CC BY 4.0 · Glob Med Genet 2022; 09(04): 268-276
DOI: 10.1055/s-0042-1756662
Review Article

Advances in Organoid Culture Research

Zhiyuan Xie
1   Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, People's Republic of China
,
Linghao Wang
1   Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, People's Republic of China
,
Yan Zhang
1   Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, People's Republic of China
2   State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
› Author Affiliations
 

Abstract

Organoids are powerful systems to facilitate the study of individuals' disorders and personalized treatments because they mimic the structural and functional characteristics of organs. However, the full potential of organoids in research has remained unrealized and the clinical applications have been limited. One of the reasons is organoids are most efficient grown in reconstituted extracellular matrix hydrogels from mouse-derived, whose poorly defined, batch-to-batch variability and immunogenicity. Another reason is that organoids lack host conditions. As a component of the tumor microenvironment, microbiota and metabolites can regulate the development and treatment in several human malignancies. Here, we introduce several engineering matrix materials and review recent advances in the coculture of organoids with microbiota and their metabolites. Finally, we discuss current trends and future possibilities to build more complex cocultures.


#

Introduction

For decades, cancer cell lines have been the most common model used in vitro and have been successful in many areas, such as exploring the mechanism of cellular signaling pathways and researching potential drug targets.[1] Nevertheless, cell line models could not maintain the genetic phenotype and heterogeneity of the original tumor cells, let alone reproduce the morphology and function of the original tumor tissue.[2] Genetically engineered mouse models and patient-derived tumor xenograft (PDX) generated in mice are important advances in mimicking tumor niche, they can largely maintain the heterogeneity of the original tumor and the interaction between tumor and its surrounding stroma.[3] However, the establishment of the PDX model takes time, the genetic operation is difficult, and the cost of molding is expensive, which is not conducive to high-through drug screening. With a higher rate of success, shorter cycles, and better cost performance, organoids can make up for the defects of the above models and they more closely resemble the original tumor in vivo.

Organoids refer to tissue analogs with a certain spatial structure formed by the three-dimensional (3D) culture of adult stem cells or pluripotent stem cells in vitro. They can simulate the cell compositions, physiological functions, and genetic characteristics of tissues and organs in vivo to the greatest extent, as well as can be long-term stable subculture.[4] Organoid culture relies on basement membrane extract (BME), like Matrigel and collagen. However, these materials have poor physical and chemical properties, unclear composition, and uncontrollable stability between batches. In contrast, the physical and chemical properties of engineering matrix materials are relatively controllable, which is more conducive to organoid culture through adjustment and optimization.

Studies have shown that microbiota can reside within tumor cells and immune cells, impact the state of the tumor immune microenvironment, and even some microbial metabolites can also enter the bloodstream and modulate cancer cells and immune cells.[5] The introduction of microbiota or microbial metabolites into organoid culture can better reproduce the tumor microbe environment in vivo and study the occurrence and development of tumors more comprehensively. A deeper understanding of the microbiota-tumor-host relationship may also provide new prospects for cancer treatment and contribute to the development of new anticancer drugs.

In this review, we discuss the application of organoid in cancer research and highlight several engineering materials which have biomaterial properties for culturing organoids. Then, we illustrate the complex relationship between microbiota and cancer, and importantly, introduce how organoids can be used in the study of microbiota-tumor-host interactions.

Organoids as a Model to Study Cancer

As early as 2009, a minigut organoid culture system was successfully established by adding R-spondin-1, epidermal growth factor, and Noggin into the Matrigel of mouse Lgr5+ intestinal stem cells, and intestinal organs with crypt-villus structures derived from adult stem cells were established based on this culture system.[6] The successful attempt and application of organoids in normal tissue make organoid culture technology develop rapidly in the field of tumor research. At present, various organoids have been successfully cultivated, including the intestine, liver, stomach, brain, kidney, heart, and skin.[7] [8] [9] [10] [11]

The organoid model has a similar tissue arrangement and spatial structure to the donor organ and can simulate partial functions of the donor organ. It provides a research platform more similar to the environment in vivo for tumor research in vitro. The organoid model has become a new research tool for scientific problems that are difficult to be solved by traditional models, such as cancer genetics, cancer processes, and the activity of antitumor drugs. Organoid technology has broad application prospects in basic and clinical research, especially in disease pathology, cell biology, precision medicine, and drug toxicity.[12]

Organoids allow the long-term expansion of normal and tumor tissues, and greatly preserve genetic diversity and morphological stability. There was heterogeneity in organoids cultured from tissues of different patient sources.[13] For the gastrointestinal and colorectal cancer (CRC) organoids, they show great similarity with the respective biopsies in morphology, the mutational spectrum, genes with an altered copy number, and expression patterns of common clinical diagnosis markers,[14] as well as they can be accurately reproduced in histopathology, hormone receptor status, and deoxyribonucleic acid (DNA) copy number variations.[15] This indicates that organoids can be used to study cell heterogeneity, construct biobanking, high-throughput drug screening, drug sensitivity test, etc., laying a foundation for the further development of personalized therapy and precision medicine.

Cancer is caused by the gradual accumulation of pathogenic gene mutations. Therefore, it is important to understand the mutational processes active in tissue homeostasis and tumorigenesis. Combined with CRISPR-Cas9 and orthotopic xenotransplantation in mice, cancer progression can be successfully regenerated in healthy tissue organoids,[16] [17] the pathogenic genes and the mechanism of signaling pathways related to cancer progression can be explored by studying the progression of precancerous lesions.[18] [19] Much effort has been exerted to model tumorigenesis and progression, to further elucidate the molecular mechanisms using tumor organoids.

However, compared with the physiological environment of normal tissues and organs, organoids still have limitations, such as a lack of connective tissue, vascular system, and immune cell. With the gradual development of precision medicine, it is believed that tumor organoids can be further developed to simulate tumors in vivo to a greater extent and become a powerful tool for personalized tumor therapy.


#

Engineering Materials for Organoid Systems

Current organoid systems mostly rely on intrinsic or extrinsic biochemical signals (e.g., growth factors) and cell-cell interactions to control stem cell fate.[20] Organoid culture methods are complicated by the nearly exclusive dependence on animal-derived hydrogels, including Matrigel[21] and collagen,[22] as the 3D matrix. As a material derived from the secretion of Engelbreth–Holm–Swarm mouse sarcoma cells and enriched for biomimetic extracellular matrix (ECM) proteins, Matrigel is an ill-defined gelatinous basement membrane protein mixture composed of laminin, collagen IV, entactin, heparan sulfate, and numerous growth factors.[21] These matrices feature complex and variable compositions that are not conducive to controlled modifications, they can cause large batch-to-batch variations in cultured organoids. There are also potential risks of immunogen[23] and pathogen transfer,[24] making them unsuitable for organoid expansion in downstream clinical applications. In addition, Matrigel differed greatly from the ECM environment of normal tissues and organs, which could not provide a tissue-specific ECM environment for cells.[25] Also Matrigel is of high cost. Based on the above points, researchers aim at finding a replacement for Matrigel.

Natural polymer-based hydrogels are favored for their similarity to human ECM and their inherent biological activity.[26] Natural hydrogels are polysaccharides, proteins, and animal-derived mixtures. Polysaccharides are thought to be suitable for creating complex 3D scaffolds with multiple forms because of their rapid gelation properties and biodegradability. The typical polysaccharides are alginate, chitosan, dextran, cellulose, hyaluronic acid, agarose, pectin, gelatin, and heparin. Most natural hydrogels lack adequate mechanical strength, as well as a single type of hydrogel cannot meet all the requirements of organoids due to the complex and dynamic microenvironments in different tissues/organs in vivo. Therefore, many researchers believe that the combination of natural and synthetic hydrogels is a better choice for organoid culture.

Engineering synthetic hydrogels are combined with signaling proteins such as integrin-binding small molecules or bioactive peptides via chemical/enzymatic crosslinking for promoting the growth of stem cells and organoids. Synthetic hydrogels can provide chemically defined matrix, precisely adjust for matrix properties, and improve organoid culture efficiency and consistency. Synthetic hydrogels are easily produced with minimal batch variation by using traditional synthetic methods.[27] A variety of tumor organoids have been successfully cultured in engineering synthetic hydrogels. The common synthetic polymers for organoid culture are polyethylene glycol, nanocellulose, and other polymer matrices such as polyisocyanopeptide (PIC).[28] The development of materials capable of spatially and dynamically controlling organoid microenvironment and surrounding matrix makes it possible to regulate organoid maturation and function.

