are transported by immersing the organoids in liquid nitrogen (Xue and Shah, 2013), the organoids can be stored by freezing and thawed when used later, however, this affects the electrical activity inside the organoids (Chakradhar, 2016). Much work is also needed in finding a more efficient and sophisticated organoid transportation method for large-scale transportation. There are few important organoid companies that are looking into manufacturing and commercialing organoids in a large scale. Some of these includeCellesce, Organome, DefiniGEN, Prellis Biologics and Novoheart. Currently these organoids are purchased mostly by researchers hence the scale is still limited. Future holds the key to large
-p
ro
of
scale commercial organoid production.
Acknowledgments
lP
re
We thank Dr Chen Sixun and Dr Wang Tianyi for insightful comments on the manuscript.
Funding
Author Contributions
na
This work was supported by A*STAR JCO Grant 15302FG152 awarded to D.C.
Jo
ur
A.A.: manuscript writing including the figures and tables; F.Y.: manuscript writing including the figures and tables; D.C: conception and design, manuscript writing; W.H.: manuscript editing. A.A, D.C, F.Y. W.H: final approval of the manuscript.
Disclosure of potential conflict of interest The authors indicated no potential conflicts of interest.
Journal Pre-proof References
Jo
ur
na
lP
re
-p
ro
of
Akkerman, N.D., L. H., 2017. Dawn of the organoid era: 3D tissue and organ cultures revolutionize the study of development, disease, and regeneration. . Bioessays. Allenby, M.C., Tahlawi, A., Morais, J.C.F., Li, K., Panoskaltsis, N., Mantalaris, A., 2018. Ceramic Hollow Fibre Constructs for Continuous Perfusion and Cell Harvest from 3D Hematopoietic Organoids. Stem Cells Int 2018, 6230214. Allison, S.J., 2019. Fluid flow enhances vascularization and maturation of kidney organoids. Nature Reviews Nephrology. Antonica, F., Kasprzyk, D.F., Opitz, R., Iacovino, M., Liao, X.-H., Dumitrescu, A.M., Refetoff, S., Peremans, K., Manto, M., Kyba, M., Costagliola, S., 2012. Generation of functional thyroid from embryonic stem cells. Nature 491, 66. Arora, N., Imran Alsous, J., Guggenheim, J.W., Mak, M., Munera, J., Wells, J.M., Kamm, R.D., Asada, H.H., Shvartsman, S.Y., Griffith, L.G., 2017. A process engineering approach to increase organoid yield. Development 144(6), 1128-1136. Bagley, J.A., Reumann, D., Bian, S., Levi-Strauss, J., Knoblich, J.A., 2017. Fused cerebral organoids model interactions between brain regions. Nat Methods 14(7), 743-751. Bals, R., Beisswenger, C., Blouquit, S., Chinet, T., 2004. Isolation and air-liquid interface culture of human large airway and bronchiolar epithelial cells. J Cyst Fibros 3 Suppl 2, 49-51. Barker, N., Huch, M., Kujala, P., van de Wetering, M., Snippert, H.J., van Es, J.H., Sato, T., Stange, D.E., Begthel, H., van den Born, M., Danenberg, E., van den Brink, S., Korving, J., Abo, A., Peters, P.J., Wright, N., Poulsom, R., Clevers, H., 2010. Lgr5+ve Stem Cells Drive SelfRenewal in the Stomach and Build Long-Lived Gastric Units In Vitro. Cell Stem Cell 6(1), 25-36. Bartfeld, S., Bayram, T., van de Wetering, M., Huch, M., Begthel, H., Kujala, P., Vries, R., Peters, P.J., Clevers, H., 2015. In Vitro Expansion of Human Gastric Epithelial Stem Cells and Their Responses to Bacterial Infection. Gastroenterology 148(1), 126-136.e126. Bartfeld, S., Clevers, H., 2015. Organoids as Model for Infectious Diseases: Culture of Human and Murine Stomach Organoids and Microinjection of Helicobacter Pylori. Journal of visualized experiments : JoVE(105), 53359. Bartfeld, S., Clevers, H., 2017. Stem cell-derived organoids and their application for medical research and patient treatment. Journal of Molecular Medicine(1432-1440 (Electronic)). Bello, S., 2019, Sep 26. Three-Organoid System Grown in Lab for the First Time. https://edgy.app/three-organoid-system-grown-in-lab-for-the-first-time?pfrom=science&fp=a6. Benton, G., George J Fau - Kleinman, H.K., Kleinman Hk Fau - Arnaoutova, I.P., Arnaoutova, I.P., 2009. Advancing science and technology via 3D culture on basement membrane matrix. Journal of Cellular Physiology(1097-4652 (Electronic)). Benton, G., Kleinman Hk Fau - George, J., George J Fau - Arnaoutova, I., Arnaoutova, I., 2011. Multiple uses of basement membrane-like matrix (BME/Matrigel) in vitro and in vivo with cancer cells. International Journal of Cancer(1097-0215 (Electronic)). Bhatia, S.N., Ingber, D.E., 2014. Microfluidic organs-on-chips. Nature Biotechnology 32, 760. Boettner, B., 2019, Feb 11. Engineered mini-kidneys come of age. https://news.harvard.edu/gazette/story/2019/02/engineered-miniature-kidneys-come-of-age/. Boucherie, C., Mukherjee S Fau - Henckaerts, E., Henckaerts E Fau - Thrasher, A.J., Thrasher Aj Fau - Sowden, J.C., Sowden Jc Fau - Ali, R.R., Ali, R.R., Brief report: self-organizing neuroepithelium from human pluripotent stem cells facilitates derivation of photoreceptors. (1549-4918 (Electronic)). Braverman, J., Yilmaz, O.H., 2018. From 3D Organoids back to 2D Enteroids. Dev Cell 44(5), 533-534. Broutier, L., Andersson-Rolf, A., Hindley, C.J., Boj, S.F., Clevers, H., Koo, B.K., Huch, M., 2016. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat Protoc 11(9), 1724-1743.
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Broutier, L., Mastrogiovanni, G., Verstegen, M.M.A., Francies, H.E., Gavarró, L.M., Bradshaw, C.R., Allen, G.E., Arnes-Benito, R., Sidorova, O., Gaspersz, M.P., Georgakopoulos, N., Koo, B.K., Dietmann, S., Davies, S.E., Praseedom, R.K., Lieshout, R., Ijzermans, J.N.M., Wigmore, S.J., Saeb-Parsy, K., Garnett, M.J., van der Laan, L.J.W., Huch, M., 2017. Human primary liver cancer–derived organoid cultures for disease modeling and drug screening. Nature Medicine 23, 1424. Campo, M.S., 2002. Animal models of papillomavirus pathogenesis. Virus Research 89(2), 249261. Castellanos-Gonzalez, A., Cabada, M.M., Nichols, J., Gomez, G., White, A.C., 2013. Human Primary Intestinal Epithelial Cells as an Improved
In Vitro Model for
Cryptosporidium parvum Infection. Infection and Immunity 81(6), 1996. Chakradhar, S., 2016. New company aims to broaden researchers' access to organoids. Nature Medicine 22, 338. Choudhury, D., Anand, S., Naing, M.W., 2018a. The Arrival of Commercial Bioprinters Towards 3D Bioprinting Revolution! International Journal of Bioprinting 4(2). Choudhury, D., Tun, H.W., Wang, T., Naing, M.W., 2018b. Organ-Derived Decellularized Extracellular Matrix: A Game Changer for Bioink Manufacturing? Trends in Biotechnology 36(8), 787-805. Cohen, J., 2018. Neanderthal brain organoids come to life. Science 360(6395), 1284. Costa, E.C., Moreira, A.F., de Melo-Diogo, D., Gaspar, V.M., Carvalho, M.P., Correia, I.J., 2016. 3D tumor spheroids: an overview on the tools and techniques used for their analysis. Biotechnol Adv 34(8), 1427-1441. Crespo, M., Vilar, E., Tsai, S.Y., Chang, K., Amin, S., Srinivasan, T., Zhang, T., Pipalia, N.H., Chen, H.J., Witherspoon, M., Gordillo, M., Xiang, J.Z., Maxfield, F.R., Lipkin, S., Evans, T., Chen, S., 2017. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing. Nat Med 23(7), 878-884. Cugola, F.R., Fernandes, I.R., Russo, F.B., Freitas, B.C., Dias, J.L., Guimaraes, K.P., Benazzato, C., Almeida, N., Pignatari, G.C., Romero, S., Polonio, C.M., Cunha, I., Freitas, C.L., Brandao, W.N., Rossato, C., Andrade, D.G., Faria Dde, P., Garcez, A.T., Buchpigel, C.A., Braconi, C.T., Mendes, E., Sall, A.A., Zanotto, P.M., Peron, J.P., Muotri, A.R., Beltrao-Braga, P.C., 2016. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 534(7606), 267-271. D'Amour, K.A., Agulnick, A.D., Eliazer, S., Kelly, O.G., Kroon, E., Baetge, E.E., 2005. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol 23(12), 1534-1541. Dakhore, S., Nayer, B., Hasegawa, K., 2018. Human Pluripotent Stem Cell Culture: Current Status, Challenges, and Advancement. Stem cells international 2018, 7396905-7396905. Daquinag, A.C., Souza, G.R., Kolonin, M.G., 2012. Adipose Tissue Engineering in ThreeDimensional Levitation Tissue Culture System Based on Magnetic Nanoparticles. Tissue Engineering Part C: Methods 19(5), 336-344. Dart, A., 2019. Organoid 2.0. Nature Reviews Cancer 19(3), 126-127. Dekkers, J.F., Wiegerinck, C.L., de Jonge, H.R., Bronsveld, I., Janssens, H.M., de Winter-de Groot, K.M., Brandsma, A.M., de Jong, N.W., Bijvelds, M.J., Scholte, B.J., Nieuwenhuis, E.E., van den Brink, S., Clevers, H., van der Ent, C.K., Middendorp, S., Beekman, J.M., 2013. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med 19(7), 939945. Delgado-Ortega, M., Olivier, M., Sizaret, P.-Y., Simon, G., Meurens, F., 2014. Newborn pig trachea cell line cultured in air-liquid interface conditions allows a partial in vitro representation of the porcine upper airway tissue. BMC Cell Biol 15, 14-14.
