1
|
Baruta G, Flannigan KL, Alston L, Thorne A, Zhang H, De Buck J, Colarusso P, Hirota SA. Mycobacterium avium subspecies paratuberculosis targets M cells in enteroid-derived monolayers through interactions with β1 integrins. Am J Physiol Gastrointest Liver Physiol 2025; 328:G482-G501. [PMID: 40112014 DOI: 10.1152/ajpgi.00250.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/09/2024] [Accepted: 02/16/2025] [Indexed: 03/22/2025]
Abstract
Paratuberculosis is an infectious disease caused by the bacterium, Mycobacterium avium subspecies paratuberculosis (MAP). MAP infection of ruminants triggers progressive wasting disease characterized by granulomatous lymphadenitis, enteritis, and severe intestinal pathology that often requires early culling of the animal. The resulting economic burden is significant, and MAP exposure in the workplace constitutes a significant zoonotic risk. Although it has been established that the MAP propagates within resident immune cells, less is known about how it traverses the epithelium. It is currently thought that MAP infects the small intestinal epithelium by targeting both enterocytes and M cells, with a potential tropism for the latter. In the current study, we developed and validated an enteroid-based in vitro assay containing functional M cells to identify the target cells for MAP's entry. Upon exposure to MAP, the bacteria were detected within both enterocytes and M cells; however, quantitative image analysis revealed significant tropism for the latter. Complementary studies using the Caco-2/Raji-B coculture system provided similar results. Since other mycobacteria have been shown to initiate cell attachment and entry by using a fibronectin-bridging process, we tested whether these interactions were involved in MAP's targeting of M cells. We found that MAP's M cell tropism was enhanced by fibronectin and that this effect was abolished when monolayers were pretreated with an integrin-blocking peptide. Our data demonstrate that MAP preferentially targets M cells and that this involves a fibronectin-bridging process. Furthermore, our study supports the utility of M cell-containing enteroids to study host-pathogen interaction at the intestinal epithelium.NEW & NOTEWORTHY We developed and validated a novel enteroid-based in vitro infection model with functional M cells and incorporated leading-edge imaging approaches to determine how MAP interacts with the intestinal epithelium. Using this model, we found that MAP preferentially enters M cells and that this process is enhanced by fibronectin opsonization and interactions with M cell-associated b1 integrins-the so-called fibronectin bridging mechanism that is used by other Mycobacterium to mediate cell attachment and entry.
Collapse
Affiliation(s)
- Grace Baruta
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Kyle L Flannigan
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Laurie Alston
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Andrew Thorne
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Hong Zhang
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jeroen De Buck
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pina Colarusso
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Simon A Hirota
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
2
|
Galli G, Melcón-Fernández E, de Garnica García MG, Martínez-Fernández B, Dehnavi M, Andrés S, Pérez-Pertejo Y, Reguera RM, García-Estrada C, Martínez-Valladares M, Balaña-Fouce R. Development of Sheep Duodenum Intestinal Organoids and Implementation of High-Throughput Screening Platform for Veterinary Applications. Int J Mol Sci 2025; 26:3452. [PMID: 40244396 PMCID: PMC11989482 DOI: 10.3390/ijms26073452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
New therapeutic molecules for farm animals are needed to address worldwide problems in the food industry, like the rise of resistance among ruminant parasites and pathogenic microbes. Since in vivo testing would involve an excessive number of animals, with consequent ethical and economic issues, the generation of sheep intestinal organoids represents a promising close-to-reality in vitro model for veterinary drug development; however, the characterization and application of such organoids remain limited. In this study, ovine intestinal organoids were generated from adult LGR5+ stem cells from the intestinal crypts of freshly slaughtered lambs, and developed in an in vitro culture system. Morphological analysis via brightfield microscopy and immunocytochemical staining revealed a pseudostratified epithelium with multiple cell types, and distinct apical-basal polarity, while RNA sequencing validated the preservation of the physiological characteristics of the original organ. The development and characterization of a robust and reproducible protocol for culturing sheep duodenum intestinal organoids in a high-throughput screening (HTS) compatible format demonstrated reliability in HTS applications, with Z'-factor tests indicating robust assay performance. Dose-response studies using pre-identified compounds showed comparable pharmacodynamic profiles between mouse and sheep organoids. These findings establish sheep intestinal organoids as an innovative tool for veterinary pharmacology and toxicology, offering a cost-effective and sustainable platform to address challenges such as drug resistance and improve livestock health.
