Skip to main content

Advances in porcine respiratory and intestinal organoids: status and potential application for virus infections

Abstract

The respiratory tract and digestive tract serve as the gateway between the host and the environment, playing an important role in protecting against viral infections. Diseases caused by viruses that infiltrate the respiratory and gastrointestinal tracts account for the major infectious diseases in pigs, resulting in significant economic losses for the swine industry. However, studies on virus-host interactions are limited due to the lack of suitable research models that can effectively stimulate the highly complex physiological characteristics found in vivo. With the advancement in stem cell technology, organoids that more closely recapitulate the structure, function, and organization of specific organs or tissues in vitro have gradually become a research hotspot. These novel ex vivo models are critical for studying viral infection, investigating viral pathogenesis, elucidating virus-host interactions and developing preventive and therapeutic approaches. Currently, respiratory organoids and intestinal organoids (IOs) have been widely applied in the study of infectious diseases. Therefore, this review primarily summarizes the development of porcine respiratory and intestinal organoids, their applications in studying infection, current limitations, and future perspectives.

Introduction

Respiratory and digestive diseases are common diseases on swine farms. African swine fever, porcine epidemic diarrhea (PED), porcine circovirus disease, porcine enzootic pneumonia and porcine colibacillosis are common diseases on large-scale pig farms [1]. Among them, viruses are one of the primary culprits responsible for the largest number of diseases and the most tremendous economic losses [2]. For example, porcine epidemic diarrhea virus (PEDV) broke out in the United States in 2013, and the virus had impacted approximately 50% of U.S. breeding herds within one year [3]. In 2018, African swine fever virus spread rapidly across China within a year and caused catastrophic damage to the pork industry of China [4]. The respiratory and digestive tracts of the host are dynamic, cellularly diverse and histologically intricate systems under tight regulation. In addition to maintaining normal physiological functions, they must form a protective barrier to resist an entry of the tremendous array of pathogens into the host organism [5,6,7]. The nasal mucosa, lysozyme, interferon (IFN), SIgA antibodies secreted by the trachea and bronchi, and the alveolar macrophages play important roles in protecting the body against respiratory viruses [8]. The intestinal barrier, which consists of the mechanical barrier, chemical barrier, microbial barrier and immune barrier, plays a key role in maintaining intestinal health and preventing the invasion of enteroviruses [9]. Therefore, the respiratory and digestive tracts play an important role in viral infections. Having a suitable model is crucial for elucidating the infection and pathogenic mechanisms of viruses, and animal models and immortalized (cancerous) cell lines are the primary models for studying host–pathogen interactions [10, 11]. Although these model systems have greatly expanded our knowledge of virology, their limitations are also evident. Universally used transformed cell lines are genetically unstable and do not recapitulate the complex composition and microenvironment of untransformed cells, nor the complex interactions between the viruses and the host's immune response [12, 13]. Animal models provide a functional read-out and a more comprehensive model of infection, but they are limited by individual variations, biological differences between species, animal welfare and ethical concerns, and are also hampered by high cost, low throughput, and poor convenience [14]. Successful cultivation of organoids bridges the gap between transformed cell lines and animal models by addressing in vivo complexity to reconcile moderate systemic complexity and reproducibility [15, 16]. Compared to three-dimensional (3D) cultures of tissue explants, organoid systems have better genetic stability and better mimic cell–cell/cell–matrix interactions [13, 17, 18]. Over the past decade, virologists have increasingly turned to organoids as tools to further elucidate virus-host interactions [19,20,21,22].

Overview of airway organoids (AOs) and intestinal organoids (IOs)

Organoids, also known as "mini-organs," are 3D structures grown from stem cells. These structures are composed of organ-specific cell types to recapitulate the cellular structure and function of native organs [15, 23]. In recent years, organoids have been widely used in many biological fields, not only for studying the interaction between hosts and pathogenic microorganisms (bacteria, viruses, and parasites) [22, 24,25,26,27], but also for immunological research, to understand epithelial cell-immune cell interactions [28,29,30]. In addition, organoids play an important role in cancer research, tissue regeneration, and clinical drug screening [31, 32]. The starting point for creating an organoid can vary considerably. In the models currently used, the tissue structure is mainly obtained from pluripotent stem cells (PSCs) and adult stem cells (ASCs). Among them, PSC-derived organoids include embryonic stem cells (ESCs)-derived organoids and induced pluripotent stem cells (iPSCs)-derived organoids [12]. In addition to producing epithelial cell types, PSCs can also differentiate into other functional cells, such as fibroblasts and muscle cells. ASCs-derived organoids are generated by directly dissociating intrinsic ASC-containing tissues and exposing them to tissue-specific growth factors [33], followed by tissue-specific enrichment by modulating individual signaling pathways such as bone morphogenic protein (BMP) or Notch [34]. In summary, the growth factors used in the culture of PSCs and ASCs-derived organoids are different because ASCs have undergone a certain degree of differentiation compared with PSCs. PSCs or ASCs grow in various extracellular matrices (such as Matrigel) and self-assemble into 3D structures. Growth factors in the medium can induce further development of organoids from stem cells to differentiated cells that mimic functional epithelium [35]. With the continuous advancement of organoid technology, porcine organoid culture technology has developed rapidly. Porcine organoids derived from ASCs, such as AOs and IOs, have been successfully established in vitro (Fig. 1). They currently play an important role in disease research. In addition, since the size and the composition of the porcine genome and the functional features of porcine organs are similar to humans, porcine organoids are often used to simulate the physiological and pathological functions of humans [36]. As a result, porcine organoids have been widely used in agriculture, veterinary medicine, and biomedicine.

Fig. 1
figure 1

Schematic diagram of porcine adult stem cells derived respiratory and intestinal organoid culture. Currently, porcine AOs are grown using basal epithelial cells isolated from the trachea. AOs are composed of the four main types of airway epithelial cells, including ciliated cells, goblet cells, basal cells, and club cells. IOs mainly include five main cell types: goblet cells, intestinal epithelial cells, enteroendocrine cells, Paneth cells and intestinal stem cells

Airway organoids (AOs)

The airway (except the nasal vestibule) to the terminal bronchioles contains five major types of epithelial cells: ciliated cells, goblet cells, small granule cells, brush cells and basal cells [37, 38]. Alveolar sac, as the basic unit of oxygen exchange, is lined with alveolar epithelium composed of flat type I alveolar epithelial (AT1) cells and cuboidal type II alveolar epithelial (AT2) cells [39]. Currently, AOs (respiratory tract and lung) have been widely used to monitor viral infection, explore pathological changes, and identify potential treatments [40,41,42].