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
DeRose, Y.S., Gligorich, K.M., Wang, G., Georgelas, A., Bowman, P., Courdy, S.J., Welm, A.L., Welm, B.E., 2013. Patient-derived models of human breast cancer: protocols for in vitro and in vivo applications in tumor biology and translational medicine. Curr Protoc Pharmacol Chapter 14, Unit14.23-Unit14.23. DiMarco, R.L., Su, J., Yan, K.S., Dewi, R., Kuo, C.J., Heilshorn, S.C., 2014. Engineering of three-dimensional microenvironments to promote contractile behavior in primary intestinal organoids. Integrative biology : quantitative biosciences from nano to macro 6(2), 127-142. DiStefano, T., Chen, H.Y., Panebianco, C., Kaya, K.D., Brooks, M.J., Gieser, L., Morgan, N.Y., Pohida, T., Swaroop, A., 2018. Accelerated and Improved Differentiation of Retinal Organoids from Pluripotent Stem Cells in Rotating-Wall Vessel Bioreactors. Stem Cell Reports 10(1), 300313. Driehuis, E., Clevers, H., 2017. CRISPR/Cas 9 genome editing and its applications in organoids. American Journal of Physiology-Gastrointestinal and Liver Physiology 312(3), G257G265. Drost, J., Clevers, H., 2018. Organoids in cancer research. Nat Rev Cancer 18(7), 407-418. Drost, J., van Boxtel, R., Blokzijl, F., Mizutani, T., Sasaki, N., Sasselli, V., de Ligt, J., Behjati, S., Grolleman, J.E., van Wezel, T., Nik-Zainal, S., Kuiper, R.P., Cuppen, E., Clevers, H., 2017. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science 358(6360), 234-238. Dutta, D., Heo, I., Clevers, H., 2017. Disease Modeling in Stem Cell-Derived 3D Organoid Systems. Trends Mol Med 23(5), 393-410. Dye, B.R., Hill, D.R., Ferguson, M.A., Tsai, Y.H., Nagy, M.S., Dyal, R., Wells, J.M., Mayhew, C.N., Nattiv, R., Klein, O.D., White, E.S., Deutsch, G.H., Spence, J.R., 2015. In vitro generation of human pluripotent stem cell derived lung organoids. Elife 4. Eicher, A.K., Berns, H.M., Wells, J.M., 2018. Translating Developmental Principles to Generate Human Gastric Organoids. Cell Mol Gastroenterol Hepatol 5(3), 353-363. Eiraku, M., Takata, N., Ishibashi, H., Kawada, M., Sakakura, E., Okuda, S., Sekiguchi, K., Adachi, T., Sasai, Y., 2011. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 472, 51. Eiraku, M., Watanabe, K., Matsuo-Takasaki, M., Kawada, M., Yonemura, S., Matsumura, M., Wataya, T., Nishiyama, A., Muguruma, K., Sasai, Y., 2008. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell 3(5), 519-532. Escaffre, O., Borisevich, V., Vergara, L.A., Wen, J.W., Long, D., Rockx, B., 2016. Characterization of Nipah virus infection in a model of human airway epithelial cells cultured at an air-liquid interface. J Gen Virol 97(5), 1077-1086. Ettayebi, K., Crawford, S.E., Murakami, K., Broughman, J.R., Karandikar, U., Tenge, V.R., Neill, F.H., Blutt, S.E., Zeng, X.-L., Qu, L., Kou, B., Opekun, A.R., Burrin, D., Graham, D.Y., Ramani, S., Atmar, R.L., Estes, M.K., 2016. Replication of human noroviruses in stem cell-derived human enteroids. Science (New York, N.Y.) 353(6306), 1387-1393. Fatehullah, A., Tan, S.H., Barker, N., 2016. Organoids as an in vitro model of human development and disease. Nat Cell Biol 18(3), 246-254. Fatimah, S.S., Chua, K., Tan, G.C., Azmi, T.I., Tan, A.E., Abdul Rahman, H., 2013. Organotypic culture of human amnion cells in air-liquid interface as a potential substitute for skin regeneration. Cytotherapy 15(8), 1030-1041. Ferreira, J.N., Hasan, R., Urkasemsin, G., Ng, K.K., Adine, C., Muthumariappan, S., Souza, G.R., 2019. A magnetic three-dimensional levitated primary cell culture system for the development of secretory salivary gland-like organoids. Journal of Tissue Engineering and Regenerative Medicine 0(0).