Collapse
Affiliation(s)
- Giulio Galli
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24007 León, Spain; (G.G.); (E.M.-F.); (Y.P.-P.); (R.M.R.); (C.G.-E.)
| | - Estela Melcón-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24007 León, Spain; (G.G.); (E.M.-F.); (Y.P.-P.); (R.M.R.); (C.G.-E.)
| | | | | | - Mahsa Dehnavi
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, Finca Marzanas s/n, Grulleros, 24346 León, Spain; (M.D.); (S.A.); (M.M.-V.)
| | - Sonia Andrés
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, Finca Marzanas s/n, Grulleros, 24346 León, Spain; (M.D.); (S.A.); (M.M.-V.)
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24007 León, Spain; (G.G.); (E.M.-F.); (Y.P.-P.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24007 León, Spain
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24007 León, Spain; (G.G.); (E.M.-F.); (Y.P.-P.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24007 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24007 León, Spain; (G.G.); (E.M.-F.); (Y.P.-P.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24007 León, Spain
| | - María Martínez-Valladares
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, Finca Marzanas s/n, Grulleros, 24346 León, Spain; (M.D.); (S.A.); (M.M.-V.)
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24007 León, Spain; (G.G.); (E.M.-F.); (Y.P.-P.); (R.M.R.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24007 León, Spain
| |
Collapse
|
3
|
Shin DL, Tsai YB, Hsu SH, Liang CC, Wu NH. Chicken intestinal organoids reveal polarity-dependent replication dynamics and immune responses of low pathogenic avian influenza viruses. Poult Sci 2025; 104:104921. [PMID: 40043674 PMCID: PMC11927706 DOI: 10.1016/j.psj.2025.104921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 03/24/2025] Open
Abstract
Low pathogenic avian influenza viruses (LPAIVs) persist in poultry populations, posing an ongoing challenge to poultry management and research. These viruses typically cause mild infections but can lead to significant economic losses due to their widespread presence and potential to disrupt poultry production. Traditional in vivo and in vitro models struggle to accurately replicate the avian intestinal environment, where these viruses often establish infection. In Taiwan, the domestic H6N1 LPAIVs cause an endemic in the local area but still lack investigation. This study addresses this gap by utilizing advanced chicken intestinal organoid (CIO) systems, apical-out (Ap-o), and basal-out (Ba-o) conformations to study the unique replication kinetics and innate immune responses of LPAIVs in a physiologically relevant setting. By comparing the Taiwan specialized H6N1 strain toward the Eurasian H9N2 virus, our results demonstrate that Ap-o organoids, which mimic natural exposure to the intestinal lumen, elicit robust interferon-stimulated gene responses, particularly higher expression of downstream gene, which effectively controls viral replication against H6N1 virus. In contrast, Ba-o organoids, representing a systemic infection route, exhibited lower upstream interferon responses, reflecting a different immune response pattern in the H9N2 strain. These results confirm that CIO is a well-suited model to study LPAIV pathogenesis. It provides key insights into the host-pathogen interactions that determine viral replication and immune evasion strategies. This model deepens our understanding of LPAIV behavior in poultry and provides a valuable tool for developing more targeted and effective control strategies for poultry health management.
Collapse
Affiliation(s)
- Dai-Lun Shin
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan
| | - Yi-Bei Tsai
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan; Department and Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei City 106, Taiwan
| | - Shu-Han Hsu
- Department and Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei City 106, Taiwan
| | - Chi-Chia Liang
- Department and Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei City 106, Taiwan
| | - Nai-Huei Wu
- Department and Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei City 106, Taiwan.