AOs can be obtained from a variety of stem cells, including iPSCs, ESCs, and adult or fetal stem cells from surgical specimens, each of which can differentiate into all cell types [43]. Depending on the type of stem cells, AOs are divided into PSC-derived and ASC-derived AOs. Currently, PSC-derived AOs play an important role in the study of lung developmental biology due to their advantages, such as not being limited by tissue sample scarcity, having stable amplification capacity, being able to differentiate into most cell types, and being accessible for gene editing [44,45,46]. The culture of PSC-derived AOs involves four essential stages: definitive endoderm, anterior foregut endoderm, lung progenitor cells, and all types of AOs [47,48,49]. However, only AOs derived from human or mouse PSCs have been reported thus far, and AOs from porcine PSCs have not yet been reported.

Although PSC-derived AOs can be differentiated to represent airway and alveolar regions, their differentiation process is complex and tedious. Thus, using PSC-derived AOs as a model remains limited. In contrast, ASC-derived AOs are widely used due to their ability to exhibit complex structures with mature components [50,51,52]. Unlike intestinal stem cells (ISCs), multiple cell types in the lung are capable of pluripotent differentiation when the lung is injured. In numerous studies researchers have attempted to develop AOs from different progenitor cells from mice and humans, including basal cells, rod cells, secretory cells and AT2 cells [53, 54]. For example, human lung organoids have been developed from normal lung tissue and directed to an immature state in a feeder-free culture system. Organoids in an immature state can faithfully generate epithelial domains that phenocopy the native airway epithelium by adopting a proximal differentiation protocol [55]. Moreover, alveolar organoids composed of AT1 and AT2 cells can be generated by adopting a distal differentiation protocol [56, 57]. Zhou et al. [55] established long-term expanded 3D human AOs with normal lung tissue, and developed an improved two-dimensional (2D) monolayer culture system for the differentiated AOs. Furthermore, due to the basal-out polarity of first-generation organoids, pathogens can rarely attach directly to the apical membrane, making it difficult to initiate infection. Boecking et al. [58] established AOs with externally oriented apical membranes, which is conducive to pathogenic infection of AOs. Before that, most studies of porcine respiratory infections were conducted using immortalized cell lines, primary airway epithelial cells, or porcine lung explants [59,60,61]. With the development of organoid technology in vitro, our laboratories achieved long-term porcine AOs (Fig. 2), which contain four main airway epithelial cell types: ciliated cells, goblet cells, basal cells and club cells [62]. The results from further experiments indicated that both 3D and 2D AOs can be successfully infected with transmissible gastroenteritis coronavirus (TGEV) and porcine respiratory coronavirus (PRCoV), and can produce significant interferon (IFN) and inflammatory responses, demonstrating that porcine AOs have become a potential universal platform for porcine respiratory infections [62].

Fig. 2
figure 2

Porcine ASC-derived airway organoids. Representative images of AO differentiation derived from the trachea of 2-day-old piglets. The tracheal epithelial cells were cultured in a Matrigel matrix, and their daily growth was observed under a microscope [62]. Scale bar = 200 mm

Intestinal organoids (IOs)

The intestinal epithelium, which consists of the villus and the crypt, is the fastest self-renewing tissue in mammals, and the ISCs located in the bottom crypt region are an indispensable driving force for its rapid renewal [63]. ISCs undergo asymmetric cell division into new stem cells and committed daughter cells, termed transit-amplifying (TA) cells. TA cells subsequently differentiate into functional cell types, including absorptive enterocytes and secretory cells (Paneth cells, goblet cells, and enteroendocrine cells) [64]. The self-proliferation and differentiation of ISCs are regulated via various signaling pathways. For example, the Wnt signaling pathway plays an important role in promoting cell proliferation and self-renewal [65], the Notch signaling pathway promotes cell differentiation, and the BMP signaling pathway inhibits the activity of the β-catenin protein [66, 67]. In recent years, with the progress of intestinal stem cell isolation technology, it has become possible to achieve long-term culture of intestinal epithelial cells in vitro. IOs can be generated by differentiation from PSCs or by derivation from isolated multipotent stem cells and progenitor cells present in vivo intestinal crypts. Among them, organoids initially created from the small intestinal crypts containing lgr5+ ISCs are called enteroids, those created from the colon crypts containing lgr5+ ISCs are called colonoids [68]. In 2009, IOs were first developed by Sato et al. [69] by culturing mouse intestinal crypts or single ISCs in an extracellular matrix. In 2011, the same research group established crypt-derived human IO cultures by adding nicotinamide and various small molecule inhibitors that are used to promote the growth of mouse organoids [70]. In the same year, Spence et al. [71] established PSC-derived human IOs. Subsequently, other researchers reported the successful culturing and establishment of IOs in cattle, pigs, dogs, cats, chickens, and bats [25, 72,73,74,75,76]. The choice of IO model depends on the purpose of the study [77]. For instance, PSC-derived IOs may be better when studying the intestinal development and differentiation in vivo, while ASC-derived IOs may be preferable for disease research because the characteristics of the tissue of origin are preserved in the organoids. Whichever the IO models, all intestinal epithelial cell types, are polarized toward the lumen, and can be cultured in an in vitro environment for prolonged periods of time. Although recent progress has been made in developing livestock iPSCs and ESC [78], all porcine IO models are developed primarily with tissue-derived intestinal epithelial stem cells (IESCs) (Fig. 1). The first step of porcine IO culture is to obtain the porcine intestinal segments, and then dissociation buffer is used to isolate epithelial crypts that contain IESCs. Next, the isolated crypts were seeded in Matrigel and the growth medium was added [79]. The three proteins Wnt3a, R-spondin1 and Noggin in the growth medium play a key role in culturing porcine IOs [79]. In addition, other supplementary factors in the culture medium, including N2 supplementation, B27 supplementation, nicotinamide, and N-acetylcysteine, ensure the long-term culture of porcine IOs [80]. Matrigel, which is widely used in organoid culture and is prepared from the secretion of EngelbrethHolm-Swarm mouse sarcoma cells [81], has several key limitations, including complex and poorly defined composition, batch-to-batch variability, high cost and safety issues [82]. Therefore, more and more studies are trying to find new organoid culture substrates. Currently, synthetic hydrogel matrices, decellularized extracellular matrices, and natural hydrogels such as type I collagen all have the potential to replace Matrigel [83]. Gonzalez et al. [73] successfully cultured porcine IOs from piglet jejunum for the first time. Since then, porcine IOs from different intestinal segments, such as the duodenum, jejunum, ileum, and colon, have been rapidly established and used as in vitro models in various research fields [22, 84,85,86] (Fig. 3). Kar et al. [87] found that the transcriptome profiles of different intestinal segments and their derived organoids showed high resemblance, which demonstrates the high complexity of porcine IO and the resemblance to in vivo tissue. Duchesne et al. [88] evaluated the effect of ISC donor age on the growth, morphology, and cellular composition of porcine IOs. This study revealed that IOs derived from young piglets bud and grow faster than those derived from older pigs, but there was no difference in the cellular composition of the organoids. In addition, Li et al. [22] reported that compared with enteroids, the proliferation and differentiation rate of colonoids is significantly slower.