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Fessart, D., Begueret, H., Delom, F., 2013. Three-dimensional culture model to distinguish normal from malignant human bronchial epithelial cells. European Respiratory Journal 42(5), 1345. Finkbeiner, S.R., Zeng, X.-L., Utama, B., Atmar, R.L., Shroyer, N.F., Estes, M.K., 2012. Stem Cell-Derived Human Intestinal Organoids as an Infection Model for Rotaviruses. mBio 3(4), e00159-00112. Foulke-Abel, J., In, J., Yin, J., Zachos, N.C., Kovbasnjuk, O., Estes, M.K., de Jonge, H., Donowitz, M., 2016. Human Enteroids as a Model of Upper Small Intestinal Ion Transport Physiology and Pathophysiology. Gastroenterology 150(3), 638-649.e638. Freedman, B.S., Brooks, C.R., Lam, A.Q., Fu, H., Morizane, R., Agrawal, V., Saad, A.F., Li, M.K., Hughes, M.R., Werff, R.V., Peters, D.T., Lu, J., Baccei, A., Siedlecki, A.M., Valerius, M.T., Musunuru, K., McNagny, K.M., Steinman, T.I., Zhou, J., Lerou, P.H., Bonventre, J.V., 2015. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat Commun 6, 8715. Gabriel, E., Wason, A., Ramani, A., Gooi, L.M., Keller, P., Pozniakovsky, A., Poser, I., Noack, F., Telugu, N.S., Calegari, F., Saric, T., Hescheler, J., Hyman, A.A., Gottardo, M., Callaini, G., Alkuraya, F.S., Gopalakrishnan, J., 2016. CPAP promotes timely cilium disassembly to maintain neural progenitor pool. EMBO J 35(8), 803-819. Gao, D., Vela, I., Sboner, A., Iaquinta, Phillip J., Karthaus, Wouter R., Gopalan, A., Dowling, C., Wanjala, Jackline N., Undvall, Eva A., Arora, Vivek K., Wongvipat, J., Kossai, M., Ramazanoglu, S., Barboza, Luendreo P., Di, W., Cao, Z., Zhang, Qi F., Sirota, I., Ran, L., MacDonald, Theresa Y., Beltran, H., Mosquera, J.-M., Touijer, Karim A., Scardino, Peter T., Laudone, Vincent P., Curtis, Kristen R., Rathkopf, Dana E., Morris, Michael J., Danila, Daniel C., Slovin, Susan F., Solomon, Stephen B., Eastham, James A., Chi, P., Carver, B., Rubin, Mark A., Scher, Howard I., Clevers, H., Sawyers, Charles L., Chen, Y., 2014. Organoid Cultures Derived from Patients with Advanced Prostate Cancer. Cell 159(1), 176-187. Gao, D., Vela, I., Sboner, A., Iaquinta, P.J., Karthaus, W.R., Gopalan, A., Dowling, C., Wanjala, J.N., Undvall, E.A., Arora, V.K., Wongvipat, J., Kossai, M., Ramazanoglu, S., Barboza, L.P., Di, W., Cao, Z., Zhang, Q.F., Sirota, I., Ran, L., MacDonald, T.Y., Beltran, H., Mosquera, J.M., Touijer, K.A., Scardino, P.T., Laudone, V.P., Curtis, K.R., Rathkopf, D.E., Morris, M.J., Danila, D.C., Slovin, S.F., Solomon, S.B., Eastham, J.A., Chi, P., Carver, B., Rubin, M.A., Scher, H.I., Clevers, H., Sawyers, C.L., Chen, Y., Organoid cultures derived from patients with advanced prostate cancer. (1097-4172 (Electronic)). Garcez, P.P., Loiola, E.C., Madeiro da Costa, R., Higa, L.M., Trindade, P., Delvecchio, R., Nascimento, J.M., Brindeiro, R., Tanuri, A., Rehen, S.K., 2016. Zika virus impairs growth in human neurospheres and brain organoids. Science. Giandomenico, S.L., Mierau, S.B., Gibbons, G.M., Wenger, L.M.D., Masullo, L., Sit, T., Sutcliffe, M., Boulanger, J., Tripodi, M., Derivery, E., Paulsen, O., Lakatos, A., Lancaster, M., 2018. Cerebral organoids at the air-liquid interface generate diverse nerve tracts with functional output. Gold, R., Linington, C., Lassmann, H., 2006. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 129(8), 1953-1971. Greggio, C., De Franceschi, F., Figueiredo-Larsen, M., Gobaa, S., Ranga, A., Semb, H., Lutolf, M., Grapin-Botton, A., 2013. Artificial three-dimensional niches deconstruct pancreas development in vitro. Development (Cambridge, England) 140(21), 4452-4462. Haegel, H., Larue, L., Ohsugi, M., Fedorov, L., Herrenknecht, K., Kemler, R., 1995. Lack of beta-catenin affects mouse development at gastrulation. Development 121(11), 3529. Hisha, H., Tanaka, T., Kanno, S., Tokuyama, Y., Komai, Y., Ohe, S., Yanai, H., Omachi, T., Ueno, H., 2013. Establishment of a Novel Lingual Organoid Culture System: Generation of
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Organoids Having Mature Keratinized Epithelium from Adult Epithelial Stem Cells. Scientific Reports 3, 3224. Hohwieler, M., Illing, A., Hermann, P.C., Mayer, T., Stockmann, M., Perkhofer, L., Eiseler, T., Antony, J.S., Müller, M., Renz, S., Kuo, C.-C., Lin, Q., Sendler, M., Breunig, M., Kleiderman, S.M., Lechel, A., Zenker, M., Leichsenring, M., Rosendahl, J., Zenke, M., Sainz, B., Mayerle, J., Costa, I.G., Seufferlein, T., Kormann, M., Wagner, M., Liebau, S., Kleger, A., 2017. Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling. Gut 66(3), 473. Homan, K.A., Gupta, N., Kroll, K.T., Kolesky, D.B., Skylar-Scott, M., Miyoshi, T., Mau, D., Valerius, M.T., Ferrante, T., Bonventre, J.V., Lewis, J.A., Morizane, R., 2019. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nature Methods 16(3), 255-262. Hsu, S.Y., Liang, S.-G., Hsueh, A.J.W., 1998. Characterization of Two LGR Genes Homologous to Gonadotropin and Thyrotropin Receptors with Extracellular Leucine-Rich Repeats and a G Protein-Coupled, Seven-Transmembrane Region. Molecular Endocrinology 12(12), 1830-1845. Huang, L., Holtzinger, A., Jagan, I., BeGora, M., Lohse, I., Ngai, N., Nostro, C., Wang, R., Muthuswamy, L.B., Crawford, H.C., Arrowsmith, C., Kalloger, S.E., Renouf, D.J., Connor, A.A., Cleary, S., Schaeffer, D.F., Roehrl, M., Tsao, M.-S., Gallinger, S., Keller, G., Muthuswamy, S.K., 2015. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell– and patient-derived tumor organoids. Nature Medicine 21, 1364. Huch, M., Dorrell, C., Boj, S.F., van Es, J.H., Li, V.S.W., van de Wetering, M., Sato, T., Hamer, K., Sasaki, N., Finegold, M.J., Haft, A., Vries, R.G., Grompe, M., Clevers, H., 2013. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494, 247. Huch M Fau - Boj, S.F., Boj Sf Fau - Clevers, H., Clevers, H., Lgr5(+) liver stem cells, hepatic organoids and regenerative medicine. (1746-076X (Electronic)). Huch, M., Koo, B.-K., 2015. Modeling mouse and human development using organoid cultures. Development 142(18), 3113. Huch, M.A.-O., Knoblich, J.A.-O., Lutolf, M.A.-O.X., Martinez-Arias, A.A.-O.X., The hope and the hype of organoid research. (1477-9129 (Electronic)). Hughes, C.S., Postovit Lm Fau - Lajoie, G.A., Lajoie, G.A., Matrigel: a complex protein mixture required for optimal growth of cell culture. (1615-9861 (Electronic)). Huh, D., Hamilton, G.A., Ingber, D.E., 2011. From 3D cell culture to organs-on-chips. Trends in Cell Biology 21(12), 745-754. Hui, E.E., Bhatia, S.N., 2007. Micromechanical control of cell-cell interactions. (0027-8424 (Print)). Hwang, C.-I., Boj, S.F., Clevers, H., Tuveson, D.A., 2016. Preclinical models of pancreatic ductal adenocarcinoma. The Journal of Pathology 238(2), 197-204. Kamb, A., Wee, S., Lengauer, C., 2006. Why is cancer drug discovery so difficult? Nature Reviews Drug Discovery 6, 115. Kang, H.-W., Atala, A., Yoo, J.J., 2015. Chapter 10 - Bioprinting of Organs for Toxicology Testing, in: Atala, A., Yoo, J.J. (Eds.), Essentials of 3D Biofabrication and Translation. Academic Press, Boston, pp. 179-186. Kaushik, G., Ponnusamy, M.P., Batra, S.K., 2018. Concise Review: Current Status of ThreeDimensional Organoids as Preclinical Models. Stem Cells 36(9), 1329-1340. Kelava, I., Lancaster, M.A., Stem Cell Models of Human Brain Development. (1875-9777 (Electronic)). Kim, J., Koo, B.-K., Yoon, K.-J., 2019. Modeling Host-Virus Interactions in Viral Infectious Diseases Using Stem-Cell-Derived Systems and CRISPR/Cas9 Technology. Viruses 11(2), 124. Kimura, H., Sakai, Y., Fujii, T., 2018. Organ/body-on-a-chip based on microfluidic technology for drug discovery. Drug Metabolism and Pharmacokinetics 33(1), 43-48.