Nanocellulose-Based Hydrogel

Nanocellulose is a kind of excellent synthetic ECM suitable for organoid growth because of its hydrophilic, renewable, nontoxic, biodegradable, and biocompatible properties, as well as superior mechanical strength and modifiable surface.[29] Cellulose, as the precursor material of cellulose hydrogel, are biological macromolecules formed by glucose molecules connected by β-1,4-glycoside bonds. Cellulose molecules contain a large number of hydrogen bonds, which makes it a temperature-sensitive material. At low temperature, cellulose only form simple entanglement around hydrophobic groups without polymerization, but as the temperature increases, hydrogen bonds are destroyed, and their hydration gradually loses. The strong hydrophobic interaction between cellulose molecules leads to the formation of a 3D network.[30]

Engineering nanocellulose hydrogel represents a performant and sustainable alternative for the organoid growth, and contributes to significantly reducing the costs in studies against diseases of global concern such as cancer. It has been reported that biologically active plant-based nanocellulose hydrogels functionalized with Arg-Gly-Asp (RGD) peptides and glycine supported the mouse intestinal organoids culture,[31] and can also be used for the breast cancer patient-derived organoids (PDOs) initiation and growth.[32]

The proliferation and cell differentiation of organoids mainly depend on whether the hydrogel in which they are located can reconstruct the microenvironment of the base membrane.[33] Nanocellulose hydrogels are made of 99.9% water and 0.1% nanocellulose fibers by crosslinking with similar mechanical properties to the standard animal-based matrix, but do not provide any biological signal. Interestingly, they can be modified and functionalized due to the chemically reactive group in surface.

RGD peptides are the adhesive sites, as a fundamental element in matrices, which can induce cell attachment and differentiation.[34] Glycine, a nonpolar amino acid, is dissolved in the hydrogel to increase its osmolality, once the osmolality of the matrix is balanced by GLY, small intestinal crypts progress into cystic organoids. Based on this, organoids cultured in the RGD-GLY nanocellulose hydrogel sustain high viability, and even organoids can be formed from single cells, although the growth rate is considerably slower than those embedded in Matrigel.[31] When nanocellulose is blended with collagen (COL-NC) instead of RGD-GLY, intestinal crypts can form organoids with cystic structures, intact lumen, sphere-like morphology, and prominent epithelial budding.[35] PDOs grown in nanocellulose hydrogel synthesized by gelatin show similar proliferation, histopathologic features, gene expression, and drug responses to original tumors and PDOs formed in standard basement-membrane extract.[32]

Besides the biological signal in hydrogels, the matrix stiffness also affects the formation and development of organoids, the mechanical properties of the hydrogels must resemble the stiffness of organs and tissues.[36] The stiffness of hydrogels is proportional to the type of solids and affected by the multiple cross-linkers and their concentration.[37] For example, the stiffness of COL-NC hydrogel is 90 Pa as same as Matrigel, while COL gels are much lower at 35 Pa[35]; RGD-TPON hydrogels form a 3D network and are closer to that of Matrigel than RGD-PON hydrogels, which offer an optimum bioactive surface and appropriate stiffness for organoid attachment and growth.[37] Usually, nanocellulose hydrogels cross-linked with MgCl2 and CaCl2, require four times more Mg2+ than Ca2+ ions to reach the stiffness of Matrigel.[37] The number of organoids in calcium cross-linked hydrogels sustains a massive decrease than the hydrogels cross-linked with Mg2+ ions. It is important to select the right matrix materials and the type of cross-linkers agent to achieve the stiffness of a specific organ or tissue.

The modification of nanocellulose by physical, chemical, or biological methods can obtain suitable properties for organoid morphogenesis in vitro, which may fill the key gap of organoid technology lacking matrix glue.


#

Nonadhesive Alginate Hydrogels

Alginate is Food and Drug Administration-approved polysaccharide derived from algae and it is a favorable 3D culture material due to its biocompatibility and ease of manipulation with gelation and viscoelastic properties. Alginate hydrogels are structurally similar to the extracellular matrices of living tissue and can be prepared by a variety of chemical/enzymatic crosslinking methods.[38]

Capeling et al[39] demonstrated that nonadhesive alginate hydrogel can support human intestinal organoid (HIO) growth in vitro. Hydrogels prepared by mixing alginate and gelatin can also exist in culture medium as suspended capsules for the cultivation of liver or intestinal organoids, and the mechanical and biological properties of this material are similar to those of tissue in vivo.[40] [41] Alginate-grown HIOs are highly similar to Matrigel-grown HIOs in vitro, and the differentiation of epithelial cells is indistinguishable from HIOs grown in Matrigel.[39] Organoids cultured in suspended alginate hydrogels are closer to real tissues in vivo, as suspended capsules can effectively exchange nutrients with the surrounding medium to support the metabolism of multicellular clusters.[40] HIOs cultured in nonadherent alginate suspended capsules can form a serosal mesothelium that resembles the human fetal intestine.[41]

Differentiation of stem cells is regulated by the stiffness of cell medium substrates.[42] Nonalginate hydrogel adjusts the mechanical strength of hydrogel by adjusting the proportion of compound composition and matching the mechanical strength of corresponding tissues in vivo, so as to be suitable for the culture of different types of organoids.[43] Nonadhesive alginate hydrogel can also form microbeads structure through microfluidic droplet devices, which can establish a high-throughput organoid model convent for drug screening.[44]

Nonadhesive alginate hydrogel becomes a simple system and conducive to large-scale production due to a lack of adhesive or biochemical clues. It is a pity that the yield of alginate-grown organoids was significantly lower than in Matrigel.[39] At the same time, alginate cannot be degraded, which is not conducive to the recovery and passage of organoids. However, these shortcomings can be improved by adhesive/degradable modification. Alginate is also cost-effective. In conclusion, nonadhesive alginate hydrogel can be used to support 3D culture systems and advance regenerative medicine.


#

PIC-Based Hydrogels

PIC is a synthetic polymer and nonimmunogenic material, it can form thermosensitive hydrogels. It is a free-flowing liquid below 16°C, and the liquid becomes a viscous hydrogel within minutes when the temperature is above 16°C.[45]

Like nanocellulose, PIC alone was not sufficient to support cell attachment or induce proliferation due to the lack of any bioactive component. When PIC hydrogels modify the adhesive RGD peptide, it can allow mammary gland organoids formation from breast fragments or purified single mammary epithelial cells[46]; PIC hydrogels can also culture human liver organoids after modification with laminin-111.[47] The expansion and differentiation rate of organoids in PIC-based hydrogel is similar to that of Matrigel[48]; while the potential for proliferation and differentiation could be maintained for many generations.[47] In addition, the proliferation rate depends on hardness, and lower hardness is the best condition for the proliferation of organoids.[47] Hydrogel hardness can also control colony formation efficiency.[46]

PIC-based hydrogel is a chemically defined and synthetic hydrogel. The thermally reversible properties make it easier for cell recovery during organoid culture and make it beneficial for clinical applications such as cell therapy or tissue engineering. For example, PIC hydrogels have also been applied in vivo for subcutaneous cell transplantations and wound healing studies without any adverse effects.[49] [50] These results highlight that cultured organoid combinations using well-defined synthetic hydrogels will pave the way for clinical applications in humans in the near future.


#

ECM Hydrogels Derived from Decellularized ECM

Decellularized tissue engineering is derived from the concept of tissue engineering. Its strategy is to use decellularization scaffolds of healthy organs as scaffold materials and then implant seed cells, such as stem cells, to induce the formation of organoids in a 3D culture system in vitro.[51] Decellularized matrix materials have attracted much attention in recent years because of their good biocompatibility, biodegradability, and tissue regeneration ability.[52] So far, reported in the article of the decellularized ECM (dECM) materials include skin,[53] muscle,[54] cartilage,[55] bone,[56] blood vessels,[57] lung,[58] liver,[59] kidney,[60] small intestine,[61] bladder,[62] trachea,[63] tissue dECM, etc. Some of them have been used in clinical treatment and have achieved good clinical efficacy.