| |
Collapse
|
4
|
Fukutome A, Sakamoto T, Asawa Y, Riu D, Kawakami H, Hoshi K, Hikita A. Establishment of a mouse organ culture model of fetal cleft lip for the evaluation of adipose-derived stem cell therapy. Regen Ther 2025; 28:41-50. [PMID: 39687332 PMCID: PMC11647479 DOI: 10.1016/j.reth.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Cleft lip and cleft palate are congenital disorders resulting from abnormal facial development. Current treatments require multiple surgeries, which have risks of scar formation and facial deformities. Recently, fetal treatments utilizing "scarless healing" have gained attention, as early intervention shows potential to suppress scarring. In the field of regenerative medicine, mesenchymal stem cell therapies using cell sheets have advanced, by which promotion of tissue repair is expected. However, researches for fetal treatment using small animal models of cleft lip are challenging due to the high fetal mortality caused by surgical invasiveness. Although organ culture methods may offer an alternative approach, no organ culture system for fetal cleft lip research has been reported. Methods In this study, a cleft lip was surgically created on the upper left side lip of E15.5 mouse fetuses. These fetuses were cultured for four days using an organ culture system. Histological evaluation was performed to evaluate cell density, tissue morphology, and epithelialization. Additionally, adipose-derived stem cell (ADSC) sheets were transplanted two days after cleft lip creation to evaluate their effect on tissue repair. Results The histological analysis showed that cell density and tissue morphology were stably maintained in the four-day culture period. Epithelialization of the incision site was observed two days after surgery, confirming the completion of cleft formation. In the ADSC-transplanted group, epithelialization of the cleft site was observed, which indicates that the stem cell sheets contributed to tissue repair. Conclusion This research demonstrates the successful development of a cleft lip organ culture model and highlights the potential of ADSC sheets in promoting tissue repair. These findings provide a foundation for future regenerative medicine strategies in fetal cleft lip therapy.
Collapse
Affiliation(s)
- Ayane Fukutome
- Department of Oral and Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomoaki Sakamoto
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yukiyo Asawa
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Dan Riu
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hiroshi Kawakami
- Division of Dentistry and Oral Surgery, Mitsui Memorial Hospital, 1 Kanda Izumicho, Chiyoda-ku, Tokyo 101-8643, Japan
| | - Kazuto Hoshi
- Department of Oral and Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
5
|
Galli G, Ruiz-Somacarrera M, González del Palacio L, Melcón-Fernández E, González-Pérez R, García-Estrada C, Martinez-Valladares M, Balaña-Fouce R. High-Throughput Screening of Five Compound Libraries for Anthelmintic Activity and Toxicity Leads to the Discovery of Two Flavonoid Compounds. Int J Mol Sci 2025; 26:1595. [PMID: 40004065 PMCID: PMC11855827 DOI: 10.3390/ijms26041595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Gastrointestinal nematode infections (GINs) in ruminants are a major constraint to efficient livestock production worldwide. Currently, only a limited number of anthelmintic drugs are available for the control of these infections, but their widespread use in preventive deworming campaigns and the incorrect administration of the drugs are responsible for the emergence of resistance. Therefore, new anthelmintic drugs are urgently needed. However, drug discovery methods for new anthelmintics based on GINs isolated from ruminants often have low throughput. In this study, a screening of five commercial collections of chemical compounds, including one collection of anti-infective drugs, three plant-based natural product collections, and one collection from the FDA-approved Chinese Pharmacopoeia, with a total of 2228 molecules, have been carried out in a high-throughput format. In the single slot screen, 32 compounds (1.44% success rate) achieved a >70% motility inhibition rate. Of these, 10 are known anthelmintic drugs, while the remaining 22 were tested against Haemonchus contortus and a resistant strain of Teladorsagia circumcincta. Four compounds (two flavonoids, chalcone and trans-chalcone), and two anti-infectives (octenidine and tolfenpyrad), showed anthelmintic activity with EC50 values below 20 µM, and were further tested for their safety against HepG2 spheroids and mouse intestinal organoids. Trans-chalcone and chalcone emerged as promising candidates for future development, showing selective indexes > 5, while tolfenpyrad and octenidine require careful evaluation due to their toxicity profiles.
Collapse
Affiliation(s)
- Giulio Galli
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (G.G.); (E.M.-F.); (R.G.-P.); (C.G.-E.)
| | - Marta Ruiz-Somacarrera
- Departamento Sanidad Animal, Instituto de Ganadería de Montaña, CSIC-Universidad de León, Grulleros, 24346 León, Spain; (M.R.-S.); (L.G.d.P.)
| | - Laura González del Palacio
- Departamento Sanidad Animal, Instituto de Ganadería de Montaña, CSIC-Universidad de León, Grulleros, 24346 León, Spain; (M.R.-S.); (L.G.d.P.)
| | - Estela Melcón-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (G.G.); (E.M.-F.); (R.G.-P.); (C.G.-E.)
| | - Rubén González-Pérez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (G.G.); (E.M.-F.); (R.G.-P.); (C.G.-E.)