Fig. 3
figure 3

Porcine ASC-derived intestinal organoids. A Representative images of the time course of porcine enteroid differentiation from intestinal crypts. During culture in Matrigel, small spheroids form on day 3 after crypt isolation, gradually mature over time, and form budding-like crypt structures on day 7 [22]. B Representative images of the time course of porcine colonoid development. Colonic crypts were isolated and differentiated into budding-like colonoid structures on day 15 [22]

The application of airway organoids (AOs) and intestinal organoids (IOs) in virology research

In the past decade, respiratory viruses, and enteroviruses such as PEDV, TGEV, pseudorabies virus and porcine reproductive and respiratory syndrome virus (PRRSV) have caused serious economic losses in the swine industry [89,90,91]. Different viruses follow different infection mechanisms in their hosts, leading to different disease symptoms, so it is important to use the model system that allows for the most accurate recreation of viral infection mechanisms. Cell lines commonly used in previous studies, such as IPEC-J2 and IPI-2I [92], are not susceptible to infection by certain viruses, and there are huge variations in the findings reported from different research groups using the same cell lines [93]. Therefore, there is an urgent need to develop a more physiological culture system for porcine virology research. Currently, porcine organoids, which can simulate better in vivo environments, are widely used in the field.

Airway organoids (AOs) in virology research

AOs provide a reliable platform for studying virus-host interactions in the nasal cavity, proximal lung, and distal lung in vitro. AOs derived from different parts of the respiratory tract provide novel opportunities to study cell tropism of viruses and the immune response of the host. Furthermore, researchers can focus on different cell types or regions in the respiratory tract to target therapeutics of various viruses [94].

AOs have been used extensively for treating respiratory pathogens in humans and rodent species. It has been reported that AOs derived from ASCs can be used to study influenza viruses and respiratory syncytial viruses [55, 95], and airway and lung organoids derived from ASCs and human PSCs have been used in research on severe acute respiratory syndrome coronavirus 2 [20, 96]. Although many viruses, such as PRCoV, swine influenza virus, and PRRSV, can infect respiratory epithelial cells (Table 1), there are few reports on porcine AOs. In 2022, our laboratories successfully generated long-term porcine AOs derived from basal epithelial cells and applied this model to assess the permissiveness of AOs for PRCoV and TGEV infection for the first time [62]. We infected 3D AOs differentiated from the tracheal epithelium with PRCoV and TGEV and observed that both PRCoV and TGEV could successfully infect pig 3D AOs and mainly infected the secretory cells and ciliated cells. Moreover, the cell-intrinsic response of AOs after viral infection helps to elucidate the pathogenic mechanism of coronavirus, and single-cell RNA sequencing results indicate that PRCoV induces a strong immune response in infected AOs. In most 3D organoid models, the apical side of the cells is facing the inside of the organoid, whereas the apical side of the 2D AO cells is exposed to the air, making it more susceptible to viral infection and therefore more suitable for virologic studies [50]. Likewise, the study found that 2D single-layer AOs were more susceptible to infection than 3D AOs [62]. In addition to 2D AO models, apical-out AO models and organoids seeded on Transwell plates to form organoids and air–liquid interface (ALI) models are helpful for viral infection. The ALI culture system can expose apical cells to the outside environment, so organoids are more susceptible to infection by pathogenic microorganisms than basal-out organoids. However, ALI culture systems rely heavily on the use of tissue culture plate inserts, which are composed of permeable membranes and have limited scalability. As a comparison, apical-out organoids both overcome this limitation and allow the apical cells to be exposed to the outside. However, these culture systems have not yet been applied to porcine AOs [97, 98]. In summary, the porcine AO model can simulate the infection process of the virus in vivo, which lays the foundation for further in-depth exploration of the pathogenic mechanism of respiratory coronavirus.

Table 1 The respiratory epithelial cells in virus infection

Intestinal organoids (IOs) in virology research

Diarrhea caused by various intestinal pathogenic microorganisms is a common clinical disease, which seriously endangers human or animal health [106]. In the development of diarrheal diseases, intestinal epithelium is the main target of pathogenic infection [107]. Since traditional in vitro cell models cannot recapitulate the highly complex physiological characteristics of the gastrointestinal tract, IO models that can better simulate the in vivo environment have become important research models. To date, the IOs have been applied for modeling host-bacterial dynamics and interactions between the intestinal epithelium and organisms such as E. coli, Clostridium difficile, and Salmonella typhi. This model has also been utilized to reveal novel and interesting aspects of host-virus interactions as well as features of replication and pathogenesis for enteric viruses [108].

A variety of viruses exist in the porcine intestine, but current studies using IOs primarily focus on porcine enteric coronaviruses, such as PEDV, TGEV, and porcine deltacoronavirus (PDCoV) [109,110,111] (Table 2). These viruses are the main causes of watery diarrhea in newborn pigs and pose a huge threat to the swine industry and public health [112]. In 2019, our laboratories first studied the susceptibility of porcine intestine to PEDV by establishing porcine IOs from porcine duodenum, jejunum, ileum, and colon. The results showed that PEDV infects multiple types of cells in IOs, including enterocytes, goblet cells and stem cells. Additionally, there are potential differences in the susceptibility of organoids derived from different intestinal segments to PEDV infection; that is, PEDV preferentially infects ileal organoids compared with colon organoids. These findings are consistent with the in vivo results [22]. Furthermore, we found that PDCoV preferably infected the jejunum and ileum, and restricted replication in the duodenum and colon in the established IOs from different intestinal segments in 2020 [21]. Our study provides further evidence that the differences in PDCoV tropism for different intestinal segments are mainly determined by host aminopeptidase N rather than IFN [21]. Additionally, Luo et al. [113] established porcine small IOs to detect the replication of PDCoV in vitro. Double immunofluorescence labeling showed that PDCoV was present in Sox9-positive intestinal cells and Villin1-positive enterocytes. In 2022, our laboratories used IOs to evaluate host epithelial cell responses to infection by three porcine enteric coronaviruses (PEDV, TGEV, and PDCoV), and the results from these studies showed that they act via a contrasting, similar, and unique mechanism to modulate global IFN responses and the expression of antigen-presentation-associated genes [112]. Besides coronaviruses, Lee et al. [114] reported that mammalian orthoreovirus type 3 (MRV3) can infect 2D and 3D porcine jejunal organoids, and that the virus can infect, replicate and activate immune responses in organoids successfully, suggesting that 2D and 3D jejunal organoids, as in vitro models, have broader application prospects beyond porcine enteric coronaviruses. Yan et al. [115] reported that apical-out enteroids and 2D filter-grown intestinal epithelial cells but not basolateral-out enteroids were more susceptible to PRV infection. Guo et al. [116] used a 3D IO model to identify the important roles of cell surface glycans in PRV infections.