Porcine gastric decellularized matrix (SEM) and intestinal decellularized matrix (IEM) hydrogel can culture gastric and intestinal (GI) organoids, respectively.[64] Hydrogels derived from healthy porcine liver ECM (PLECM) or human liver ECM (HLECM) can replace mouse tumor-derived BME for the culture and expansion of intrahepatic cholangiocyte organoids.[65] SEM and IEM hydrogels could effectively cultivate GI organoids with similar structure and functional characteristics as Matrigel organoids, even though some indexes were closer to natural tissues and organs,[64] let alone organoids in dECM hydrogels can be long-term culture, passage, and freezing storage. LECM hydrogels support the proliferation of bile duct cell-like organoids and maintain a bile duct cell-like phenotype, and no species-specific effects were observed between HLECM or PLECM hydrogels.[65]

Although more and more dECM materials are being used in clinical therapy, dECM faces many challenges in its applications, including optimal decellularization schemes, degradation of materials, and regeneration regulation. The organoids cultured in dECM hydrogel had a low proliferation rate, and the intestinal villi/crypt structures of IEM are less than the organoids cultured in Matrigel.[64] But these limitations could be optimized by further improving protein retention other than collagen after decellularization.[66] So far, some cell fragments and nuclear residues still exist in the prepared dECM materials, even the dECM materials that have been commercialized and widely used in clinical practice. At the same time, nondegradation or incomplete degradation can hinder the growth of regenerative tissues and organs.

In summary, engineering matrices offer the possibility of customizing biochemical and mechanical properties according to organoid type and application that can support several different types of organoid cultures. This will become increasingly important once multiple organoid types need to be grown and maintained in a single cell culture system. However, these synthetic and highly defined hydrogels are still in their infancy. They can only be used in certain applications and are still not a complete replacement for widely used commercially available natural matrices.


#
#

The Microbiota, Microbial Metabolites in Organoids

As understanding of the microbiota grows, we gradually understand the microbiota and microbial metabolites within mammalians. According to the classification of natural attributes, the microbiota within human has been identified in dozens of bacteria phyla, including Bacteroidetes, Firmicutes, Proteobacteria, Actinobacteria, Verrucomicrobia, Fusobacteria, and so on.[67] Ninety-eight percent of microbiota can be classified into the first four groups. Microbiota ferments dietary fiber and carbohydrates liberated from host mucins, and produces various fatty acids and amino acids through different metabolic pathways.[68] In addition, some gases and nitrogenous compounds are produced, particularly N-nitroso compounds. These metabolites accumulate in the bloodstream and have systemic effects on the host both protective and detrimental.[69] Accumulating evidence suggests that microbiota and microbial metabolites are involved in numerous biological processes, such as transformation process, tumor progression, and the response to anticancer therapies including immune checkpoint blockade.[70] [71] [72] [73] [74] [75] [76]

To better dissect host-microbiota interactions, it is necessary to adopt models that mimic health and disease features as closely as possible. Traditional in vitro and in vivo experimental models are difficult to reproduce the host-microbiota relationship as represented by humans. Therefore, it is crucial to establish high-quality experimental models to understand the crosstalk of host, tumor, microbiota, and microbial metabolites. At present, many studies have reconstructed the complex tumor microenvironment in vivo by fusing immune cells, mesenchymal cells, and endothelial cells in organoids. If microbiota is fused, the complex physiological and pathological conditions of microbiota-tumor-host can be better simulated and the influence mechanisms of microbiota and their metabolites on tumor progression can be better studied.[77]

Protumorigenic Microbiota and Microbial Metabolites

Microbiota can regulate some signaling pathways, affect the invasion and metastasis of tumor cells, and promote the occurrence and development of tumor. The Wnt/β-catenin and PI3K/AKT signaling pathway are involved in a wide range of biological processes, including cell migration, proliferation, differentiation, apoptosis, and immune response.[78] [79] The alteration of these signaling pathways is a major cause of carcinogenesis. Fusobacterium nucleatum can activate the Wnt/β-catenin pathway through the upregulation of cyclin-dependent kinase 5 (Cdk5), and promote tumor cell proliferation and migration.[80] Peptostreptococcus anaerobius could drive CRC through increasing the gene expression and protein level of PI3K and AKT.[81]

Microbial metabolites can also regulate tumor cell growth and affect the differentiation of immune cells. For example, formate can drive tumor progression and invasion by triggering AhR signaling, and regulating cancer stemness.[82] Butyrate may locally or systemically regulate the balance of anti-inflammatory and proinflammatory cytokines, and disrupt the ratio of regulatory T cells and Thelper17 cell subsets.[83] [84] The high concentrations of secondary bile acids can be produced with gut microbiota enzyme and induce CRC.[85] [86] And gut microbiota can use bile acids as a messenger to alter immune function and influence antitumor immunosurveillance.[87] Polyamines are mainly metabolized from arginine in host tissues, its synthesis also occurs in gut microbiota.[88] Polyamines are involved in a range of essential physiological functions, such as the maintenance of the structural integrity of membranes and nucleic acids, gene regulation, and translation.[89] [90] Enterotoxigenic Bacteroides fragilis can upregulate polyamine production by host cells,[90] and affect the activity of various signaling pathways in tumor cells.[91]


#

Antitumorigenic Microbiota and Microbial Metabolites

In addition to promoting tumor, most microbiota and their metabolites also inhibit tumor growth. A cocktail of Lactobacillus spp inhibits tumor cell growth via downregulating Wnt/β-catenin target genes.[92] Motility-associated killing factor A (MakA), a cytotoxin from Vibrio cholera, can induce cell apoptosis in several cancer cell lines,[93] like ileocecal cancer and colon carcinoma, and it can inhibit β-catenin-mediated tumor cell proliferation and reduce tumor burden via altering β-catenin integrity. Leuconostoc mesenteroides, isolated from dairy products, induced apoptosis and DNA fragmentation in a CRC cell line, as well as downregulated AKT in cells treated with bacterial conditioned media.[94] Some microbiota can induce tumor cells death by affecting the redox balance of tumor cells and upregulating genes related to oxidative stress response.[95] Indoles and their derivatives can regulate the differentiation of immune cells and increase the tolerance of the immune system.[96] [97]

Immune checkpoint block (ICB)-mediated antitumor responses mainly depend on cytotoxic T cells capable of recognizing and killing tumor cells. ICB has greatly improved the clinical efficacy of malignant tumor therapy and has great potential to be explored.[98] [99] However, the immune response efficiency of ICB therapy is at a low level due to individual differences of patients.[100] It is necessary to find new ways to improve ICB response. More and more evidence indicate microbiota can improve the efficiency of ICB treatment efficiency.[101] [102] For example, different types of Bacteroides may affect anticancer immunotherapy with CTLA-4 blockade.[103] Bifidobacterium and Akkermansia muciniphila can promote antitumor immunity and improve the therapeutic effect of anti-PD-L1 and anti-PD-1.[71] [104] Microbial metabolites can also enhance the antitumor immune response and improve the immunotherapy response, such as inosine, polypeptide, and L-arginine.[105] [106] [107] [108]


#

Coculture of Microbiota and Organoids

At present, most research models of microbiota are based on germ-free mice,[109] which have two defects: first, the cells and microbiota of this model are all from mice, which are different from human species; Second, due to the extremely complex microbiota and its effects, mouse models cannot be finely controlled in the experimental process. Therefore, it is difficult to obtain more detailed and in-depth results of mechanism properties. Organoids can be used to establish in vitro research models of complex microbial communities in direct contact with the mucus layer of human intestinal cells, complementing the significant deficiencies of mouse models in experimental control, scalability, and reproducibility of interactions between the human gut and host-specific symbiotic microbiota. It is extremely valuable for further study of the interaction between host and microbiota.

The coculture technology of organoids with microbiota has been used widely and made substantial progress in disease research. Engevik et al[110] found colony-stimulating factor (CSF) induce interleukin-10 expression in dendritic cells and regulate the host immune system by coculture of Lactobacillus reuteri, organoids, and dendritic cells. A genotoxic polyacetyl-1-peptide (colibactin) in PPKS+ Escherichia coli that causes DNA double-strand cross-linking and damage which is associated with CRC,[111] this study provides important evidence for further treatment and research of CRC through studying the direct effect and causal relationship between gut microbiota genotoxic substances and oncogene mutations. To investigate how the tumor-resident microbiota influence the tumor cell activity, researchers established a coculture system of organoids with microbiota,[112] organoids imaging results showed that microbiota help tumor cells survive in the circulatory system by regulating the cytoskeleton, thereby promoting tumor metastasis.