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (G.G.); (E.M.-F.); (R.G.-P.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Maria Martinez-Valladares
- Departamento Sanidad Animal, Instituto de Ganadería de Montaña, CSIC-Universidad de León, Grulleros, 24346 León, Spain; (M.R.-S.); (L.G.d.P.)
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (G.G.); (E.M.-F.); (R.G.-P.); (C.G.-E.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
6
|
Wang H, He X, Zhang M, Fan N, Yang Z, Shen T, Guo J, Song Y, Cao G, Liu Y, Li X, Nashun B. Development of Sheep Intestinal Organoids for Studying Deoxynivalenol-Induced Toxicity. Int J Mol Sci 2025; 26:955. [PMID: 39940725 PMCID: PMC11816529 DOI: 10.3390/ijms26030955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Sheep are an important livestock species whose gastrointestinal tract is essential for overall health. Feed contaminants such as bacterial toxins and mycotoxins severely damage the sheep intestine, yet the mechanisms remain mostly elusive partially due to the lack of physiologically relevant in vitro models. Here, we investigated molecular mechanisms underlying deoxynivalenol (DON)-induced toxicity by developing intestinal organoids from isolated intestinal crypts of Hu sheep. The organoids had a central lumen and monolayer epithelium, and could be continuously passaged, cryopreserved, and resuscitated. Histological and transcriptomic analysis showed that the intestinal organoids recapitulate the cell lineages and gene expression characteristics of the original intestinal tissues. Statistical analysis indicated that DON exposure significantly inhibited organoid formation efficiency, as well as the proliferation and activity of intestinal organoid cells. RNA-seq and Western blotting analysis further revealed that DON exposure induces intestinal toxicity by inhibiting the PI3K/AKT/GSK3β/β-catenin signaling pathway. Our study provides a novel example of organoid application in toxicity studies and reveals the signaling pathway involved in DON-induced toxicity in sheep, which is of great significance for improving mitigation strategies for DON.
Collapse
Affiliation(s)
- Hongyu Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Xige He
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Miaomiao Zhang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Na Fan
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Zongxuan Yang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Ting Shen
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Jiaojiao Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Yongli Song
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Guifang Cao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot 011517, China
| | - Yongbin Liu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Xihe Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot 011517, China
| | - Buhe Nashun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| |
Collapse
|
7
|
Premachandre CK, Quah PS, Tran BM, Vincan E, Deliyannis G, Wong CY, Diaz-Méndez A, Jackson DC, Reading PC, Browning GF, Vaz PK, Wawegama NK. Bovine tracheal organoids for studying Mycoplasma bovis respiratory infections. Vet Microbiol 2025; 300:110340. [PMID: 39675119 DOI: 10.1016/j.vetmic.2024.110340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/08/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
In vitro three-dimensional organoid models simulate key aspects of the structure and function of in vivo organs and have been used to study physiology, host-pathogen interactions, pathogenesis and pharmacodynamics. Although most organoid studies have been developed using human or mouse tissues, recent advancements have enabled the establishment of intestinal and respiratory tract organoids from domestic animal samples. Mycoplasma bovis causes chronic respiratory tract infections in cattle with significant health and economic consequences. The pathogenesis and virulence factors of M. bovis have been studied in several in vitro infection models, but the use of organoids has not been examined previously. In this study, we assessed the feasibility of using a matrix-embedded bovine tracheal organoid system to study respiratory infections with M. bovis. Bovine tracheal organoids were inoculated with M. bovis strain MbovMil and incubated for 72 hours to investigate the ability of M. bovis to proliferate, attach and invade the organoids. M. bovis was able to infect the organoids, resulting in a mean 260-fold increase in the titre of viable M. bovis by 72 hours post-inoculation. Examination of the infected organoids using transmission electron microscopy revealed the presence of mycoplasmas within the organoid cells and membrane bound clusters of M. bovis inside the intercellular junctions. Our findings indicate that bovine tracheal organoids can be used as a model system for studying respiratory tract infections caused by M. bovis.