Table 2 Porcine IOs in virus infection

Because the interior of the IOs is an intestinal cavity and the exterior is wrapped in Matrigel, this structure could limit viral infection. To address this problem, researchers have developed several solutions: (1) Change the 3D structure to a 2D structure. Cell suspension of IOs was resuspended in medium during passage and then plated in Matrigel precoated tissue culture plates or seeded on permeable filter supports using the Transwell system [84, 85, 115, 119]. Compared with plating in tissue culture plates, the Transwell system has advantages in studying cell migration and invasion, and cell–cell interactions [120]. (2) Use microinjection technology to inject microorganisms into the cavities of organoids [121]. (3) Establish the apical-out organoid model. Li et al. [111] have successfully established porcine apical-out IOs, in which TGEV infection can be effective, inducing type I and type III IFN antiviral responses and inflammatory responses.

Limitations of porcine airway organoids (AOs) and intestinal organoids (IOs) in studies with viruses and future perspectives

By recapitulating the intricate cellular organization and microenvironments found in real organs, these organoids are critical for virological research. Although porcine organoids have many advantages, similar to all other model systems, there are important limitations to be considered. Airways and IOs maintain cellular diversity but lack other cell types critical to airway and intestinal function, including immune cells, lymphocytes, endothelial cells, neurons, smooth muscle, and fibroblasts. To address these limitations, many efforts have been made to develop co-cultural organoid models that can be cultured with other cell types [122]. These models allow for a more comprehensive study of interactions between virus-infected epithelial cells and other non-epithelial cells, including monocytes, dendritic cells, lymphocytes, stromal cells, adipocytes, endothelial cells, and neurons. As co-culture systems continue to be refined, we believe that more valuable platforms will be developed for studying pathogenesis beyond the epithelium. Additionally, organoids can be employed in preclinical drug development and toxicology studies, bridging the gap between traditional cell-based assays and animal testing. With the development of this technology, organoids-on-chip will provide a valuable high-throughput screening platform for drug discovery and medical research.

The development of PSC-derived organoids containing mesenchymal cells is also important for increasing organoid complexity. The successful cultivation of porcine PSCs makes it possible to induce the differentiation of stem cells into organoids [78]. In recent years, with the rapid rise of gene editing technology, the potential for generating genetically engineered swine organoids had opened exciting avenues for studying gene function, gene editing, and regenerative medicine applications. At present, studies have successfully transduced lentiviral vectors into porcine IOs, and other researchers have used CRISPR Cas9 technology for gene editing in organoids, but this technology has not been used in porcine organoids [123,124,125,126]. The application of bioprinting technology has accelerated the construction process of organoids. Bioprinting can construct 3D living organs and tissues by designing and selectively distributing cells, bioactive materials, and cytokines to print specific structures of organoids quickly and accurately, which provides a new technology for simulating the in vivo microenvironment to a higher degree [127]. Given that the core and surface of organoids are separated, and the nutrients required for growing cells in the core and the waste produced are limited, microfluidic technologies that can improve material transport and produce more uniform organoids have been developed to overcome these limitations [128]. However, these techniques have not yet been reported in porcine organoids.

Conclusion

Organoids provide an ideal in vitro model for basic research on porcine viral diseases, which is developing rapidly in terms of complexity and standardization. As the field of organoid technology continues to evolve, porcine AOs and IOs are of great value in elucidating the mechanisms of virus recognition by cells, virus entry and replication, and virus-host interactions, and can provide a new theoretical basis for disease treatment and prevention.

Availability of data and materials

Not applicable.

Abbreviations

AOs:

Airway organoids

IOs:

Intestinal organoids

iPSCs:

Induced pluripotent stem cells

PSCs:

Pluripotent stem cells

ASCs:

Adult tissue stem cells

BMP:

Bone morphogenic protein

AT1:

Type I alveolar epithelial

AT2:

Type II alveolar epithelial

PEDV:

Porcine epidemic diarrhea virus

PRRSV:

Porcine reproductive and respiratory syndrome virus

ESCs:

Embryonic stem cells

TGEV:

Transmissible gastroenteritis coronavirus

IFN:

Interferon

ISCs:

Intestinal stem cells

TA:

Termed transit-amplifying

IESCs:

Intestinal epithelial stem cells

PRV:

Porcine rotavirus

PRCoV:

Porcine respiratory coronavirus

PDCoV:

Porcine deltacoronavirus

ALI:

Air–liquid interface

MRV3:

Mammalian orthoreovirus type 3

3D:

Three-dimensional

2D:

Two-dimensional

References

  1. Bojkovski J, Prodanov-Radulović J, Prodanović R, Vujanac I, Nedić S, Zdravković N, et al. Common pig diseases on commercial farms: a review. In: Lucrări Ştiinţifice Medicină Veterinară Timişoara. Timisoara: Agroprint; 2018. p. 14–27.

  2. Vanderwaal K, Deen J. Global trends in infectious diseases of swine. Proc Natl Acad Sci U S A. 2018;115:11495–500.

  3. Goede D, Morrison RB. Production impact & time to stability in sow herds infected with porcine epidemic diarrhea virus (PEDV). Prev Vet Med. 2016;123:202–7.

    Article  PubMed  Google Scholar 

  4. Liu J, Liu B, Shan B, Wei S, An T, Shen G, et al. Prevalence of African Swine Fever in China, 2018–2019. J Med Virol. 2020;92:1023–34.

  5. Bals R, Hiemstra P. Innate immunity in the lung: how epithelial cells fight against respiratory pathogens. Eur Respir J. 2004;23:327–33.

    Article  CAS  PubMed  Google Scholar 

  6. Bowen RA. The gastrointestinal barrier. In: Wingfield WE, Raffe MR, editors. The veterinary ICU book. London: CRC Press; 2020. p. 40–46.

  7. Song D, Cahn D, Duncan GA. Mucin biopolymers and their barrier function at airway surfaces. Langmuir. 2020;36:12773–83.

    Article  CAS  PubMed  Google Scholar 

  8. Krejci J, Nechvatalova K, Blahutkova M, Faldyna M. The respiratory tract in pigs and its immune system: a review. Vet Med Czech. 2013;58:206–20.

    Article  Google Scholar 

  9. Yang S, Yang N, Huang X, Li Y, Liu G, Jansen CA, et al. Pigs’ intestinal barrier function is more refined with aging. Dev Comp Immunol. 2022;136:104512.