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Koehler, K.R., Mikosz, A.M., Molosh, A.I., Patel, D., Hashino, E., 2013. Generation of inner ear sensory epithelia from pluripotent stem cells in 3D culture. Nature 500, 217. Koike, M., Kurosawa, H., Amano, Y., 2005. A Round-bottom 96-well Polystyrene Plate Coated with 2-methacryloyloxyethyl Phosphorylcholine as an Effective Tool for Embryoid Body Formation. Cytotechnology 47(1-3), 3-10. Kopper, O., de Witte, C.J., Lõhmussaar, K., Valle-Inclan, J.E., Hami, N., Kester, L., Balgobind, A.V., Korving, J., Proost, N., Begthel, H., van Wijk, L.M., Revilla, S.A., Theeuwsen, R., van de Ven, M., van Roosmalen, M.J., Ponsioen, B., Ho, V.W.H., Neel, B.G., Bosse, T., Gaarenstroom, K.N., Vrieling, H., Vreeswijk, M.P.G., van Diest, P.J., Witteveen, P.O., Jonges, T., Bos, J.L., van Oudenaarden, A., Zweemer, R.P., Snippert, H.J.G., Kloosterman, W.P., Clevers, H., 2019. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nature medicine 25(5), 838-849. Krefft, O., Jabali, A., Iefremova, V., Koch, P., Ladewig, J., 2018. Generation of Standardized and Reproducible Forebrain-type Cerebral Organoids from Human Induced Pluripotent Stem Cells. J Vis Exp(131). Lancaster, M.A., 2018. Brain organoids get vascularized. Nat Biotechnol 36(5), 407-408. Lancaster, M.A., Knoblich, J.A., 2014. Generation of cerebral organoids from human pluripotent stem cells. Nat Protoc 9(10), 2329-2340. Lancaster, M.A., Knoblich, J.A., 2014. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 345(6194). Lancaster, M.A., Renner, M., Martin, C.-A., Wenzel, D., Bicknell, L.S., Hurles, M.E., Homfray, T., Penninger, J.M., Jackson, A.P., Knoblich, J.A., 2013. Cerebral organoids model human brain development and microcephaly. Nature 501, 373. Lazear, Helen M., Govero, J., Smith, Amber M., Platt, Derek J., Fernandez, E., Miner, Jonathan J., Diamond, Michael S., 2016. A Mouse Model of Zika Virus Pathogenesis. Cell Host & Microbe 19(5), 720-730. Lee, G.Y., Kenny, P.A., Lee, E.H., Bissell, M.J., 2007. Three-dimensional culture models of normal and malignant breast epithelial cells. Nature Methods 4, 359. Lee, S.H., Hu, W., Matulay, J.T., Silva, M.V., Owczarek, T.B., Kim, K., Chua, C.W., Barlow, L.J., Kandoth, C., Williams, A.B., Bergren, S.K., Pietzak, E.J., Anderson, C.B., Benson, M.C., Coleman, J.A., Taylor, B.S., Abate-Shen, C., McKiernan, J.M., Al-Ahmadie, H., Solit, D.B., Shen, M.M., 2018. Tumor Evolution and Drug Response in Patient-Derived Organoid Models of Bladder Cancer. Cell 173(2), 515-528.e517. Lee, V., Singh, G., Trasatti, J.P., Bjornsson, C., Xu, X., Tran, T.N., Yoo, S.-S., Dai, G., Karande, P., 2013. Design and Fabrication of Human Skin by Three-Dimensional Bioprinting. Tissue Engineering Part C: Methods 20(6), 473-484. Li, F., 2019. What is the difference between spheroids and organoids and what they can be used for? https://cellculturedish.com/questions/difference-spheroids-organoids-can-used/. Li, X., Nadauld, L., Ootani, A., Corney, D.C., Pai, R.K., Gevaert, O., Cantrell, M.A., Rack, P.G., Neal, J.T., Chan, C.W., Yeung, T., Gong, X., Yuan, J., Wilhelmy, J., Robine, S., Attardi, L.D., Plevritis, S.K., Hung, K.E., Chen, C.Z., Ji, H.P., Kuo, C.J., 2014. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med 20(7), 769-777. Li, X., Nadauld, L., Ootani, A., Corney, D.C., Pai, R.K., Gevaert, O., Cantrell, M.A., Rack, P.G., Neal, J.T., Chan, C.W.M., Yeung, T., Gong, X., Yuan, J., Wilhelmy, J., Robine, S., Attardi, L.D., Plevritis, S.K., Hung, K.E., Chen, C.-Z., Ji, H.P., Kuo, C.J., 2014. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nature Medicine 20, 769. Li, X., Ootani, A., Kuo, C., 2016. An Air-Liquid Interface Culture System for 3D Organoid Culture of Diverse Primary Gastrointestinal Tissues. Methods Mol Biol 1422, 33-40. Ling, K.H., Mason, J.O., Cheah, P.-S., 2019. Challenges and future perspectives for 3D cerebral organoids as a model for complex brain disorders. Neuroscience Research Notes 2(1), 1-6.
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Liu, P., Wakamiya, M., Shea, M.J., Albrecht, U., Behringer, R.R., Bradley, A., 1999. Requirement for Wnt3 in vertebrate axis formation. Nature Genetics 22, 361. Liu, Y., Qi, Z., Li, X., Du, Y., Chen, Y.G., 2018. Monolayer culture of intestinal epithelium sustains Lgr5(+) intestinal stem cells. Cell Discov 4, 32. Lokody, I., 2013. Correcting genetic defects with CRISPR–Cas9. Nature Reviews Genetics 15, 63. Lu, Y.-C., Fu, D.-J., An, D., Chiu, A., Schwartz, R., Nikitin, A.Y., Ma, M., 2017. Scalable Production and Cryostorage of Organoids Using Core–Shell Decoupled Hydrogel Capsules. Advanced Biosystems 1(12), 1700165. Maimets, M., Rocchi, C., Bron, R., Pringle, S., Kuipers, J., Giepmans, Ben N.G., Vries, Robert G.J., Clevers, H., de Haan, G., van Os, R., Coppes, Robert P., 2016. Long-Term In Vitro Expansion of Salivary Gland Stem Cells Driven by Wnt Signals. Stem Cell Reports 6(1), 150162. Mansour, A.A., Goncalves, J.T., Bloyd, C.W., Li, H., Fernandes, S., Quang, D., Johnston, S., Parylak, S.L., Jin, X., Gage, F.H., 2018. An in vivo model of functional and vascularized human brain organoids. Nat Biotechnol 36(5), 432-441. Mariani, J., Coppola, G., Zhang, P., Abyzov, A., Provini, L., Tomasini, L., Amenduni, M., Szekely, A., Palejev, D., Wilson, M., Gerstein, M., Grigorenko, E.L., Chawarska, K., Pelphrey, K.A., Howe, J.R., Vaccarino, F.M., 2015. FOXG1-Dependent Dysregulation of GABA/Glutamate Neuron Differentiation in Autism Spectrum Disorders. Cell 162(2), 375-390. Matano, M., Date, S., Shimokawa, M., Takano, A., Fujii, M., Ohta, Y., Watanabe, T., Kanai, T., Sato, T., 2015. Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids. Nature Medicine 21, 256. McCracken, K.W., Catá, E.M., Crawford, C.M., Sinagoga, K.L., Schumacher, M., Rockich, B.E., Tsai, Y.-H., Mayhew, C.N., Spence, J.R., Zavros, Y., Wells, J.M., 2014. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature 516, 400. McMurtrey, R.J., 2016. Analytic Models of Oxygen and Nutrient Diffusion, Metabolism Dynamics, and Architecture Optimization in Three-Dimensional Tissue Constructs with Applications and Insights in Cerebral Organoids. (1937-3392 (Electronic)). Merck, 2019. 