Systematic protocols have been developed to guide the coculture of intestinal organoids with microbiota.[113] Organoids can form lumens that represent the intestinal lumen, microbiota was injected into lumen by microinjection and represent faithfully the microbiota–epithelium orientation. This method was successfully used to inoculate human gastric organs with Helicobacter pylori.[114] Microinjection is a technical skill, it will be failed if the organoids are too small, too differentiated, too dense, or the microbes are too overgrown, and most of the intestinal organoids have to be as similar in size as possible. This operation also does not allow for high-throughput scaling, one of the advantages is that it is more in line with the actual physiological state of the body.

Importantly, organoids coculture with single microbiota does not take into account the overall complexity of the gut microbiota ecosystem, such as complex interactions with immune compartments. The intestinal organoids and organs-on-chips[115] approach has made significant progress and can achieve long-term coculture with a variety of microbiota species. Jalili-Firoozinezhad et al[116] developed a two-channel microfluidic organoid chip device, Intestine Chip System, the system has the capability of maintaining a complex microbiota community in coculture with human intestinal epithelial cells, and to study host-microbiota interactions through direct contact with human intestinal epithelial cells via an in vitro covering simulated mucous layer. Compared with aerobic culture conditions, microfluidic chip-controlled chamber of oxygen gradient increased the intestinal barrier function and provides maintenance of microbiota diversity related to physiological levels, including more than 200 from 11 different genera classification of obligate anaerobic bacteria. Microfluidic organoid chip can be used as a tool to develop microbiota-related therapies, probiotics, and nutrients.


#
#
#

Conclusion

As 3D micro-organs cultivated in vitro, organoids have the potential to construct human organ disease models. They are recognized as important tools in biological research and can make up for the shortcomings of cell lines and animal models in clinical application. Organoid research is still in its infancy and there are many problems to be solved. For example, animal-derived matrix materials used to support organoid growth are not conducive to organoid expansion in downstream clinical applications due to their complex and variable components, differences between batches, and risks of immunogen and pathogen transfer. Engineering hydrogels can be synthesized using traditional methods to provide chemically well-defined matrices that can be precisely tuned to improve organoid culture efficiency and consistency. Compared with Matrigel, the engineering materials currently developed have a slightly lower efficiency in organoid culture, but they still have broad development and application prospects.

Microbiota and microbial metabolites play a key role in human health and disease. Different types of microbiota and microbial metabolites play different roles in the occurrence and development of tumors, immunology, and cancer therapy. But it is still having a lot to learn for the internal mechanism and how to adjust the microbiota to enhance response to cancer immunotherapy. Traditional experimental models cannot well simulate the relationship between host and microbiota, so it is difficult to obtain more detailed and in-depth results of mechanism properties. To better analyze the host-microbiota interaction, microbiota and organoid technology can be combined to reconstruct the complex tumor microenvironment in vivo, providing new details for studying the occurrence and development mechanism of various diseases including cancers. At present, the application of microbiota and organoid coculture technology has played an important role in the study of some diseases, opening up new possibilities for understanding and treating diseases. However, this coculture model is still in its infancy and there is much more space for improvement in the future.


#
#

Conflict of Interest

None declared.