    Article  CAS  PubMed  Google Scholar 

  10. Barron SL, Saez J, Owens RM. In vitro models for studying respiratory host–pathogen interactions. Advanced Biology. 2021;5:2000624.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ayyavoo V. Modeling human viral diseases: trials and triumphs. Front Virol. 2021;1:722297.

  12. Corrò C, Novellasdemunt L, Li VS. A brief history of organoids. Am J Physiol Cell Physiol. 2020;319:C151–65.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Mead BE, Karp JM. All models are wrong, but some organoids may be useful. Genome Biol. 2019;20:66.

  14. Hofer M, Lutolf MP. Engineering organoids. Nat Rev Mater. 2021;6:402–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lehmann R, Lee CM, Shugart EC, Benedetti M, Charo RA, Gartner Z, et al. Human organoids: a new dimension in cell biology. Mol Biol Cell. 2019;30:1129–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol. 2020;21:571–84.

  17. Yin X, Mead BE, Safaee H, Langer R, Karp JM, Levy O. Engineering stem cell organoids. Cell Stem Cell. 2016;18:25–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fuchs E, Blau HM. Tissue stem cells: architects of their niches. Cell Stem Cell. 2020;27:532–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Li P, Li Y, Wang Y, Liu J, Lavrijsen M, Li Y, et al. Recapitulating hepatitis E virus–host interactions and facilitating antiviral drug discovery in human liver–derived organoids. Sci Adv. 2022;8:eabj5908.

  20. Salahudeen AA, Choi SS, Rustagi A, Zhu J, Van Unen V, De La OSM, et al. Progenitor identification and SARS-CoV-2 infection in human distal lung organoids. Nature. 2020;588:670–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yin L, Chen J, Li L, Guo S, Xue M, Zhang J, et al. Aminopeptidase N expression, not interferon responses, determines the intestinal segmental tropism of porcine deltacoronavirus. J Virol. 2020;94(14):e00480–20.

  22. Li L, Fu F, Guo S, Wang H, He X, Xue M, et al. Porcine intestinal enteroids: a new model for studying enteric coronavirus porcine epidemic diarrhea virus infection and the host innate response. J Virol. 2019;93(5):e01682–18.

  23. De Oliveira LF, Mendes Filho D, Marques BL, Maciel GF, Parreira RC, Do Carmo Neto JR. Organoids as a novel tool in modelling infectious diseases. Semin Cell Dev Biol. 2023;144:87–96.

  24. Duque-Correa MA, Maizels RM, Grencis RK, Berriman M. Organoids–new models for host–helminth interactions. Trends Parasitol. 2020;36:170–81.

  25. Derricott H, Luu L, Fong WY, Hartley CS, Johnston LJ, Armstrong SD, et al. Developing a 3D intestinal epithelium model for livestock species. Cell Tissue Res. 2019;375:409–24.

    Article  PubMed  Google Scholar 

  26. Li Y, Yang N, Chen J, Huang X, Zhang N, Yang S, et al. Next-generation porcine intestinal organoids: an apical-out organoid model for swine enteric virus infection and immune response investigations. J Virol. 2021;95:e00006–21.

  27. Resende TP, Medida RL, Vannucci FA, Saqui-Salces M, Gebhart C. Evaluation of swine enteroids as in vitro models for Lawsonia intracellularis infection. J Anim Sci. 2020;98:skaa011.

  28. Bar-Ephraim YE, Kretzschmar K, Clevers H. Organoids in immunological research. Nat Rev Immunol. 2020;20:279–93.

    Article  CAS  PubMed  Google Scholar 

  29. Biton M, Haber AL, Rogel N, Burgin G, Beyaz S, Schnell A, et al. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell. 2018;175:1307–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. De Lau W, Kujala P, Schneeberger K, Middendorp S, Li VS, Barker N, et al. Peyer’s patch M cells derived from Lgr5(+) stem cells require SpiB and are induced by RankL in cultured "miniguts". Mol Cell Biol. 2012;32:3639–47.

  31. Jiang X, Oyang L, Peng Q, Liu Q, Xu X, Wu N, et al. Organoids: opportunities and challenges of cancer therapy. Front Cell Dev Biol. 2023;11:1232528.

  32. Xue Z, Zhao J. Bioelectric interface technologies in cells and organoids. Adv Mater Interfaces. 2023;10:2300550.

  33. Olayanju A, Jones L, Greco K, Goldring CE, Ansari T. Application of porcine gastrointestinal organoid units as a potential in vitro tool for drug discovery and development. J Appl Toxicol. 2019;39:4–15.

    Article  CAS  PubMed  Google Scholar 

  34. Beumer J, Clevers H. Cell fate specification and differentiation in the adult mammalian intestine. Nat Rev Mol Cell Biol. 2021;22:39–53.

    Article  CAS  PubMed  Google Scholar 

  35. Clevers H. Modeling development and disease with organoids. Cell. 2016;165:1586–97.

    Article  CAS  PubMed  Google Scholar 

  36. Meurens F, Summerfield A, Nauwynck H, Saif L, Gerdts V. The pig: a model for human infectious diseases. Trends Microbiol. 2012;20:50–7.

    Article  CAS  PubMed  Google Scholar 

  37. Davis JD, Wypych TP. Cellular and functional heterogeneity of the airway epithelium. Mucosal Immunol. 2021;14:978–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Blundell R. The biology of small airway epithelium. Int J Mol Med Adv Sci. 2006;2:354–9.

    Google Scholar 

  39. Chen Y, Dong Y, Du X. Lung development: AT1 and AT2 property. Biocell. 2020;44:1–5.

    Article  CAS  Google Scholar 

  40. Rajan A, Weaver AM, Aloisio GM, Jelinski J, Johnson HL, Venable SF, et al. The human nose organoid respiratory virus model: an ex vivo human challenge model to study respiratory syncytial virus (RSV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis and evaluate therapeutics. MBio. 2022;13:e03511–e3521.

  41. Rijsbergen L, Lamers M, Comvalius A, Koutstaal R, Schipper D, Duprex W, et al. Human respiratory syncytial virus subgroup A and B infections in nasal, bronchial, small-airway, and organoid-derived respiratory cultures. mSphere. 2021;6(3):e00237–21.

  42. Li C, Huang J, Yu Y, Wan Z, Chiu MC, Liu X, et al. Human airway and nasal organoids reveal escalating replicative fitness of SARS-CoV-2 emerging variants. Proc Natl Acad Sci U S A. 2023;120:e2300376120.

  43. Peng L, Gao L, Wu X, Fan Y, Liu M, Chen J, et al. Lung organoids as model to study SARS-CoV-2 infection. Cells. 2022;11:2758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wang R, Mccauley KB, Kotton DN, Hawkins F. Differentiation of human airway-organoids from induced pluripotent stem cells (iPSCs). Methods Cell Biol. 2020;159:95–114.