Collapse
Affiliation(s)
- Chintha K Premachandre
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Pin Shie Quah
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Bang Manh Tran
- Department of Infectious Diseases, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Elizabeth Vincan
- Department of Infectious Diseases, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Georgia Deliyannis
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Andrés Diaz-Méndez
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - David C Jackson
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne at the Peter Doherty institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Glenn F Browning
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Paola K Vaz
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nadeeka K Wawegama
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
8
|
Yang HW, Hu T, Ait-Ali T. Lawsonia intracellularis regulates nuclear factor-κB signalling pathway during infection. PLoS One 2024; 19:e0310804. [PMID: 39325775 PMCID: PMC11426430 DOI: 10.1371/journal.pone.0310804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Lawsonia intracellularis is the etiological agent of proliferative enteropathy (PE) in pigs, horses and wide range of mammals. Little is known about the role of innate immune response during L. intracellularis infection. In this study, we investigated the nuclear factor-κB (NF-κB)-regulated immune response against infection of a clinical strain Dkp23 and a live-attenuated Enterisol vaccine strain in PK-15 cells. We found that expression of NF-κB target genes TNF-α, IFN-γ, IL-6 and IL-8 were modulated during the course of infection. At 5 dpi, there was a significant increase in p65 NF-κB activation, including protein nuclear translocation and phosphorylation, synchronous with the induction of IL-6, IFN-γ and IL-8 expression in L. intracellularis infected cells, especially for Enterisol vaccine strain-infected cells. This result suggests that NF-κB signalling level is induced when L. intracellularis bacterial load peaks at 5 dpi. The induction of pro-inflammatory cytokines expression is consistent with the decreased viability of L. intracellularis-infected cells especially that of the vaccine strain. There were no significant changes in NF-κB signalling between vaccine and Dkp23 infection in PK-15 cells, except for moderate levels of differences in NF-κB target genes expression which might be a reflection of differences in intracellular bacterial load. Overall, the data presented here indicate a correlation between the induction of NF-κB signalling and the L. intracellularis bacterial load in PK-15 cells.
Collapse
Affiliation(s)
- Huan W Yang
- Department of Biochemistry, The University of Illinois Champaign-Urbana, Champaign, IL, United States of America
| | - Tuanjun Hu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | | |
Collapse
|
9
|
Zheng X, Wang Y, Zhou X, Deng T, Zhao Y, Fu Z, Wei Y, Ma W, Zhang S, Guan W, Chen F. Plant essential oils combined with organic acids restored lipopolysaccharide-induced leaky intestine via gut microbial modulation in weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:419-432. [PMID: 39309969 PMCID: PMC11415868 DOI: 10.1016/j.aninu.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 09/25/2024]
Abstract
Intestine derived lipopolysaccharide (LPS) is closely related to systemic inflammation and disorders, yet little is known about its roles in the weanling stress of piglets and its potential as a nutritional intervention target. This study aimed to investigate the potential of essential oils (EO) and organic acids (OA) in mitigating weaning stress in piglets by modulating the circulation of intestine derived LPS. Seventy-two weaned piglets at 21 d old with body weight of 8.12 ± 0.168 kg were randomly divided into a control group (CON) and an experimental group, each consisting of six pens with six piglets per pen, and were fed either a basal diet or a basal diet supplemented with 3 kg/t OA + 500 g/t EO (EO + OA). On the 14th day of the feeding trial, 12 weaned piglets were randomly selected from the CON group, and 6 piglets were selected from the experimental group. Based on diet composition and stress treatment, these 18 piglets were divided into the following three groups: 1) CON group. Piglets were fed a basal diet and received an intraperitoneal injection of saline as a control. 2) LPS group. Piglets were fed a basal diet and received an intraperitoneal injection of LPS (100 μg/kg body weight) to induce stress. 