  • References

  • 1 Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol 2020; 21 (10) 571-584
  • 2 Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer 2018; 18 (07) 407-418
  • 3 Jin K, Teng L, Shen Y, He K, Xu Z, Li G. Patient-derived human tumour tissue xenografts in immunodeficient mice: a systematic review. Clin Transl Oncol 2010; 12 (07) 473-480
  • 4 McCauley HA, Wells JM. Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish. Development 2017; 144 (06) 958-962
  • 5 Nejman D, Livyatan I, Fuks G. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 2020; 368 (6494): 973-980
  • 6 Sato T, Vries RG, Snippert HJ. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009; 459 (7244): 262-265
  • 7 Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 2014; 345 (6194): 1247125
  • 8 Huch M, Dorrell C, Boj SF. et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 2013; 494 (7436): 247-250
  • 9 Barker N, Huch M, Kujala P. et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 2010; 6 (01) 25-36
  • 10 Lancaster MA, Renner M, Martin CA. et al. Cerebral organoids model human brain development and microcephaly. Nature 2013; 501 (7467): 373-379
  • 11 Xia Y, Nivet E, Sancho-Martinez I. et al. Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor-like cells. Nat Cell Biol 2013; 15 (12) 1507-1515
  • 12 Li M, Izpisua Belmonte JC. Organoids - preclinical models of human disease. N Engl J Med 2019; 380 (06) 569-579
  • 13 Kopper O, de Witte CJ, Lõhmussaar K. et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med 2019; 25 (05) 838-849
  • 14 Vlachogiannis G, Hedayat S, Vatsiou A. et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018; 359 (6378): 920-926
  • 15 Sachs N, de Ligt J, Kopper O. et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 2018; 172 (1-2): 373-386.e10
  • 16 Matano M, Date S, Shimokawa M. et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med 2015; 21 (03) 256-262
  • 17 Drost J, van Jaarsveld RH, Ponsioen B. et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 2015; 521 (7550): 43-47
  • 18 Miura A, Yamada D, Nakamura M. et al. Oncogenic potential of human pluripotent stem cell-derived lung organoids with HER2 overexpression. Int J Cancer 2021; 149 (08) 1593-1604
  • 19 Fumagalli A, Drost J, Suijkerbuijk SJE. et al. Genetic dissection of colorectal cancer progression by orthotopic transplantation of engineered cancer organoids. Proc Natl Acad Sci U S A 2017; 114 (12) E2357-E2364
  • 20 Yin X, Mead BE, Safaee H, Langer R, Karp JM, Levy O. Engineering stem cell organoids. Cell Stem Cell 2016; 18 (01) 25-38
  • 21 Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol 2005; 15 (05) 378-386
  • 22 Jabaji Z, Brinkley GJ, Khalil HA. et al. Type I collagen as an extracellular matrix for the in vitro growth of human small intestinal epithelium. PLoS One 2014; 9 (09) e107814
  • 23 Liu H, Bockhorn J, Dalton R. et al. Removal of lactate dehydrogenase-elevating virus from human-in-mouse breast tumor xenografts by cell-sorting. J Virol Methods 2011; 173 (02) 266-270
  • 24 Peterson NC. From bench to cageside: risk assessment for rodent pathogen contamination of cells and biologics. ILAR J 2008; 49 (03) 310-315
  • 25 Henke E, Nandigama R, Ergün S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci 2020; 6: 160
  • 26 Kaur S, Kaur I, Rawal P, Tripathi DM, Vasudevan A. Non-matrigel scaffolds for organoid cultures. Cancer Lett 2021; 504: 58-66
  • 27 Kratochvil MJ, Seymour AJ, Li TL, Paşca SP, Kuo CJ, Heilshorn SC. Engineered materials for organoid systems. Nat Rev Mater 2019; 4 (09) 606-622
  • 28 Magno V, Meinhardt A, Werner C. Polymer hydrogels to guide organotypic and organoid cultures. Adv Funct Mater 2020; 30 (48) 2000097
  • 29 Heidarian P, Kaynak A, Paulino M, Zolfagharian A, Varley RJ, Kouzani AZ. Dynamic nanocellulose hydrogels: recent advancements and future outlook. Carbohydr Polym 2021; 270: 118357-118357
  • 30 Zhu L, Zhang Y-Q. Postoperative anti-adhesion ability of a novel carboxymethyl chitosan from silkworm pupa in a rat cecal abrasion model. Mater Sci Eng C 2016; 61: 387-395
  • 31 Curvello R, Kerr G, Micati DJ. et al. Engineered plant-based nanocellulose hydrogel for small intestinal organoid growth. Adv Sci (Weinh) 2020; 8 (01) 2002135
  • 32 Prince E, Cruickshank J, Ba-Alawi W. et al. Biomimetic hydrogel supports initiation and growth of patient-derived breast tumor organoids. Nat Commun 2022; 13 (01) 1466
  • 33 Meran L, Baulies A, Li VSW. Intestinal stem cell niche: the extracellular matrix and cellular components. Stem Cells Int 2017; 2017: 7970385-11
  • 34 Wang H, Cui J, Zheng Z. et al; Assembly of RGD-Modified Hydrogel Micromodules into Permeable Three-Dimensional Hollow Microtissues Mimicking in Vivo Tissue Structures. Assembly of RGD-modified hydrogel micromodules into permeable three-dimensional hollow microtissues mimicking in vivo tissue structures. ACS Appl Mater Interfaces 2017; 9 (48) 41669-41679
  • 35 Curvello R, Alves D, Abud HE, Garnier G. A thermo-responsive collagen-nanocellulose hydrogel for the growth of intestinal organoids. Mater Sci Eng C 2021; 124: 112051-112051
  • 36 Guimarães CF, Gasperini L, Marques AP, Reis RL. The stiffness of living tissues and its implications for tissue engineering. Nat Rev Mater 2020; 5 (05) 351-370
  • 37 Curvello R, Garnier G. Cationic cross-linked nanocellulose-based matrices for the growth and recovery of intestinal organoids. Biomacromolecules 2021; 22 (02) 701-709
  • 38 Lee KY, Mooney DJ. Alginate: properties and biomedical applications. Prog Polym Sci 2012; 37 (01) 106-126
  • 39 Capeling MM, Czerwinski M, Huang S. et al. Nonadhesive alginate hydrogels support growth of pluripotent stem cell-derived intestinal organoids. Stem Cell Reports 2019; 12 (02) 381-394
  • 40 Dong H, Li Z, Bian S. et al. Culture of patient-derived multicellular clusters in suspended hydrogel capsules for pre-clinical personalized drug screening. Bioact Mater 2022; 18: 164-177
  • 41 Capeling MM, Huang S, Childs CJ. et al. Suspension culture promotes serosal mesothelial development in human intestinal organoids. Cell Rep 2022; 38 (07) 110379-110379
  • 42 Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell 2006; 126 (04) 677-689
  • 43 Cassel de Camps C, Aslani S, Stylianesis N. et al. Hydrogel mechanics influence the growth and development of embedded brain organoids. ACS Appl Bio Mater 2022; 5 (01) 214-224
  • 44 Fang G, Lu H, Rodriguez de la Fuente L. et al. Mammary tumor organoid culture in non-adhesive alginate for luminal mechanics and high-throughput drug screening. Adv Sci (Weinh) 2021; 8 (21) e2102418
  • 45 Deshpande SR, Hammink R, Nelissen FHT, Rowan AE, Heus HA. Biomimetic stress sensitive hydrogel controlled by DNA nanoswitches. Biomacromolecules 2017; 18 (10) 3310-3317
  • 46 Zhang Y, Tang C, Span PN. et al. Polyisocyanide hydrogels as a tunable platform for mammary gland organoid formation. Adv Sci (Weinh) 2020; 7 (18) 2001797
  • 47 Ye S, Boeter JWB, Mihajlovic M. et al. A chemically defined hydrogel for human liver organoid culture. Adv Funct Mater 2020; 30 (48) 2000893
  • 48 Sorrentino G, Rezakhani S, Yildiz E. et al. Mechano-modulatory synthetic niches for liver organoid derivation. Nat Commun 2020; 11 (01) 3416-3416
  • 49 Op 't Veld RC, van den Boomen OI, Lundvig DMS. et al. Thermosensitive biomimetic polyisocyanopeptide hydrogels may facilitate wound repair. Biomaterials 2018; 181: 392-401
  • 50 Zimoch J, Padial JS, Klar AS. et al. Polyisocyanopeptide hydrogels: a novel thermo-responsive hydrogel supporting pre-vascularization and the development of organotypic structures. Acta Biomater 2018; 70: 129-139
  • 51 Eweida AM, Marei MK. Naturally occurring extracellular matrix scaffolds for dermal regeneration: do they really need cells?. BioMed Res Int 2015; 2015: 839694-839699
  • 52 Hillebrandt KH, Everwien H, Haep N, Keshi E, Pratschke J, Sauer IM. Strategies based on organ decellularization and recellularization. Transpl Int 2019; 32 (06) 571-585
  • 53 Zhang Q, Johnson JA, Dunne LW. et al. Decellularized skin/adipose tissue flap matrix for engineering vascularized composite soft tissue flaps. Acta Biomater 2016; 35: 166-184
  • 54 Mase Jr VJ, Hsu JR, Wolf SE. et al. Clinical application of an acellular biologic scaffold for surgical repair of a large, traumatic quadriceps femoris muscle defect. Orthopedics 2010; 33 (07) 511-511
  • 55 Jia L, Hua Y, Zeng J. et al. Bioprinting and regeneration of auricular cartilage using a bioactive bioink based on microporous photocrosslinkable acellular cartilage matrix. Bioact Mater 2022; 16: 66-81
  • 56 Paduano F, Marrelli M, Alom N. et al. Decellularized bone extracellular matrix and human dental pulp stem cells as a construct for bone regeneration. J Biomater Sci Polym Ed 2017; 28 (08) 730-748
  • 57 Li R, Xu J, Rao Z. et al. Facilitate angiogenesis and neurogenesis by growth factors integrated decellularized matrix hydrogel. Tissue Eng Part A 2021; 27 (11-12): 771-787