    Article  CAS  PubMed  Google Scholar 

  45. Shi Y, Inoue H, Wu JC, Yamanaka S. Induced pluripotent stem cell technology: a decade of progress. Nat Rev Drug Discovery. 2017;16:115–30.

    Article  CAS  PubMed  Google Scholar 

  46. Jung J-H, Yang S-R, Kim WJ, Rhee CK, Hong S-H. Human pluripotent stem cell-derived alveolar organoids: cellular heterogeneity and maturity. Tuberc Respir Dis. 2024;87:52.

  47. Tiwari SK, Wang S, Smith D, Carlin AF, Rana TM. Revealing tissue-specific SARS-CoV-2 infection and host responses using human stem cell-derived lung and cerebral organoids. Stem Cell Reports. 2021;16:437–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Han Y, Duan X, Yang L, Nilsson-Payant BE, Wang P, Duan F, et al. Identification of SARS-CoV-2 inhibitors using lung and colonic organoids. Nature. 2021;589:270–5.

    Article  CAS  PubMed  Google Scholar 

  49. Chen Y-W, Huang SX, De Carvalho ALRT, Ho S-H, Islam MN, Volpi S, et al. A three-dimensional model of human lung development and disease from pluripotent stem cells. Nat Cell Biol. 2017;19:542–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lamers MM, Van Der Vaart J, Knoops K, Riesebosch S, Breugem TI, Mykytyn AZ, et al. An organoid-derived bronchioalveolar model for SARS-CoV-2 infection of human alveolar type II-like cells. EMBO J. 2021;40:e105912.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Katsura H, Sontake V, Tata A, Kobayashi Y, Edwards CE, Heaton BE, et al. Human lung stem cell-based alveolospheres provide insights into SARS-CoV-2-mediated interferon responses and pneumocyte dysfunction. Cell Stem Cell. 2020;27:890–904.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wang T, Zhang N, Fan S, Zhao L, Song W, Gong Y, et al. Establishment of human distal lung organoids for SARS-CoV-2 infection. Cell Discovery. 2021;7:108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Barkauskas CE, Chung M-I, Fioret B, Gao X, Katsura H, Hogan BL. Lung organoids: current uses and future promise. Development. 2017;144:986–97.

  54. Choi J, Iich E, Lee J-H. Organogenesis of adult lung in a dish: differentiation, disease and therapy. Dev Biol. 2016;420:278–86.

  55. Zhou J, Li C, Sachs N, Chiu MC, Wong BH-Y, Chu H, et al. Differentiated human airway organoids to assess infectivity of emerging influenza virus. Proc Natl Acad Sci U S A. 2018;115:6822–7.

  56. Chiu MC, Li C, Liu X, Song W, Wan Z, Yu Y, et al. Human nasal organoids model SARS-CoV-2 upper respiratory infection and recapitulate the differential infectivity of emerging variants. MBio. 2022;13:e01944–e2022.

  57. Chiu MC, Li C, Liu X, Yu Y, Huang J, Wan Z, et al. A bipotential organoid model of respiratory epithelium recapitulates high infectivity of SARS-CoV-2 Omicron variant. Cell discovery. 2022;8:57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Boecking CA, Walentek P, Zlock LT, Sun DI, Wolters PJ, Ishikawa H, et al. A simple method to generate human airway epithelial organoids with externally orientated apical membranes. Am J Physiol Lung Cell Mol Physiol. 2022;322:L420–37.

  59. Shin D-L, Yang W, Peng J-Y, Sawatsky B, Von Messling V, Herrler G, et al. Avian influenza a virus infects swine airway epithelial cells without prior adaptation. Viruses. 2020;12:589.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Stadejek W, Chiers K, Van Reeth K. Infectivity and transmissibility of an avian H3N1 influenza virus in pigs. Vet Res. 2023;54:4.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Peng J-Y, Shin D-L, Li G, Wu N-H, Herrler G. Time-dependent viral interference between influenza virus and coronavirus in the infection of differentiated porcine airway epithelial cells. Virulence. 2021;12:1111–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Jiang C, Li L, Xue M, Zhao L, Liu X, Wang W, et al. Long-term expanding porcine airway organoids provide insights into the pathogenesis and innate immunity of porcine respiratory coronavirus infection. J Virol. 2022;96:e00738–e822.

  63. Guevara-Garcia A, Soleilhac M, Minc N, Delacour D. Regulation and functions of cell division in the intestinal tissue. Semin Cell Dev Biol. 2023;150–151:3–14.

    Article  PubMed  Google Scholar 

  64. Santos AJ, Lo Y-H, Mah AT, Kuo CJ. The intestinal stem cell niche: homeostasis and adaptations. Trends Cell Biol. 2018;28:1062–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mori-Akiyama Y, Van Den Born M, Van Es JH, Hamilton SR, Adams HP, Zhang J, et al. SOX9 is required for the differentiation of paneth cells in the intestinal epithelium. Gastroenterology. 2007;133:539–46.

  66. Milano J, Mckay J, Dagenais C, Foster-Brown L, Pognan F, Gadient R, et al. Modulation of notch processing by γ-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation. Toxicol Sci. 2004;82:341–58.

    Article  CAS  PubMed  Google Scholar 

  67. He XC, Zhang J, Tong W-G, Tawfik O, Ross J, Scoville DH, et al. BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt–β-catenin signaling. Nat Genet. 2004;36:1117–21.

    Article  CAS  PubMed  Google Scholar 

  68. Wallach T, Bayrer JR. Intestinal organoids: new frontiers in the study of intestinal disease and physiology. J Pediatr Gastroenterol Nutr. 2017;64:180.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Sato T, Vries RG, Snippert HJ, Van De Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–5.

    Article  CAS  PubMed  Google Scholar 

  70. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van Den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762–72.

    Article  CAS  PubMed  Google Scholar 

  71. Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470:105–9.

    Article  PubMed  Google Scholar 

  72. Chandra L, Borcherding DC, Kingsbury D, Atherly T, Ambrosini YM, Bourgois-Mochel A, et al. Derivation of adult canine intestinal organoids for translational research in gastroenterology. BMC Biol. 2019;17:1–21.

    Article  Google Scholar 

  73. Gonzalez LM, Williamson I, Piedrahita JA, Blikslager AT, Magness ST. Cell lineage identification and stem cell culture in a porcine model for the study of intestinal epithelial regeneration. PLoS ONE. 2013;8:e66465.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kramer N, Pratscher B, Meneses A, Tschulenk W, Walter I, Swoboda A, et al. Generation of differentiating and long-living intestinal organoids reflecting the cellular diversity of canine intestine. Cells. 2020;9:822.