3D Organoid Culture: New In Vitro Models of Development and Disease. https://www.sigmaaldrich.com/technical-documents/articles/biology/cell-culture/3d-organoidculture.html. Mitaka, T., 2002. Reconstruction of hepatic organoid by hepatic stem cells. Journal of HepatoBiliary-Pancreatic Surgery 9(6), 697-703. Morizane, R., Bonventre, J.V., 2017. Generation of nephron progenitor cells and kidney organoids from human pluripotent stem cells. Nat Protoc 12(1), 195-207. Muguruma, K., Nishiyama, A., Kawakami, H., Hashimoto, K., Sasai, Y., 2015. Self-Organization of Polarized Cerebellar Tissue in 3D Culture of Human Pluripotent Stem Cells. Cell Reports 10(4), 537-550. Nadkarni, R.R., Abed, S., Draper, J.S., 2016. Organoids as a model system for studying human lung development and disease. Biochemical and Biophysical Research Communications 473(3), 675-682. Nadkarni, R.R., Abed, S., Draper, J.S., 2016. Organoids as a model system for studying human lung development and disease. Biochem Biophys Res Commun 473(3), 675-682. Nakamura, T., 2019. Recent progress in organoid culture to model intestinal epithelial barrier functions. Int Immunol 31(1), 13-21. Nanduri, Lalitha S.Y., Baanstra, M., Faber, H., Rocchi, C., Zwart, E., de Haan, G., van Os, R., Coppes, Robert P., 2014. Purification and Ex Vivo Expansion of Fully Functional Salivary Gland Stem Cells. Stem Cell Reports 3(6), 957-964. Neal, J.T., Li, X., Zhu, J., Giangarra, V., Grzeskowiak, C.L., Ju, J., Liu, I.H., Chiou, S.-H., Salahudeen, A.A., Smith, A.R., Deutsch, B.C., Liao, L., Zemek, A.J., Zhao, F., Karlsson, K.,
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Schultz, L.M., Metzner, T.J., Nadauld, L.D., Tseng, Y.-Y., Alkhairy, S., Oh, C., Keskula, P., Mendoza-Villanueva, D., De La Vega, F.M., Kunz, P.L., Liao, J.C., Leppert, J.T., Sunwoo, J.B., Sabatti, C., Boehm, J.S., Hahn, W.C., Zheng, G.X.Y., Davis, M.M., Kuo, C.J., 2018. Organoid Modeling of the Tumor Immune Microenvironment. Cell 175(7), 1972-1988.e1916. Nie, J., Hashino, E., 2017. Organoid technologies meet genome engineering. EMBO reports 18(3), 367-376. Noordhoek, J., Gulmans, V., van der Ent, K., Beekman, J.M., 2016. Intestinal organoids and personalized medicine in cystic fibrosis: a successful patient-oriented research collaboration. Curr Opin Pulm Med 22(6), 610-616. Nugraha, B., Buono, M.F., von Boehmer, L., Hoerstrup, S.P., Emmert, M.Y., 2019. Human Cardiac Organoids for Disease Modeling. Clin Pharmacol Ther 105(1), 79-85. O'Rourke, K.P., Ackerman, S., Dow, L.E., Lowe, S.W., 2016. Isolation, Culture, and Maintenance of Mouse Intestinal Stem Cells. Bio-protocol 6(4), e1733. O'Rourke, K.P., Loizou, E., Livshits, G., Schatoff, E.M., Baslan, T., Manchado, E., Simon, J., Romesser, P.B., Leach, B., Han, T., Pauli, C., Beltran, H., Rubin, M.A., Dow, L.E., Lowe, S.W., 2017. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat Biotechnol 35(6), 577-582. Ootani, A., Li, X., Sangiorgi, E., Ho, Q.T., Ueno, H., Toda, S., Sugihara, H., Fujimoto, K., Weissman, I.L., Capecchi, M.R., Kuo, C.J., 2009. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat Med 15(6), 701-706. Pageau, S.C., Sazonova, O.V., Wong, J.Y., Soto, A.M., Sonnenschein, C., 2011. The effect of stromal components on the modulation of the phenotype of human bronchial epithelial cells in 3D culture. Biomaterials 32(29), 7169-7180. Paşca, S.P., 2019. Assembling human brain organoids. Science 363(6423), 126. Porotto, M., Ferren, M., Chen, Y.W., Siu, Y., Makhsous, N., Rima, B., Briese, T., Greninger, A.L., Snoeck, H.W., Moscona, A., 2019. Authentic Modeling of Human Respiratory Virus Infection in Human Pluripotent Stem Cell-Derived Lung Organoids. mBio 10(3), e00723-00719. Pruniéras, M., Régnier, M., Woodley, D., 1983. Methods for Cultivation of Keratinocytes with an Air-Liquid Interface. Journal of Investigative Dermatology 81(1), S28-S33. Purcell, O., Lu, T.K., 2014. Synthetic analog and digital circuits for cellular computation and memory. Curr Opin Biotechnol 29, 146-155. Qian, X., Nguyen, H.N., Song, M.M., Hadiono, C., Ogden, S.C., Hammack, C., Yao, B., Hamersky, G.R., Jacob, F., Zhong, C., Yoon, K.J., Jeang, W., Lin, L., Li, Y., Thakor, J., Berg, D.A., Zhang, C., Kang, E., Chickering, M., Nauen, D., Ho, C.Y., Wen, Z., Christian, K.M., Shi, P.Y., Maher, B.J., Wu, H., Jin, P., Tang, H., Song, H., Ming, G.L., 2016. Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure. Cell 165(5), 1238-1254. Quadrato, G., Nguyen, T., Macosko, E.Z., Sherwood, J.L., Min Yang, S., Berger, D.R., Maria, N., Scholvin, J., Goldman, M., Kinney, J.P., Boyden, E.S., Lichtman, J.W., Williams, Z.M., McCarroll, S.A., Arlotta, P., 2017. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545(7652), 48-53. Research, A.S., 2019. New spinal-cord-like cell clusters could be used to study and find treatments for nerve cell degeneration. https://research.a-star.edu.sg/research/8032/spinalorganoids-mimic-neurodegenerative-disease. Rossi, G., Manfrin, A., Lutolf, M.P., 2018. Progress and potential in organoid research. Nat Rev Genet 19(11), 671-687. Sachs, N., de Ligt, J., Kopper, O., Gogola, E., Bounova, G., Weeber, F., Balgobind, A.V., Wind, K., Gracanin, A., Begthel, H., Korving, J., van Boxtel, R., Duarte, A.A., Lelieveld, D., van Hoeck, A., Ernst, R.F., Blokzijl, F., Nijman, I.J., Hoogstraat, M., van de Ven, M., Egan, D.A., Zinzalla, V., Moll, J., Boj, S.F., Voest, E.E., Wessels, L., van Diest, P.J., Rottenberg, S., Vries, R.G.J., Cuppen, E., Clevers, H., 2018. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell 172(1), 373-386.e310.
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Saglam-Metiner, P., Gulce-Iz, S., Biray-Avci, C., 2019. Bioengineering-inspired threedimensional culture systems: Organoids to create tumor microenvironment. Gene 686, 203-212. Sato, T., Clevers, H., 2013. Growing Self-Organizing Mini-Guts from a Single Intestinal Stem Cell: Mechanism and Applications. Science 340(6137), 1190. Sato, T., Stange, D.E., Ferrante, M., Vries, R.G., Van Es, J.H., Van den Brink, S., Van Houdt, W.J., Pronk, A., Van Gorp, J., Siersema, P.D., Clevers, H., 2011. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology 141(5), 1762-1772. Sato, T., Stange, D.E., Ferrante, M., Vries, R.G.J., van Es, J.H., van den Brink, S., van Houdt, W.J., Pronk, A., van Gorp, J., Siersema, P.D., Clevers, H., 2011. Long-term Expansion of Epithelial Organoids From Human Colon, Adenoma, Adenocarcinoma, and Barrett's Epithelium. Gastroenterology 141(5), 1762-1772. Sato, T., van Es, J.H., Snippert, H.J., Stange, D.E., Vries, R.G., van den Born, M., Barker, N., Shroyer, N.F., van de Wetering, M., Clevers, H., 2010. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415. Sato, T., Vries, R.G., Snippert, H.J., van de Wetering, M., Barker, N., Stange, D.E., van Es, J.H., Abo, A., Kujala, P., Peters, P.J., Clevers, H., 2009. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459(7244), 262-265. Schwank, G., Koo, B.-K., Sasselli, V., Dekkers, Johanna F., Heo, I., Demircan, T., Sasaki, N., Boymans, S., Cuppen, E., van der Ent, Cornelis K., Nieuwenhuis, Edward E.S., Beekman, Jeffrey M., Clevers, H., 2013. Functional Repair of CFTR by CRISPR/Cas9 in Intestinal Stem Cell Organoids of Cystic Fibrosis Patients. Cell Stem Cell 13(6), 653-658. Seidlitz, T., Merker, S.R., Rothe, A., Zakrzewski, F., von Neubeck, C., Grützmann, K., Sommer, U., Schweitzer, C., Schölch, S., Uhlemann, H., Gaebler, A.-M., Werner, K., Krause, M., Baretton, G.B., Welsch, T., Koo, B.-K., Aust, D.E., Klink, B., Weitz, J., Stange, D.E., 2019. Human gastric cancer modelling using organoids. Gut 68(2), 207-217. Sekiya, S., Kikuchi, T., Shimizu, T., 2019. Perfusion culture maintained with an air-liquid interface to stimulate epithelial cell organization in renal organoids in vitro. BMC Biomedical Engineering 1(1), 15. Shook, D., Keller, R., 2003. Mechanisms, mechanics and function of epithelial–mesenchymal transitions in early development. Mechanisms of Development 120(11), 1351-1383. Simian, M., Hirai Y Fau - Navre, M., Navre M Fau - Werb, Z., Werb Z Fau - Lochter, A., Lochter A Fau - Bissell, M.J., Bissell, M.J., 2001. The interplay of matrix metalloproteinases, morphogens and growth factors is necessary for branching of mammary epithelial cells. (09501991 (Print)). Skardal, A., Shupe, T., Atala, A., 2016. Organoid-on-a-chip and body-on-a-chip systems for drug screening and disease modeling. Drug Discov Today 21(9), 1399-1411. Souza, G.R., Molina, J.R., Raphael, R.M., Ozawa, M.G., Stark, D.J., Levin, C.S., Bronk, L.F., Ananta, J.S., Mandelin, J., Georgescu, M.-M., Bankson, J.A., Gelovani, J.G., Killian, T.C., Arap, W., Pasqualini, R., 2010. Three-dimensional tissue culture based on magnetic cell levitation. Nature Nanotechnology 5, 291. Spence, J.R., Mayhew, C.N., Rankin, S.A., Kuhar, M.F., Vallance, J.E., Tolle, K., Hoskins, E.E., Kalinichenko, V.V., Wells, S.I., Zorn, A.M., Shroyer, N.F., Wells, J.M., 2011. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470(7332), 105-109. Spencer, J.A., Ferraro, F., Roussakis, E., Klein, A., Wu, J., Runnels, J.M., Zaher, W., Mortensen, L.J., Alt, C., Turcotte, R., Yusuf, R., Cote, D., Vinogradov, S.A., Scadden, D.T., Lin, C.P., Direct measurement of local oxygen concentration in the bone marrow of live animals. (1476-4687 (Electronic)). Stange, Daniel E., Koo, B.-K., Huch, M., Sibbel, G., Basak, O., Lyubimova, A., Kujala, P., Bartfeld, S., Koster, J., Geahlen, Jessica H., Peters, Peter J., van Es, Johan H.,
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
van de Wetering, M., Mills, Jason C., Clevers, H., 2013. Differentiated Troy+ Chief Cells Act as Reserve Stem Cells to Generate All Lineages of the Stomach Epithelium. Cell 155(2), 357-368. Stevens, K.R., Kreutziger Kl Fau - Dupras, S.K., Dupras Sk Fau - Korte, F.S., Korte Fs Fau Regnier, M., Regnier M Fau - Muskheli, V., Muskheli V Fau - Nourse, M.B., Nourse Mb Fau Bendixen, K., Bendixen K Fau - Reinecke, H., Reinecke H Fau - Murry, C.E., Murry, C.E., 2009. Physiological function and transplantation of scaffold-free and vascularized human cardiac muscle tissue. (1091-6490 (Electronic)). Strange, D.P., Zarandi, N.P., Trivedi, G., Atala, A., Bishop, C.E., Sadri-Ardekani, H., Verma, S., 2018. Human testicular organoid system as a novel tool to study Zika virus pathogenesis. Emerging Microbes & Infections 7(1), 82. Sun, T., Jackson, S., Haycock, J.W., MacNeil, S., 2006. Culture of skin cells in 3D rather than 2D improves their ability to survive exposure to cytotoxic agents. Journal of Biotechnology 122(3), 372-381. Sutherland Rm Fau - McCredie, J.A., McCredie Ja Fau - Inch, W.R., Inch, W.R., Growth of multicell spheroids in tissue culture as a model of nodular carcinomas. (0027-8874 (Print)). Taguchi, A., Kaku, Y., Ohmori, T., Sharmin, S., Ogawa, M., Sasaki, H., Nishinakamura, R., 2014. Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell 14(1), 53-67. Takasato, M., Er, P.X., Becroft, M., Vanslambrouck, J.M., Stanley, E.G., Elefanty, A.G., Little, M.H., 2014. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat Cell Biol 16(1), 118-126. Takasato, M., Er, P.X., Chiu, H.S., Little, M.H., 2016. Generation of kidney organoids from human pluripotent stem cells. Nat Protoc 11(9), 1681-1692. Takasato, M., Little, M.H., 2015. The origin of the mammalian kidney: implications for recreating the kidney in vitro. Development 142(11), 1937-1947. Takebe, T., Sekine, K., Enomura, M., Koike, H., Kimura, M., Ogaeri, T., Zhang, R.-R., Ueno, Y., Zheng, Y.-W., Koike, N., Aoyama, S., Adachi, Y., Taniguchi, H., 2013. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499, 481. Takebe, T., Sekine K Fau - Enomura, M., Enomura M Fau - Koike, H., Koike H Fau - Kimura, M., Kimura M Fau - Ogaeri, T., Ogaeri T Fau - Zhang, R.-R., Zhang Rr Fau - Ueno, Y., Ueno Y Fau - Zheng, Y.-W., Zheng Yw Fau - Koike, N., Koike N Fau - Aoyama, S., Aoyama S Fau Adachi, Y., Adachi Y Fau - Taniguchi, H., Taniguchi, H., Vascularized and functional human liver from an iPSC-derived organ bud transplant. (1476-4687 (Electronic)). Thorne, C.A., Chen, I.W., Sanman, L.E., Cobb, M.H., Wu, L.F., Altschuler, S.J., 2018. Enteroid Monolayers Reveal an Autonomous WNT and BMP Circuit Controlling Intestinal Epithelial Growth and Organization. Dev Cell 44(5), 624-633 e624. Toda, S., Watanabe, K., Yokoi, F., Matsumura, S., Suzuki, K., Ootani, A., Aoki, S., Koike, N., Sugihara, H., 2002. A new organotypic culture of thyroid tissue maintains three-dimensional follicles with C cells for a long term. Biochemical and Biophysical Research Communications 294(4), 906-911. Tseng, H., Gage, J.A., Raphael, R.M., Moore, R.H., Killian, T.C., Grande-Allen, K.J., Souza, G.R., 2013. Assembly of a Three-Dimensional Multitype Bronchiole Coculture Model Using Magnetic Levitation. Tissue Engineering Part C: Methods 19(9), 665-675. Tucker, B.A., Solivan-Timpe, F., Roos, B.R., Anfinson, K.R., Robin, A.L., Wiley, L.A., Mullins, R.F., Fingert, J.H., 2014. Duplication of TBK1 Stimulates Autophagy in iPSC-derived Retinal Cells from a Patient with Normal Tension Glaucoma. J Stem Cell Res Ther 3(5), 161. Tuveson, D., Clevers, H., 2019. Cancer modeling meets human organoid technology. Science 364(6444), 952. Upadhyay, S., Palmberg, L., 2018. Air-Liquid Interface: Relevant In Vitro Models for Investigating Air Pollutant-Induced Pulmonary Toxicity. Toxicol Sci 164(1), 21-30.