  • 58 Balestrini JL, Niklason LE. Extracellular matrix as a driver for lung regeneration. Ann Biomed Eng 2015; 43 (03) 568-576
  • 59 Kajbafzadeh A-M, Javan-Farazmand N, Monajemzadeh M, Baghayee A. Determining the optimal decellularization and sterilization protocol for preparing a tissue scaffold of a human-sized liver tissue. Tissue Eng Part C Methods 2013; 19 (08) 642-651
  • 60 Bonandrini B, Figliuzzi M, Papadimou E. et al. Recellularization of well-preserved acellular kidney scaffold using embryonic stem cells. Tissue Eng Part A 2014; 20 (9-10): 1486-1498
  • 61 Li M, Zhang C, Zhong Y, Zhao J. A novel approach to utilize icariin as icariin-derived ECM on small intestinal submucosa scaffold for bone repair. Ann Biomed Eng 2017; 45 (11) 2673-2682
  • 62 Iorio T, Blumberg D. Short-term results of treating primary and recurrent anal fistulas with a novel extracellular matrix derived from porcine urinary bladder. Am Surg 2015; 81 (05) 498-502
  • 63 Sun W, Yang Y, Wang L. et al. Utilization of an acellular cartilage matrix-based photocrosslinking hydrogel for tracheal cartilage regeneration and circumferential tracheal repair. Adv Funct Mater 2022; 32: 2201257
  • 64 Kim S, Min S, Choi YS. et al. Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 2022; 13 (01) 1692
  • 65 Willemse J, van Tienderen G, van Hengel E. et al. Hydrogels derived from decellularized liver tissue support the growth and differentiation of cholangiocyte organoids. Biomaterials 2022; 284: 121473-121473
  • 66 Simsa R, Rothenbücher T, Gürbüz H. et al. Brain organoid formation on decellularized porcine brain ECM hydrogels. PLoS One 2021; 16 (01) e0245685-e0245685
  • 67 Manichanh C, Borruel N, Casellas F, Guarner F. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol 2012; 9 (10) 599-608
  • 68 Krautkramer KA, Fan J, Bäckhed F. Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol 2021; 19 (02) 77-94
  • 69 Nicholson JK, Holmes E, Kinross J. et al. Host-gut microbiota metabolic interactions. Science 2012; 336 (6086): 1262-1267
  • 70 Matson V, Fessler J, Bao R. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018; 359 (6371): 104-108
  • 71 Routy B, Le Chatelier E, Derosa L. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. In: Science, United States. Vol. 359. American Association for the Advancement of Science; United States: 2018: 91-97
  • 72 Dejea CM, Fathi P, Craig JM. et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science 2018; 359 (6375): 592-597
  • 73 Wong SH, Zhao L, Zhang X. et al. Gavage of fecal samples from patients with colorectal cancer promotes intestinal carcinogenesis in germ-free and conventional mice. Gastroenterology 2017; 153 (06) 1621-1633.e6
  • 74 Viaud S, Saccheri F, Mignot G. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013; 342 (6161): 971-976
  • 75 Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer 2017; 17 (05) 271-285
  • 76 Frankel AE, Coughlin LA, Kim J. et al. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia 2017; 19 (10) 848-855
  • 77 Rubert J, Schweiger PJ, Mattivi F, Tuohy K, Jensen KB, Lunardi A. Intestinal organoids: a tool for modelling diet-microbiome-host interactions. Trends Endocrinol Metab 2020; 31 (11) 848-858
  • 78 Duchartre Y, Kim YM, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol 2016; 99: 141-149
  • 79 Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in human disease. Cell 2017; 170 (04) 605-635
  • 80 Li X, Huang J, Yu T. et al. Fusobacterium nucleatum promotes the progression of colorectal cancer through Cdk5-activated Wnt/β-catenin signaling. Front Microbiol 2021; 11: 545251
  • 81 Long X, Wong CC, Tong L. et al. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat Microbiol 2019; 4 (12) 2319-2330
  • 82 Ternes D, Tsenkova M, Pozdeev VI. et al. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat Metab 2022; 4 (04) 458-475
  • 83 Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol 2017; 17 (04) 219-232
  • 84 Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A 2010; 107 (27) 12204-12209
  • 85 Jia W, Xie G, Jia W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol 2018; 15 (02) 111-128
  • 86 Wang S, Dong W, Liu L. et al. Interplay between bile acids and the gut microbiota promotes intestinal carcinogenesis. Mol Carcinog 2019; 58 (07) 1155-1167
  • 87 Ma C, Han M, Heinrich B. et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018; 360 (6391): eaan5931
  • 88 Hanfrey CC, Pearson BM, Hazeldine S. et al. Alternative spermidine biosynthetic route is critical for growth of Campylobacter jejuni and is the dominant polyamine pathway in human gut microbiota. J Biol Chem 2011; 286 (50) 43301-43312
  • 89 Di Martino ML, Campilongo R, Casalino M, Micheli G, Colonna B, Prosseda G. Polyamines: emerging players in bacteria-host interactions. Int J Med Microbiol 2013; 303 (08) 484-491
  • 90 Pegg AE. Toxicity of polyamines and their metabolic products. Chem Res Toxicol 2013; 26 (12) 1782-1800
  • 91 Holbert CE, Cullen MT, Casero Jr RA, Stewart TM. Polyamines in cancer: integrating organismal metabolism and antitumour immunity. Nat Rev Cancer 2022; 22 (08) 467-480
  • 92 Ghanavati R, Akbari A, Mohammadi F. et al. Lactobacillus species inhibitory effect on colorectal cancer progression through modulating the Wnt/β-catenin signaling pathway. Mol Cell Biochem 2020; 470 (1-2): 1-13
  • 93 Nadeem A, Aung KM, Ray T. et al. Suppression of β-catenin signaling in colon carcinoma cells by a bacterial protein. Int J Cancer 2021; 149 (02) 442-459
  • 94 Zununi Vahed S, Barzegari A, Rahbar Saadat Y, Goreyshi A, Omidi Y. Leuconostoc mesenteroides-derived anticancer pharmaceuticals hinder inflammation and cell survival in colon cancer cells by modulating NF-κB/AKT/PTEN/MAPK pathways. Biomed Pharmacother 2017; 94: 1094-1100
  • 95 Watschinger C, Moschen AR. Lactobacillus reuteri-an old acquaintance takes on a new task in colorectal tumor surveillance. Cancer Cell 2022; 40 (02) 125-127
  • 96 Cervantes-Barragan L, Chai JN, Tianero MD. et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 2017; 357 (6353): 806-810
  • 97 Dodd D, Spitzer MH, Van Treuren W. et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017; 551 (7682): 648-652
  • 98 Pardoll D. Cancer and the immune system: basic concepts and targets for intervention. Semin Oncol 2015; 42 (04) 523-538
  • 99 Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013; 39 (01) 1-10
  • 100 Wang Y, Ma R, Liu F, Lee SA, Zhang L. Modulation of gut microbiota: a novel paradigm of enhancing the efficacy of programmed death-1 and programmed death ligand-1 blockade therapy. Front Immunol 2018; 9 (MAR): 374-374
  • 101 Elkrief A, Derosa L, Zitvogel L, Kroemer G, Routy B. The intimate relationship between gut microbiota and cancer immunotherapy. Gut Microbes 2019; 10 (03) 424-428
  • 102 Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med 2019; 25 (03) 377-388
  • 103 Vétizou M, Pitt JM, Daillère R. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015; 350 (6264): 1079-1084
  • 104 Sivan A, Corrales L, Hubert N. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015; 350 (6264): 1084-1089
  • 105 Mager LF, Burkhard R, Pett N. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 2020; 369 (6510): 1481-1489
  • 106 Griffin ME, Espinosa J, Becker JL. et al. Enterococcus peptidoglycan remodeling promotes checkpoint inhibitor cancer immunotherapy. Science 2021; 373 (6558): 1040-1046
  • 107 Chen LC, Chen YY. Bacteria recycle tumour waste to fuel immune cells. Nature 2021; 598 (7882): 570-571
  • 108 Canale FP, Basso C, Antonini G. et al. Metabolic modulation of tumours with engineered bacteria for immunotherapy. Nature 2021; 598 (7882): 662-666
  • 109 Arrieta MC, Walter J, Finlay BB. Human microbiota-associated mice: a model with challenges. Cell Host Microbe 2016; 19 (05) 575-578
  • 110 Engevik MA, Ruan W, Esparza M. et al. Immunomodulation of dendritic cells by Lactobacillus reuteri surface components and metabolites. Physiol Rep 2021; 9 (02) e14719
  • 111 Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A. et al; Genomics England Research Consortium. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature 2020; 580 (7802): 269-273
  • 112 Fu A, Yao B, Dong T. et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell 2022; 185 (08) 1356-1372.e26
  • 113 Puschhof J, Pleguezuelos-Manzano C, Martinez-Silgado A. et al. Intestinal organoid cocultures with microbes. Nat Protoc 2021; 16 (10) 4633-4649
  • 114 Bartfeld S, Bayram T, van de Wetering M. et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology 2015; 148 (01) 126-136.e6
  • 115 Puschhof J, Pleguezuelos-Manzano C, Clevers H. Organoids and organs-on-chips: insights into human gut-microbe interactions. Cell Host Microbe 2021; 29 (06) 867-878
  • 116 Jalili-Firoozinezhad S, Gazzaniga FS, Calamari EL. et al. A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip. Nat Biomed Eng 2019; 3 (07) 520-531