3) EO + OA + LPS group. Piglets were fed a basal diet supplemented with EO and OA and received an intraperitoneal injection of LPS (100 μg/kg body weight) to induce stress. The results showed that EO + OA significantly ameliorated the oxidative imbalance and inflammation disorder induced by LPS in piglets' serum and intestine by inhibiting the activation of the Toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB)/mitogen-activated protein kinase (MAPK) signaling pathway. Furthermore, compared to the LPS group, supplementation with EO + OA restored LPS-induced reductions in Bcl-2 protein expression in the piglets' intestines (P < 0.05) and mitigated morphological damage; it also enhanced both the protein expression and relative gene expression of the tight junction proteins occludin and claudin-1 (P < 0.05), and reduced the plasma diamine oxidase activity (DAO) and LPS content (P < 0.05). Compared to the CON group, supplementation with EO + OA altered the composition of the intestinal microbiota, increasing beneficial bacteria relative abundance (Faecalibacterium) (P < 0.05) and decreasing harmful bacteria relative abundance [Rikenellaceae_RC9_gut_group (P < 0.01), Negativibacillus (P < 0.05)]. Further analysis revealed that plasma LPS content in piglets was negatively correlated with the relative abundance of Faecalibacterium (r = -0.662, P = 0.021), Akkermansia (r = -0.492, P = 0.031), and average daily gain (ADG) (r = -0.912, P = 0.041). Plasma LPS content was also positively correlated with the plasma inflammatory factors interleukin (IL)-1β (r = 0.591, P = 0.021), IL-6 (r = 0.623, P = 0.021), IL-12 (r = 561, P = 0.031) contents, and the relative abundance of Negativibacillus (r = 0.712, P = 0.041). In summary, the addition of EO + OA prevents the leakage of intestine derived LPS into the circulation by improving intestinal integrity and microbiota composition, thereby enhancing antioxidant and anti-inflammatory abilities and growth performance of weaned piglets.
Collapse
Affiliation(s)
- Xiaoyu Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yibo Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xuemei Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tanyi Deng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yueqi Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhichao Fu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yulong Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wen Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Fang Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
10
|
Yan Y, Zhu S, Jia M, Chen X, Qi W, Gu F, Valencak TG, Liu JX, Sun HZ. Advances in single-cell transcriptomics in animal research. J Anim Sci Biotechnol 2024; 15:102. [PMID: 39090689 PMCID: PMC11295521 DOI: 10.1186/s40104-024-01063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/12/2024] [Indexed: 08/04/2024] Open
Abstract
Understanding biological mechanisms is fundamental for improving animal production and health to meet the growing demand for high-quality protein. As an emerging biotechnology, single-cell transcriptomics has been gradually applied in diverse aspects of animal research, offering an effective method to study the gene expression of high-throughput single cells of different tissues/organs in animals. In an unprecedented manner, researchers have identified cell types/subtypes and their marker genes, inferred cellular fate trajectories, and revealed cell‒cell interactions in animals using single-cell transcriptomics. In this paper, we introduce the development of single-cell technology and review the processes, advancements, and applications of single-cell transcriptomics in animal research. We summarize recent efforts using single-cell transcriptomics to obtain a more profound understanding of animal nutrition and health, reproductive performance, genetics, and disease models in different livestock species. Moreover, the practical experience accumulated based on a large number of cases is highlighted to provide a reference for determining key factors (e.g., sample size, cell clustering, and cell type annotation) in single-cell transcriptomics analysis. We also discuss the limitations and outlook of single-cell transcriptomics in the current stage. This paper describes the comprehensive progress of single-cell transcriptomics in animal research, offering novel insights and sustainable advancements in agricultural productivity and animal health.