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Usui, T., Sakurai, M., Umata, K., Yamawaki, H., Ohama, T., Sato, K., 2018. Preparation of Human Primary Colon Tissue-Derived Organoid Using Air Liquid Interface Culture. Curr Protoc Toxicol 75, 22 26 21-22 26 27. van de Wetering, M., Francies, H.E., Francis, J.M., Bounova, G., Iorio, F., Pronk, A., van Houdt, W., van Gorp, J., Taylor-Weiner, A., Kester, L., McLaren-Douglas, A., Blokker, J., Jaksani, S., Bartfeld, S., Volckman, R., van Sluis, P., Li, V.S., Seepo, S., Sekhar Pedamallu, C., Cibulskis, K., Carter, S.L., McKenna, A., Lawrence, M.S., Lichtenstein, L., Stewart, C., Koster, J., Versteeg, R., van Oudenaarden, A., Saez-Rodriguez, J., Vries, R.G., Getz, G., Wessels, L., Stratton, M.R., McDermott, U., Meyerson, M., Garnett, M.J., Clevers, H., 2015. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161(4), 933-945. van der Hee, B., Loonen, L.M.P., Taverne, N., Taverne-Thiele, J.J., Smidt, H., Wells, J.M., 2018. Optimized procedures for generating an enhanced, near physiological 2D culture system from porcine intestinal organoids. Stem Cell Res 28, 165-171. van der Worp, H.B., Howells, D.W., Sena, E.S., Porritt, M.J., Rewell, S., O'Collins, V., Macleod, M.R., 2010. Can Animal Models of Disease Reliably Inform Human Studies? PLOS Medicine 7(3), e1000245. van de Wetering, M., Francies, Hayley E., Francis, Joshua M., Bounova, G., Iorio, F., Pronk, A., van Houdt, W., van Gorp, J., Taylor-Weiner, A., Kester, L., McLaren-Douglas, A., Blokker, J., Jaksani, S., Bartfeld, S., Volckman, R., van Sluis, P., Li, Vivian S.W., Seepo, S., Sekhar Pedamallu, C., Cibulskis, K., Carter, Scott L., McKenna, A., Lawrence, Michael S., Lichtenstein, L., Stewart, C., Koster, J., Versteeg, R., van Oudenaarden, A., Saez-Rodriguez, J., Vries, Robert G.J., Getz, G., Wessels, L., Stratton, Michael R., McDermott, U., Meyerson, M., Garnett, Mathew J., Clevers, H., 2015. Prospective Derivation of a Living Organoid Biobank of Colorectal Cancer Patients. Cell 161(4), 933-945. Vaughan, M.B., Ramirez Rd Fau - Wright, W.E., Wright We Fau - Minna, J.D., Minna Jd Fau Shay, J.W., Shay, J.W., 2006. A three-dimensional model of differentiation of immortalized human bronchial epithelial cells. (0301-4681 (Print)). Vlachogiannis, G., Hedayat, S., Vatsiou, A., Jamin, Y., Fernández-Mateos, J., Khan, K., Lampis, A., Eason, K., Huntingford, I., Burke, R., Rata, M., Koh, D.-M., Tunariu, N., Collins, D., HulkkiWilson, S., Ragulan, C., Spiteri, I., Moorcraft, S.Y., Chau, I., Rao, S., Watkins, D., Fotiadis, N., Bali, M., Darvish-Damavandi, M., Lote, H., Eltahir, Z., Smyth, E.C., Begum, R., Clarke, P.A., Hahne, J.C., Dowsett, M., de Bono, J., Workman, P., Sadanandam, A., Fassan, M., Sansom, O.J., Eccles, S., Starling, N., Braconi, C., Sottoriva, A., Robinson, S.P., Cunningham, D., Valeri, N., 2018. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science (New York, N.Y.) 359(6378), 920-926. Vlachogiannis, G., Hedayat, S., Vatsiou, A., Jamin, Y., Fernandez-Mateos, J., Khan, K., Lampis, A., Eason, K., Huntingford, I., Burke, R., Rata, M., Koh, D.M., Tunariu, N., Collins, D., HulkkiWilson, S., Ragulan, C., Spiteri, I., Moorcraft, S.Y., Chau, I., Rao, S., Watkins, D., Fotiadis, N., Bali, M., Darvish-Damavandi, M., Lote, H., Eltahir, Z., Smyth, E.C., Begum, R., Clarke, P.A., Hahne, J.C., Dowsett, M., de Bono, J., Workman, P., Sadanandam, A., Fassan, M., Sansom, O.J., Eccles, S., Starling, N., Braconi, C., Sottoriva, A., Robinson, S.P., Cunningham, D., Valeri, N., 2018. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 359(6378), 920-926. Vogt, N., 2019. Organoids that model the fetal placenta. Nat Methods 16(2), 144. Volkner, M., Zschatzsch, M., Rostovskaya, M., Overall, R.W., Busskamp, V., Anastassiadis, K., Karl, M.O., 2016. Retinal Organoids from Pluripotent Stem Cells Efficiently Recapitulate Retinogenesis. Stem Cell Reports 6(4), 525-538. Wang, P., Mokhtari, R., Pedrosa, E., Kirschenbaum, M., Bayrak, C., Zheng, D., Lachman, H.M., 2017. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in cerebral organoids derived from iPS cells. Molecular Autism 8(1), 11.
Journal Pre-proof
Jo
ur
na
lP
re
-p
ro
of
Wang, Y., Wang, L., Zhu, Y., Qin, J., 2018. Human brain organoid-on-a-chip to model prenatal nicotine exposure. Lab Chip 18(6), 851-860. Watson, C.L., Mahe, M.M., Munera, J., Howell, J.C., Sundaram, N., Poling, H.M., Schweitzer, J.I., Vallance, J.E., Mayhew, C.N., Sun, Y., Grabowski, G., Finkbeiner, S.R., Spence, J.R., Shroyer, N.F., Wells, J.M., Helmrath, M.A., 2014. An in vivo model of human small intestine using pluripotent stem cells. Nat Med 20(11), 1310-1314. Weaver, V.M., Petersen, O.W., Wang, F., Larabell, C.A., Briand, P., Damsky, C., Bissell, M.J., 1997. Reversion of the Malignant Phenotype of Human Breast Cells in Three-Dimensional Culture and In Vivo by Integrin Blocking Antibodies. The Journal of Cell Biology 137(1), 231245. Weeber, F., Ooft, S.N., Dijkstra, K.K., Voest, E.E., 2017. Tumor Organoids as a Pre-clinical Cancer Model for Drug Discovery. Cell Chem Biol 24(9), 1092-1100. Xue, X., Shah, Y.M., 2013. In vitro organoid culture of primary mouse colon tumors. Journal of visualized experiments : JoVE(75), e50210-e50210. Yan, H.H.N., Siu, H.C., Law, S., Ho, S.L., Yue, S.S.K., Tsui, W.Y., Chan, D., Chan, A.S., Ma, S., Lam, K.O., Bartfeld, S., Man, A.H.Y., Lee, B.C.H., Chan, A.S.Y., Wong, J.W.H., Cheng, P.S.W., Chan, A.K.W., Zhang, J., Shi, J., Fan, X., Kwong, D.L.W., Mak, T.W., Yuen, S.T., Clevers, H., Leung, S.Y., 2018. A Comprehensive Human Gastric Cancer Organoid Biobank Captures Tumor Subtype Heterogeneity and Enables Therapeutic Screening. Cell Stem Cell 23(6), 882897.e811. Yin, X., Mead, B.E., Safaee, H., Langer, R., Karp, J.M., Levy, O., 2016. Engineering Stem Cell Organoids. Cell Stem Cell 18(1), 25-38. Yin, Y., Bijvelds, M., Dang, W., Xu, L., van der Eijk, A.A., Knipping, K., Tuysuz, N., Dekkers, J.F., Wang, Y., de Jonge, J., Sprengers, D., van der Laan, L.J.W., Beekman, J.M., ten Berge, D., Metselaar, H.J., de Jonge, H., Koopmans, M.P.G., Peppelenbosch, M.P., Pan, Q., 2015. Modeling rotavirus infection and antiviral therapy using primary intestinal organoids. Antiviral Research 123, 120-131. Yu, F., Choudhury, D., 2019. Microfluidic bioprinting for organ-on-a-chip models. Drug Discovery Today. Yu, F., Hunziker, W., Choudhury, D., 2019. Engineering Microfluidic Organoid-on-a-Chip Platforms. Micromachines (Basel) 10(3). Zhang, Y.S., Arneri, A., Bersini, S., Shin, S.-R., Zhu, K., Goli-Malekabadi, Z., Aleman, J., Colosi, C., Busignani, F., Dell'Erba, V., Bishop, C., Shupe, T., Demarchi, D., Moretti, M., Rasponi, M., Dokmeci, M.R., Atala, A., Khademhosseini, A., 2016. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 110, 45-59. Zomer-van Ommen, D.D., Pukin, A.V., Fu, O., Quarles van Ufford, L.H.C., Janssens, H.M., Beekman, J.M., Pieters, R.J., 2016. Functional Characterization of Cholera Toxin Inhibitors Using Human Intestinal Organoids. Journal of Medicinal Chemistry 59(14), 6968-6972.
Journal Pre-proof Table 1. Summary of various organoids fabricated using different methods
Magneti c Levitati on method using BioAssemb ler plates
Lung Orga noid, Saliv ary Orga noid, adipo se tissue grow th
Organ oid forma tion by using only a scaffo ld such as Matri gel.
200– 250 μm diame ter (Saliv ary organ oids),
The respe ctive tissu e cells
―
―
Crypt Isolatio n, cell dissocia tion and cell culture techniq ue
Intest inal Orga noid
Adul t intest inal stem cells (Lgr 5+ are mark ers)
Produci ng organoi ds by
Intest inal Orga noid
huma n ESCs and
Demonst rated applicati on
Challen ges in using this method ology
―
―
Prompt cell-cell interacti ons and 3D structur es develop ment and accurate ly mirrors the in vivo tissue represe ntation
Current methodeasy and effective.