Address for correspondence

Yan Zhang
Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University
No.1954 Huashan Road, Shanghai 200030
People's Republic of China   

Publication History

Article published online:
14 December 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol 2020; 21 (10) 571-584
  • 2 Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer 2018; 18 (07) 407-418
  • 3 Jin K, Teng L, Shen Y, He K, Xu Z, Li G. Patient-derived human tumour tissue xenografts in immunodeficient mice: a systematic review. Clin Transl Oncol 2010; 12 (07) 473-480
  • 4 McCauley HA, Wells JM. Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish. Development 2017; 144 (06) 958-962
  • 5 Nejman D, Livyatan I, Fuks G. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 2020; 368 (6494): 973-980
  • 6 Sato T, Vries RG, Snippert HJ. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009; 459 (7244): 262-265
  • 7 Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 2014; 345 (6194): 1247125
  • 8 Huch M, Dorrell C, Boj SF. et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 2013; 494 (7436): 247-250
  • 9 Barker N, Huch M, Kujala P. et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 2010; 6 (01) 25-36
  • 10 Lancaster MA, Renner M, Martin CA. et al. Cerebral organoids model human brain development and microcephaly. Nature 2013; 501 (7467): 373-379
  • 11 Xia Y, Nivet E, Sancho-Martinez I. et al. Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor-like cells. Nat Cell Biol 2013; 15 (12) 1507-1515
  • 12 Li M, Izpisua Belmonte JC. Organoids - preclinical models of human disease. N Engl J Med 2019; 380 (06) 569-579
  • 13 Kopper O, de Witte CJ, Lõhmussaar K. et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med 2019; 25 (05) 838-849
  • 14 Vlachogiannis G, Hedayat S, Vatsiou A. et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018; 359 (6378): 920-926
  • 15 Sachs N, de Ligt J, Kopper O. et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 2018; 172 (1-2): 373-386.e10
  • 16 Matano M, Date S, Shimokawa M. et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med 2015; 21 (03) 256-262
  • 17 Drost J, van Jaarsveld RH, Ponsioen B. et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 2015; 521 (7550): 43-47
  • 18 Miura A, Yamada D, Nakamura M. et al. Oncogenic potential of human pluripotent stem cell-derived lung organoids with HER2 overexpression. Int J Cancer 2021; 149 (08) 1593-1604
  • 19 Fumagalli A, Drost J, Suijkerbuijk SJE. et al. Genetic dissection of colorectal cancer progression by orthotopic transplantation of engineered cancer organoids. Proc Natl Acad Sci U S A 2017; 114 (12) E2357-E2364
  • 20 Yin X, Mead BE, Safaee H, Langer R, Karp JM, Levy O. Engineering stem cell organoids. Cell Stem Cell 2016; 18 (01) 25-38
  • 21 Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol 2005; 15 (05) 378-386
  • 22 Jabaji Z, Brinkley GJ, Khalil HA. et al. Type I collagen as an extracellular matrix for the in vitro growth of human small intestinal epithelium. PLoS One 2014; 9 (09) e107814
  • 23 Liu H, Bockhorn J, Dalton R. et al. Removal of lactate dehydrogenase-elevating virus from human-in-mouse breast tumor xenografts by cell-sorting. J Virol Methods 2011; 173 (02) 266-270
  • 24 Peterson NC. From bench to cageside: risk assessment for rodent pathogen contamination of cells and biologics. ILAR J 2008; 49 (03) 310-315
  • 25 Henke E, Nandigama R, Ergün S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci 2020; 6: 160
  • 26 Kaur S, Kaur I, Rawal P, Tripathi DM, Vasudevan A. Non-matrigel scaffolds for organoid cultures. Cancer Lett 2021; 504: 58-66
  • 27 Kratochvil MJ, Seymour AJ, Li TL, Paşca SP, Kuo CJ, Heilshorn SC. Engineered materials for organoid systems. Nat Rev Mater 2019; 4 (09) 606-622
  • 28 Magno V, Meinhardt A, Werner C. Polymer hydrogels to guide organotypic and organoid cultures. Adv Funct Mater 2020; 30 (48) 2000097
  • 29 Heidarian P, Kaynak A, Paulino M, Zolfagharian A, Varley RJ, Kouzani AZ. Dynamic nanocellulose hydrogels: recent advancements and future outlook. Carbohydr Polym 2021; 270: 118357-118357
  • 30 Zhu L, Zhang Y-Q. Postoperative anti-adhesion ability of a novel carboxymethyl chitosan from silkworm pupa in a rat cecal abrasion model. Mater Sci Eng C 2016; 61: 387-395
  • 31 Curvello R, Kerr G, Micati DJ. et al. Engineered plant-based nanocellulose hydrogel for small intestinal organoid growth. Adv Sci (Weinh) 2020; 8 (01) 2002135
  • 32 Prince E, Cruickshank J, Ba-Alawi W. et al. Biomimetic hydrogel supports initiation and growth of patient-derived breast tumor organoids. Nat Commun 2022; 13 (01) 1466
  • 33 Meran L, Baulies A, Li VSW. Intestinal stem cell niche: the extracellular matrix and cellular components. Stem Cells Int 2017; 2017: 7970385-11
  • 34 Wang H, Cui J, Zheng Z. et al; Assembly of RGD-Modified Hydrogel Micromodules into Permeable Three-Dimensional Hollow Microtissues Mimicking in Vivo Tissue Structures. Assembly of RGD-modified hydrogel micromodules into permeable three-dimensional hollow microtissues mimicking in vivo tissue structures. ACS Appl Mater Interfaces 2017; 9 (48) 41669-41679
  • 35 Curvello R, Alves D, Abud HE, Garnier G. A thermo-responsive collagen-nanocellulose hydrogel for the growth of intestinal organoids. Mater Sci Eng C 2021; 124: 112051-112051
  • 36 Guimarães CF, Gasperini L, Marques AP, Reis RL. The stiffness of living tissues and its implications for tissue engineering. Nat Rev Mater 2020; 5 (05) 351-370
  • 37 Curvello R, Garnier G. Cationic cross-linked nanocellulose-based matrices for the growth and recovery of intestinal organoids. Biomacromolecules 2021; 22 (02) 701-709
  • 38 Lee KY, Mooney DJ. Alginate: properties and biomedical applications. Prog Polym Sci 2012; 37 (01) 106-126
  • 39 Capeling MM, Czerwinski M, Huang S. et al. Nonadhesive alginate hydrogels support growth of pluripotent stem cell-derived intestinal organoids. Stem Cell Reports 2019; 12 (02) 381-394
  • 40 Dong H, Li Z, Bian S. et al. Culture of patient-derived multicellular clusters in suspended hydrogel capsules for pre-clinical personalized drug screening. Bioact Mater 2022; 18: 164-177
  • 41 Capeling MM, Huang S, Childs CJ. et al. Suspension culture promotes serosal mesothelial development in human intestinal organoids. Cell Rep 2022; 38 (07) 110379-110379
  • 42 Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell 2006; 126 (04) 677-689
  • 43 Cassel de Camps C, Aslani S, Stylianesis N. et al. Hydrogel mechanics influence the growth and development of embedded brain organoids. ACS Appl Bio Mater 2022; 5 (01) 214-224
  • 44 Fang G, Lu H, Rodriguez de la Fuente L. et al. Mammary tumor organoid culture in non-adhesive alginate for luminal mechanics and high-throughput drug screening. Adv Sci (Weinh) 2021; 8 (21) e2102418
  • 45 Deshpande SR, Hammink R, Nelissen FHT, Rowan AE, Heus HA. Biomimetic stress sensitive hydrogel controlled by DNA nanoswitches. Biomacromolecules 2017; 18 (10) 3310-3317
  • 46 Zhang Y, Tang C, Span PN. et al. Polyisocyanide hydrogels as a tunable platform for mammary gland organoid formation. Adv Sci (Weinh) 2020; 7 (18) 2001797
  • 47 Ye S, Boeter JWB, Mihajlovic M. et al. A chemically defined hydrogel for human liver organoid culture. Adv Funct Mater 2020; 30 (48) 2000893
  • 48 Sorrentino G, Rezakhani S, Yildiz E. et al. Mechano-modulatory synthetic niches for liver organoid derivation. Nat Commun 2020; 11 (01) 3416-3416
  • 49 Op 't Veld RC, van den Boomen OI, Lundvig DMS. et al. Thermosensitive biomimetic polyisocyanopeptide hydrogels may facilitate wound repair. Biomaterials 2018; 181: 392-401
  • 50 Zimoch J, Padial JS, Klar AS. et al. Polyisocyanopeptide hydrogels: a novel thermo-responsive hydrogel supporting pre-vascularization and the development of organotypic structures. Acta Biomater 2018; 70: 129-139
  • 51 Eweida AM, Marei MK. Naturally occurring extracellular matrix scaffolds for dermal regeneration: do they really need cells?. BioMed Res Int 2015; 2015: 839694-839699
  • 52 Hillebrandt KH, Everwien H, Haep N, Keshi E, Pratschke J, Sauer IM. Strategies based on organ decellularization and recellularization. Transpl Int 2019; 32 (06) 571-585
  • 53 Zhang Q, Johnson JA, Dunne LW. et al. Decellularized skin/adipose tissue flap matrix for engineering vascularized composite soft tissue flaps. Acta Biomater 2016; 35: 166-184
  • 54 Mase Jr VJ, Hsu JR, Wolf SE. et al. Clinical application of an acellular biologic scaffold for surgical repair of a large, traumatic quadriceps femoris muscle defect. Orthopedics 2010; 33 (07) 511-511
  • 55 Jia L, Hua Y, Zeng J. et al. Bioprinting and regeneration of auricular cartilage using a bioactive bioink based on microporous photocrosslinkable acellular cartilage matrix. Bioact Mater 2022; 16: 66-81
  • 56 Paduano F, Marrelli M, Alom N. et al. Decellularized bone extracellular matrix and human dental pulp stem cells as a construct for bone regeneration. J Biomater Sci Polym Ed 2017; 28 (08) 730-748