  75. Powell RH, Behnke MS. WRN conditioned media is sufficient for in vitro propagation of intestinal organoids from large farm and small companion animals. Biology open. 2017;6:698–705.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Zhou J, Li C, Liu X, Chiu MC, Zhao X, Wang D, et al. Infection of bat and human intestinal organoids by SARS-CoV-2. Nat Med. 2020;26:1077–83.

    Article  CAS  PubMed  Google Scholar 

  77. Tsuruta S, Uchida H, Akutsu H. Intestinal organoids generated from human pluripotent stem cells. JMA journal. 2020;3:9–19.

    PubMed  Google Scholar 

  78. Gao X, Nowak-Imialek M, Chen X, Chen D, Herrmann D, Ruan D, et al. Establishment of porcine and human expanded potential stem cells. Nat Cell Biol. 2019;21:687–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhang M, Lv L, Cai H, Li Y, Gao F, Yu L, et al. Long-term expansion of porcine intestinal organoids serves as an in vitro model for swine enteric coronavirus infection. Front Microbiol. 2022;13:865336.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Yin Y, Liu P-Y, Shi Y, Li P. Single-cell sequencing and organoids: a powerful combination for modelling organ development and diseases. Rev Physiol Biochem Pharmacol. 2021;179:189–210.

  81. Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol. 2005;15:378–86.

  82. Czerwinski M, Spence JR. Hacking the matrix. Cell Stem Cell. 2017;20:9–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kozlowski MT, Crook CJ, Ku HT. Towards organoid culture without Matrigel. Communications biology. 2021;4:1387.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Hoffmann P, Schnepel N, Langeheine M, Künnemann K, Grassl GA, Brehm R, et al. Intestinal organoid-based 2D monolayers mimic physiological and pathophysiological properties of the pig intestine. PLoS ONE. 2021;16:e0256143.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Vermeire B, Gonzalez LM, Jansens RJ, Cox E, Devriendt B. Porcine small intestinal organoids as a model to explore ETEC–host interactions in the gut. Vet Res. 2021;52:94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Barnett AM, Mullaney JA, Hendriks C, Le Borgne L, Mcnabb WC, Roy NC. Porcine colonoids and enteroids keep the memory of their origin during regeneration. Am J Physiol Cell Physiol. 2021;320:C794–805.

    Article  CAS  PubMed  Google Scholar 

  87. Kar SK, Te Pas MF, Rikkers R, Madsen O, Taverne N, Ellen ED, et al. Variabilities and similarities of adult stem cells derived intestinal organoids originating from different intestinal segments in pig. BioRxiv [Preprint]. 2022. https://doi.org/10.1101/2022.12.14.520382.

  88. Duchesne C, Randuineau G, Le Normand L, Rome V, Laraqui S, Arnaud AP, et al. Initial pig developmental stage influences intestinal organoid growth but not phenotype. BioRxiv [Preprint]. 2024. https://doi.org/10.1101/2024.01.26.577507.

  89. Miyabe FM, Dall Agnol AM, Leme RA, Oliveira TES, Headley SA, Fernandes T, et al. Porcine rotavirus B as primary causative agent of diarrhea outbreaks in newborn piglets. Sci Rep. 2020;10:22002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Li C, Xu H, Zhao J, Gong B, Sun Q, Xiang L, et al. Epidemiological investigation and genetic evolutionary analysis of PRRSV-1 on a pig farm in China. Front Microbiol. 2022;13:1067173.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Zhang Y, Chen Y, Zhou J, Wang X, Ma L, Li J, et al. Porcine epidemic diarrhea virus: an updated overview of virus epidemiology, virulence variation patterns and virus-host interactions. Viruses. 2022;14:2434.

  92. Jung K, Miyazaki A, Hu H, Saif LJ. Susceptibility of porcine IPEC-J2 intestinal epithelial cells to infection with porcine deltacoronavirus (PDCoV) and serum cytokine responses of gnotobiotic pigs to acute infection with IPEC-J2 cell culture-passaged PDCoV. Vet Microbiol. 2018;221:49–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Wang X, Fang L, Liu S, Ke W, Wang D, Peng G, et al. Susceptibility of porcine IPI-2I intestinal epithelial cells to infection with swine enteric coronaviruses. Vet Microbiol. 2019;233:21–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Edwards CE, Tata A, Baric RS. Human lung organoids as a model for respiratory virus replication and countermeasure performance in human hosts. Transl Res. 2022;250:36–45.

    Article  CAS  PubMed  Google Scholar 

  95. Sachs N, Papaspyropoulos A, Zomer-Van Ommen DD, Heo I, Böttinger L, Klay D, et al. Long-term expanding human airway organoids for disease modeling. EMBO J. 2019;38:e100300.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Huff S, Kummetha IR, Tiwari SK, Huante MB, Clark AE, Wang S, et al. Discovery and mechanism of SARS-CoV-2 main protease inhibitors. J Med Chem. 2021;65:2866–79.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Stroulios G, Brown T, Moreni G, Kondro D, Dei A, Eaves A, et al. Apical-out airway organoids as a platform for studying viral infections and screening for antiviral drugs. Sci Rep. 2022;12:7673.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Aloisio GM, Nagaraj D, Murray AM, Schultz EM, Mcbride T, Aideyan L, et al. Pediatric human nose organoids demonstrate greater susceptibility, epithelial responses, and cytotoxicity than adults during RSV infection. BioRxiv [Preprint]. 2024. https://doi.org/10.1101/2024.02.01.578466.

  99. Khatri M, Goyal SM, Saif YM. Oct4+ stem/progenitor swine lung epithelial cells are targets for influenza virus replication. J Virol. 2012;86:6427–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Punyadarsaniya D, Liang C-H, Winter C, Petersen H, Rautenschlein S, Hennig-Pauka I, et al. Infection of differentiated porcine airway epithelial cells by influenza virus: differential susceptibility to infection by porcine and avian viruses. PLoS ONE. 2011;6:e28429.

  101. Atanasova K, Gucht SV, Barbé F, Lefebvre D, Chiers K, Reeth KV. Lung cell tropism and inflammatory cytokine-profile of porcine respiratory coronavirus infection. Open Vet Sci J. 2008;2:117–26.

  102. Jung K, Renukaradhya GJ, Alekseev KP, Fang Y, Tang Y, Saif LJ. Porcine reproductive and respiratory syndrome virus modifies innate immunity and alters disease outcome in pigs subsequently infected with porcine respiratory coronavirus: implications for respiratory viral co-infections. J Gen Virol. 2009;90:2713–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Wang G, He Y, Tu Y, Liu Y, Zhou E-M, Han Z, et al. Comparative analysis of apoptotic changes in peripheral immune organs and lungs following experimental infection of piglets with highly pathogenic and classical porcine reproductive and respiratory syndrome virus. Virol J. 2014;11:1–5.