―
(Daquin ag et al.; Ferreira et al., 2019; Tseng et al., 2013)
Organoi ds were cultured along with their characte ristic features for more than 8 months
Facilitates further studies on gene therapy and regenerati ve medicine
Matrigel (a mouse sarcoma product) , not compati ble with human organ transpla ntations
(Sato et al., 2009)
The cells of the intestin
Facilitates molecular study on human
―
(Spence et al., 2011)
―
―
―
14 days
―
Goblet cells, Paneth cells, enteroen docrine cells and maturati on of brush border are observe d
―
―
Minim um 28 days impro
―
Presence of apical microvil li brush
Enables futuristic possibiliti es of studying inherited kidney diseases
Variabili ty of hPSCs cell lines affects different iation efficienc y and interstiti al space cells of kidney organoid s- not well defined
Referen ce
Each organoid produce d contains multiple nephron structure s
of
kidney organo ids (mass produc tion) within 12 days from NPCs
Advant age of this method
Allows examin ation of cells at each stage of differen tiation. The protoco l is effectiv e for both 2D and 3D organoi ds
ro
0.5 mm in diame ter
Three to six wells of a 24well plate are suffic ient to gener ate 96 organ oids in 3D cultur e.
Cost of the proc ess
-p
Size of the organ oid
Durati on to get the compl ete organ oid
re
hiPS Cs & hES Cs
Jo
3D Cultu re Syste m for Orga noids
Kidn ey Orga noid
Cell Sour ce
Num ber of orga noids
lP
Suspe nsion cultur e syste m for organ oids
96 well ultralow attachm ent plate method for suspens ion cultures
Type of Orga noid Prod uced
na
Meth ods to prod uce organ oids
Comple xity of organoi d – in terms of cells present in the organoi d formed etc.
ur
Orga noid cultu re syste m
Landm ark studies with slight variatio n in the protoco l
(Moriza ne and Bonvent re, 2017)
Journal Pre-proof
The epitheliu m of hGOs exhibits surface mucous cells, contains antral gland cells and endocrin e cells
―
Duct tissu e or singl e cell
―
10-30 organ oids per well
Organ oid forma tion Via Embr yoid Form ation & Spinn ing
Cerebra l organoi d culture system
Retin al organ oid
huma n ESCs
Cere bral Orga noid
Mou se ESCs , ESCs or iPSC of huma ns.
Mainly used to create models to study disease caused by Helicobac tor pylori
―
Allows detailed study of epitheli almesenc hymal interacti ons
To study genetic modificati ons in organoids
Organoi ds produce d have large, continu ous epitheli al structur es
Phenotypi c and molecular heterogen eity build scope for organoido gensis and downstrea m applicatio n
study the cause of hypopla sia seen in microce phaly and can be maintai ned as
The organoid thus obtained can be used as a model to study neurodev elopment and neurologi
―
60 organ oids in total at the end of their study
21 days
―
Robust retinoge nesis is observe d with significa nt appeara nce of CRX cells & photorec eptors
Grow s up to 4mm in diame ter
Two 60m m plate conta ins 32 organ oids in total
8-10 days: basic neural identiti es appear . 20-30 days: define d brain region
$150 per orga noid not inclu ding the biore actor
Histolog ical analysis revealed regions of cerebral cortex, choroid plexus, retina and
ur
Jo
Retinal organoi ds produce d by neuroep ithelial Trisecti on method.
congenita l gut disorders
Ductal markers surround ing the central lumen resembli ng liver buds
re
Liver and Pancr eatic Orga noid
It takes about 1 to 4 weeks for organo ids to develo p.
al organoi ds acquire both secretiv e and absorpti ve functio ns Robust method to generat e hGOs to study stomac h develop ment and mechan isms involve d
of
―
This is a 34 days protoc ol to produc e human gastric organo ids
-p
―
border similar to the ones found in the mature intestine
na
Protoco l to produce both liver and pancrea s organoi d
Gastr ic Orga noid
huma n ESCs and hiPS Cs
ved develo pment observ ed in the organo ids
lP
Gastric Organoi d differen tiation protocol - from spheroi ds to organoi ds
hiPS Cs
ro
manipul ating a series of growth factors
―
Suitable only for adult epithelia l tissue, impedes exponen tial longterm expansio n of tissue in-vitro 1/3 of organoid s degrade d before day 21 and low OV formatio n frequenc y severely affects organoid number produce d. Tissue maturati on and neural & nonneural tissue crosstalks due to lack of surround
(McCra cken et al., 2014)
(Broutie r et al., 2016)
(Volkne r et al., 2016)
(Lancast er and Knoblic h, 2014; Lancast er et al., 2013)
Journal Pre-proof
huma n iPSC s
―
―
33 days
Large polarize d neuroepi thelial surround ing the fluid filled cavities with expressi on of CD133 at the apical surface resembli ng the ventricul ar zone
―
ro
Cere bral Orga noid
meninge s
re
-p
Organoi d produce d combini ng stem cell biology & Organ on chip technol ogy
s to form
―
31 huma n cereb ral organ oids
Organ oids are grown for 9 month s or more to establi sh mature brain feature s
Protoco l to produce mature cerebral organoi ds
Cere bral Orga noid
huma n PSCs
―
cal diseases
ing embryon ic tissue
The organoids thus obtained are a better model to study drug toxicity, brain developm ent and neurologi cal diseases
―
(Wang et al., 2018)
―
Organoi ds produce d exhibits a high number of SCone photorec eptors with typical morphol ogy along with a high expressi on of Soptin
Retinal organoi ds show improv ed maturat ion (Day25 organoi ds resembl ed Day 32 organoi ds of static culture system)
Use of bioreactor accelerate s the productio n, growth and differenti ation of organoids and allows retinal diseases and therapeuti c studies
Retinal organoid s produce d show signs of degener ation after day 25
(DiStefa no et al., 2018)
―
Mature structure s of brain that includes dendritic spinelike structure s are observe d
Maturat ion of organoi ds can be attained by light simulati on of photose nsitive cells
These mature brain organoids have the potential to model brain functions exhibited by adult brain
The role of light in modulati ng the neuronal network and photose nsitivity of cellsyet to be discover ed
(Quadra to et al., 2017)
lP ―
na
mous e PSCs
ur
Retin al organ oid
Appea rance of optic cup, increas e in growth till 25th day.
Jo
Retinal organoi ds producti on using rotating wall vessel bioreact or
such for over a year (around 15 months) Minima l cell death in the core of the brain organoi d observe d via TUNEL Assay indicati ng high viabilit y of brain organoi d on chip with improv ed maturat ion
of
Biore actor
Journal Pre-proof
―
7-day monol ayer culture . 18 days 3 D culture to genera te kidney organo ids
―
Inducti on all the four types of progeni tors essentia l to generat e kidney organoi d that highly matches the first trimeste r human kidney.
Robust renal model to study human kidney developm ent, diseases & nephrotox ic drug screening and
Organoi ds remaine d healthy over a very long period (over 5 months)
Provides avenues to study developm ent and response to injury of human CNS
Organoi d culture conditio ns require further optimiza tion to reach Maturati on
(Takasat o et al., 2016)
―
(Giando menico et al., 2018)
―
-p
1-3 organ oids per mold
ur
―
re
huma n ESCs
lP
Cere bral Orga noid
Appro x 5060 and in some cases 90 days
ro
of
hPS Cs and hiPS Cs
na
Cerebra l organoi d generat ed using airliquid interfac e method
Kidn ey Orga noid
Jo
Organ oid produ ction using AirLiqui d interf ace Meth od
Differe ntiation Protoco l to produce kidney organoi d
6mm in diame ter at the end of 18th day of organ oid cultur e
Collecti ng ducts, loops of Henle, proxima l tubules, early mesangi al & distal tubules, glomeru li, tubular structure s and nephron s are observe d. Remark able selforganiza tion of callosal and corticof ugal tracts observe d
Journal Pre-proof Captions for Figures and Tables
Figure 1: Summary of the methods for fabrication of organoids Fig 2: Suspension culture system for fabrication of organoids Fig 3: Crypt isolation culture system: Isolation of crypt from primary intestine tissues Fig 3A: Crypt isolation culture system: Generating organoids from isolated crypts Fig 4: Cerebral organoid culture system. Fig 5: Air-liquid interface method for fabrication of organoids
Jo
ur
na
lP
re
-p
ro
of
Fig 6: Summary of factors crucial for the manufacturing of organoids
Figure 1
Figure 2
Figure 3A
Figure 3B
Figure 4
Figure 5
Figure 6