  • 57 Li R, Xu J, Rao Z. et al. Facilitate angiogenesis and neurogenesis by growth factors integrated decellularized matrix hydrogel. Tissue Eng Part A 2021; 27 (11-12): 771-787
  • 58 Balestrini JL, Niklason LE. Extracellular matrix as a driver for lung regeneration. Ann Biomed Eng 2015; 43 (03) 568-576
  • 59 Kajbafzadeh A-M, Javan-Farazmand N, Monajemzadeh M, Baghayee A. Determining the optimal decellularization and sterilization protocol for preparing a tissue scaffold of a human-sized liver tissue. Tissue Eng Part C Methods 2013; 19 (08) 642-651
  • 60 Bonandrini B, Figliuzzi M, Papadimou E. et al. Recellularization of well-preserved acellular kidney scaffold using embryonic stem cells. Tissue Eng Part A 2014; 20 (9-10): 1486-1498
  • 61 Li M, Zhang C, Zhong Y, Zhao J. A novel approach to utilize icariin as icariin-derived ECM on small intestinal submucosa scaffold for bone repair. Ann Biomed Eng 2017; 45 (11) 2673-2682
  • 62 Iorio T, Blumberg D. Short-term results of treating primary and recurrent anal fistulas with a novel extracellular matrix derived from porcine urinary bladder. Am Surg 2015; 81 (05) 498-502
  • 63 Sun W, Yang Y, Wang L. et al. Utilization of an acellular cartilage matrix-based photocrosslinking hydrogel for tracheal cartilage regeneration and circumferential tracheal repair. Adv Funct Mater 2022; 32: 2201257
  • 64 Kim S, Min S, Choi YS. et al. Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 2022; 13 (01) 1692
  • 65 Willemse J, van Tienderen G, van Hengel E. et al. Hydrogels derived from decellularized liver tissue support the growth and differentiation of cholangiocyte organoids. Biomaterials 2022; 284: 121473-121473
  • 66 Simsa R, Rothenbücher T, Gürbüz H. et al. Brain organoid formation on decellularized porcine brain ECM hydrogels. PLoS One 2021; 16 (01) e0245685-e0245685
  • 67 Manichanh C, Borruel N, Casellas F, Guarner F. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol 2012; 9 (10) 599-608
  • 68 Krautkramer KA, Fan J, Bäckhed F. Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol 2021; 19 (02) 77-94
  • 69 Nicholson JK, Holmes E, Kinross J. et al. Host-gut microbiota metabolic interactions. Science 2012; 336 (6086): 1262-1267
  • 70 Matson V, Fessler J, Bao R. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018; 359 (6371): 104-108
  • 71 Routy B, Le Chatelier E, Derosa L. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. In: Science, United States. Vol. 359. American Association for the Advancement of Science; United States: 2018: 91-97
  • 72 Dejea CM, Fathi P, Craig JM. et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science 2018; 359 (6375): 592-597
  • 73 Wong SH, Zhao L, Zhang X. et al. Gavage of fecal samples from patients with colorectal cancer promotes intestinal carcinogenesis in germ-free and conventional mice. Gastroenterology 2017; 153 (06) 1621-1633.e6
  • 74 Viaud S, Saccheri F, Mignot G. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013; 342 (6161): 971-976
  • 75 Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer 2017; 17 (05) 271-285
  • 76 Frankel AE, Coughlin LA, Kim J. et al. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia 2017; 19 (10) 848-855
  • 77 Rubert J, Schweiger PJ, Mattivi F, Tuohy K, Jensen KB, Lunardi A. Intestinal organoids: a tool for modelling diet-microbiome-host interactions. Trends Endocrinol Metab 2020; 31 (11) 848-858
  • 78 Duchartre Y, Kim YM, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol 2016; 99: 141-149
  • 79 Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in human disease. Cell 2017; 170 (04) 605-635
  • 80 Li X, Huang J, Yu T. et al. Fusobacterium nucleatum promotes the progression of colorectal cancer through Cdk5-activated Wnt/β-catenin signaling. Front Microbiol 2021; 11: 545251
  • 81 Long X, Wong CC, Tong L. et al. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat Microbiol 2019; 4 (12) 2319-2330
  • 82 Ternes D, Tsenkova M, Pozdeev VI. et al. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat Metab 2022; 4 (04) 458-475
  • 83 Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol 2017; 17 (04) 219-232
  • 84 Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A 2010; 107 (27) 12204-12209
  • 85 Jia W, Xie G, Jia W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol 2018; 15 (02) 111-128
  • 86 Wang S, Dong W, Liu L. et al. Interplay between bile acids and the gut microbiota promotes intestinal carcinogenesis. Mol Carcinog 2019; 58 (07) 1155-1167
  • 87 Ma C, Han M, Heinrich B. et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018; 360 (6391): eaan5931
  • 88 Hanfrey CC, Pearson BM, Hazeldine S. et al. Alternative spermidine biosynthetic route is critical for growth of Campylobacter jejuni and is the dominant polyamine pathway in human gut microbiota. J Biol Chem 2011; 286 (50) 43301-43312
  • 89 Di Martino ML, Campilongo R, Casalino M, Micheli G, Colonna B, Prosseda G. Polyamines: emerging players in bacteria-host interactions. Int J Med Microbiol 2013; 303 (08) 484-491
  • 90 Pegg AE. Toxicity of polyamines and their metabolic products. Chem Res Toxicol 2013; 26 (12) 1782-1800
  • 91 Holbert CE, Cullen MT, Casero Jr RA, Stewart TM. Polyamines in cancer: integrating organismal metabolism and antitumour immunity. Nat Rev Cancer 2022; 22 (08) 467-480
  • 92 Ghanavati R, Akbari A, Mohammadi F. et al. Lactobacillus species inhibitory effect on colorectal cancer progression through modulating the Wnt/β-catenin signaling pathway. Mol Cell Biochem 2020; 470 (1-2): 1-13
  • 93 Nadeem A, Aung KM, Ray T. et al. Suppression of β-catenin signaling in colon carcinoma cells by a bacterial protein. Int J Cancer 2021; 149 (02) 442-459
  • 94 Zununi Vahed S, Barzegari A, Rahbar Saadat Y, Goreyshi A, Omidi Y. Leuconostoc mesenteroides-derived anticancer pharmaceuticals hinder inflammation and cell survival in colon cancer cells by modulating NF-κB/AKT/PTEN/MAPK pathways. Biomed Pharmacother 2017; 94: 1094-1100
  • 95 Watschinger C, Moschen AR. Lactobacillus reuteri-an old acquaintance takes on a new task in colorectal tumor surveillance. Cancer Cell 2022; 40 (02) 125-127
  • 96 Cervantes-Barragan L, Chai JN, Tianero MD. et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 2017; 357 (6353): 806-810
  • 97 Dodd D, Spitzer MH, Van Treuren W. et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017; 551 (7682): 648-652
  • 98 Pardoll D. Cancer and the immune system: basic concepts and targets for intervention. Semin Oncol 2015; 42 (04) 523-538
  • 99 Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013; 39 (01) 1-10
  • 100 Wang Y, Ma R, Liu F, Lee SA, Zhang L. Modulation of gut microbiota: a novel paradigm of enhancing the efficacy of programmed death-1 and programmed death ligand-1 blockade therapy. Front Immunol 2018; 9 (MAR): 374-374
  • 101 Elkrief A, Derosa L, Zitvogel L, Kroemer G, Routy B. The intimate relationship between gut microbiota and cancer immunotherapy. Gut Microbes 2019; 10 (03) 424-428
  • 102 Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med 2019; 25 (03) 377-388
  • 103 Vétizou M, Pitt JM, Daillère R. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015; 350 (6264): 1079-1084
  • 104 Sivan A, Corrales L, Hubert N. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015; 350 (6264): 1084-1089
  • 105 Mager LF, Burkhard R, Pett N. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 2020; 369 (6510): 1481-1489
  • 106 Griffin ME, Espinosa J, Becker JL. et al. Enterococcus peptidoglycan remodeling promotes checkpoint inhibitor cancer immunotherapy. Science 2021; 373 (6558): 1040-1046
  • 107 Chen LC, Chen YY. Bacteria recycle tumour waste to fuel immune cells. Nature 2021; 598 (7882): 570-571
  • 108 Canale FP, Basso C, Antonini G. et al. Metabolic modulation of tumours with engineered bacteria for immunotherapy. Nature 2021; 598 (7882): 662-666
  • 109 Arrieta MC, Walter J, Finlay BB. Human microbiota-associated mice: a model with challenges. Cell Host Microbe 2016; 19 (05) 575-578
  • 110 Engevik MA, Ruan W, Esparza M. et al. Immunomodulation of dendritic cells by Lactobacillus reuteri surface components and metabolites. Physiol Rep 2021; 9 (02) e14719
  • 111 Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A. et al; Genomics England Research Consortium. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature 2020; 580 (7802): 269-273
  • 112 Fu A, Yao B, Dong T. et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell 2022; 185 (08) 1356-1372.e26
  • 113 Puschhof J, Pleguezuelos-Manzano C, Martinez-Silgado A. et al. Intestinal organoid cocultures with microbes. Nat Protoc 2021; 16 (10) 4633-4649
  • 114 Bartfeld S, Bayram T, van de Wetering M. et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology 2015; 148 (01) 126-136.e6
  • 115 Puschhof J, Pleguezuelos-Manzano C, Clevers H. Organoids and organs-on-chips: insights into human gut-microbe interactions. Cell Host Microbe 2021; 29 (06) 867-878
  • 116 Jalili-Firoozinezhad S, Gazzaniga FS, Calamari EL. et al. A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip. Nat Biomed Eng 2019; 3 (07) 520-531