    Article  Google Scholar 

  104. Liu C, Zhang W, Gong W, Zhang D, She R, Xu B, et al. Comparative respiratory pathogenicity and dynamic tissue distribution of Chinese highly pathogenic porcine reproductive and respiratory syndrome virus and its attenuated strain in piglets. J Comp Pathol. 2015;153:38–49.

    Article  CAS  PubMed  Google Scholar 

  105. Gómez-Laguna J, Salguero FJ, Pallarés FJ, Carrasco L. Immunopathogenesis of porcine reproductive and respiratory syndrome in the respiratory tract of pigs. Vet J. 2013;195:148–55.

    Article  PubMed  Google Scholar 

  106. Zhang F, Luo S, Gu J, Li Z, Li K, Yuan W, et al. Prevalence and phylogenetic analysis of porcine diarrhea associated viruses in southern China from 2012 to 2018. BMC Vet Res. 2019;15:1–9.

    Article  Google Scholar 

  107. Shen X, Yin L, Pan X, Zhao R, Zhang D. Porcine epidemic diarrhea virus infection blocks cell cycle and induces apoptosis in pig intestinal epithelial cells. Microb Pathog. 2020;147:104378.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Dutta D, Clevers H. Organoid culture systems to study host–pathogen interactions. Curr Opin Immunol. 2017;48:15–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Zhang S, Zhang S, Hou Y, Huang Y, Cai J, Wang G, et al. Porcine deltacoronavirus infection disrupts the intestinal mucosal barrier and inhibits intestinal stem cell differentiation to goblet cells via the notch signaling pathway. J Virol. 2023;97:e0068923.

  110. Yang N, Zhang Y, Fu Y, Li Y, Yang S, Chen J, et al. Transmissible gastroenteritis virus infection promotes the self-renewal of porcine intestinal stem cells via Wnt/β-Catenin pathway. J Virol. 2022;96:e00962–e1022.

  111. Li Y, Yang N, Chen J, Huang X, Zhang N, Yang S, et al. Next-generation porcine intestinal organoids: an apical-out organoid model for swine enteric virus infection and immune response investigations. J Virol. 2020;94:e01006–20.

  112. Yin L, Liu X, Hu D, Luo Y, Zhang G, Liu P. Swine enteric coronaviruses (PEDV, TGEV, and PDCoV) induce divergent interferon-stimulated gene responses and antigen presentation in porcine intestinal enteroids. Front Immunol. 2022;12:826882.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Luo H, Zheng J, Chen Y, Wang T, Zhang Z, Shan Y, et al. Utility evaluation of porcine enteroids as PDCoV infection model in vitro. Front Microbiol. 2020;11:821.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Lee S-A, Lee HJ, Gu N-Y, Park Y-R, Kim E-J, Kang S-J, et al. Evaluation of porcine intestinal organoids as an in vitro model for mammalian orthoreovirus 3 infection. J Vet Sci. 2023;24:e53.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Yan M, Su A, Pavasutthipaisit S, Spriewald R, Graßl GA, Beineke A, et al. Infection of porcine enteroids and 2D differentiated intestinal epithelial cells with rotavirus A to study cell tropism and polarized immune response. Emerg Microbes Infect. 2023;12:2239937.

  116. Guo Y, Candelero-Rueda RA, Saif LJ, Vlasova AN. Infection of porcine small intestinal enteroids with human and pig rotavirus A strains reveals contrasting roles for histo-blood group antigens and terminal sialic acids. PLoS Pathog. 2021;17:e1009237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Yang Q-Y, Yang Y-L, Tang Y-X, Qin P, Wang G, Xie J-Y, et al. Bile acids promote the caveolae-associated entry of swine acute diarrhea syndrome coronavirus in porcine intestinal enteroids. PLoS Pathog. 2022;18:e1010620.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Guo Y, Raev S, Kick MK, Raque M, Saif LJ, Vlasova AN. Rotavirus C replication in porcine intestinal enteroids reveals roles for cellular cholesterol and Sialic Acids. Viruses. 2022;14:1825.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Van Der Hee B, Loonen L, Taverne N, Taverne-Thiele J, Smidt H, Wells J. Optimized procedures for generating an enhanced, near physiological 2D culture system from porcine intestinal organoids. Stem Cell Res. 2018;28:165–71.

    Article  PubMed  Google Scholar 

  120. Barnett AM, Mullaney JA, Mcnabb WC, Roy NC. Culture media and format alter cellular composition and barrier integrity of porcine colonoid-derived monolayers. Tissue Barriers. 2024;12:2222632.

  121. Beaumont M, Blanc F, Cherbuy C, Egidy G, Giuffra E, Lacroix-Lamandé S, et al. Intestinal organoids in farm animals. Vet Res. 2021;52:1–15.

    Article  Google Scholar 

  122. Hentschel V, Seufferlein T, Armacki M. Intestinal organoids in coculture: redefining the boundaries of gut mucosa ex vivo modeling. Am J Physiol Gastrointest Liver Physiol. 2021;321:G693–704.

  123. Khalil HA, Lei NY, Brinkley G, Scott A, Wang J, Kar UK, et al. A novel culture system for adult porcine intestinal crypts. Cell Tissue Res. 2016;365:123–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Ramakrishna G, Babu PE, Singh R, Trehanpati N. Application of CRISPR-Cas9 based gene editing to study the pathogenesis of colon and liver cancer using organoids. Hep Intl. 2021;15:1309–17.

    Article  Google Scholar 

  125. Gopal S, Rodrigues AL, Dordick JS. Exploiting CRISPR Cas9 in three-dimensional stem cell cultures to model disease. Front Bioeng Biotechnol. 2020;8:692.

  126. Fujii M, Clevers H, Sato T. Modeling human digestive diseases with CRISPR-Cas9–modified organoids. Gastroenterology. 2019;156:562–76.

    Article  CAS  PubMed  Google Scholar 

  127. Groll J, Boland T, Blunk T, Burdick JA, Cho D-W, Dalton PD, et al. Biofabrication: reappraising the definition of an evolving field. Biofabrication. 2016;8:013001.

    Article  PubMed  Google Scholar 

  128. Velasco V, Shariati SA, Esfandyarpour R. Microtechnology-based methods for organoid models. Microsyst Nanoeng. 2020;6:76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by the grant from the Chinese Universities Scientific Fund (2023RC046) and North Carolina Agricultural Research Services. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

C.L.: data curation, investigation, and writing. X.D.: data curation and investigation; P.L.: conceptualization, resources, validation, and supervision. X.L.: conceptualization, supervision, edition, and revision. All authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Pinghuang Liu or Xi Lin.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, C., Dong, X., Liu, P. et al. Advances in porcine respiratory and intestinal organoids: status and potential application for virus infections. One Health Adv. 2, 22 (2024). https://doi.org/10.1186/s44280-024-00052-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s44280-024-00052-0

Keywords