Collapse
Affiliation(s)
- Yunan Yan
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Senlin Zhu
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Minghui Jia
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyi Chen
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wenlingli Qi
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Fengfei Gu
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Teresa G Valencak
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Agency for Health and Food Safety Austria, 1220, Vienna, Austria
| | - Jian-Xin Liu
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hui-Zeng Sun
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
11
|
Wang Q, Guo F, Zhang Q, Hu T, Jin Y, Yang Y, Ma Y. Organoids in gastrointestinal diseases: from bench to clinic. MedComm (Beijing) 2024; 5:e574. [PMID: 38948115 PMCID: PMC11214594 DOI: 10.1002/mco2.574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/15/2024] [Accepted: 04/26/2024] [Indexed: 07/02/2024] Open
Abstract
The etiology of gastrointestinal (GI) diseases is intricate and multifactorial, encompassing complex interactions between genetic predisposition and gut microbiota. The cell fate change, immune function regulation, and microenvironment composition in diseased tissues are governed by microorganisms and mutated genes either independently or through synergistic interactions. A comprehensive understanding of GI disease etiology is imperative for developing precise prevention and treatment strategies. However, the existing models used for studying the microenvironment in GI diseases-whether cancer cell lines or mouse models-exhibit significant limitations, which leads to the prosperity of organoids models. This review first describes the development history of organoids models, followed by a detailed demonstration of organoids application from bench to clinic. As for bench utilization, we present a layer-by-layer elucidation of organoid simulation on host-microbial interactions, as well as the application in molecular mechanism analysis. As for clinical adhibition, we provide a generalized interpretation of organoid application in GI disease simulation from inflammatory disorders to malignancy diseases, as well as in GI disease treatment including drug screening, immunotherapy, and microbial-targeting and screening treatment. This review draws a comprehensive and systematical depiction of organoids models, providing a novel insight into the utilization of organoids models from bench to clinic.
Collapse
Affiliation(s)
- Qinying Wang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Fanying Guo
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qinyuan Zhang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - TingTing Hu
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - YuTao Jin
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yongzhi Yang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yanlei Ma
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
12
|
Yan J, Monlong J, Cougoule C, Lacroix-Lamandé S, Wiedemann A. Mapping the scientific output of organoids for animal and human modeling infectious diseases: a bibliometric assessment. Vet Res 2024; 55:81. [PMID: 38926765 PMCID: PMC11210181 DOI: 10.1186/s13567-024-01333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/11/2024] [Indexed: 06/28/2024] Open
Abstract
The escalation of antibiotic resistance, pandemics, and nosocomial infections underscores the importance of research in both animal and human infectious diseases. Recent advancements in three-dimensional tissue cultures, or "organoids", have revolutionized the development of in vitro models for infectious diseases. Our study conducts a bibliometric analysis on the use of organoids in modeling infectious diseases, offering an in-depth overview of this field's current landscape. We examined scientific contributions from 2009 onward that focused on organoids in host‒pathogen interactions using the Web of Science Core Collection and OpenAlex database. Our analysis included temporal trends, reference aging, author, and institutional productivity, collaborative networks, citation metrics, keyword cluster dynamics, and disruptiveness of organoid models. VOSviewer, CiteSpace, and Python facilitated this analytical assessment. The findings reveal significant growth and advancements in organoid-based infectious disease research. Analysis of keywords and impactful publications identified three distinct developmental phases in this area that were significantly influenced by outbreaks of Zika and SARS-CoV-2 viruses. The research also highlights the synergistic efforts between academia and publishers in tackling global pandemic challenges. Through mostly consolidating research efforts, organoids are proving to be a promising tool in infectious disease research for both human and animal infectious disease. Their integration into the field necessitates methodological refinements for better physiological emulation and the establishment of extensive organoid biobanks. These improvements are crucial for fully harnessing the potential of organoids in understanding infectious diseases and advancing the development of targeted treatments and vaccines.
Collapse
Affiliation(s)
- Jin Yan
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China.
- Research Center of Digestive Disease, Central South University, Changsha, China.
- IRSD - Digestive Health Research Institute, University of Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France.
| | - Jean Monlong
- IRSD - Digestive Health Research Institute, University of Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Céline Cougoule
- Institut de Pharmacologie Et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Agnès Wiedemann
- IRSD - Digestive Health Research Institute, University of Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France.
| |
Collapse
|
13
|
Xu G, Qiao Z, Schraauwen R, Avan A, Peppelenbosch MP, Bijvelds MJC, Jiang S, Li P. Evidence for cross-species transmission of human coronavirus OC43 through bioinformatics and modeling infections in porcine intestinal organoids. Vet Microbiol 2024; 293:110101. [PMID: 38718529 DOI: 10.1016/j.vetmic.2024.110101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 05/15/2024]
Abstract
Cross-species transmission of coronaviruses has been continuously posing a major challenge to public health. Pigs, as the major animal reservoirs for many zoonotic viruses, frequently mediate viral transmission to humans. This study comprehensively mapped the relationship between human and porcine coronaviruses through in-depth bioinformatics analysis. We found that human coronavirus OC43 and porcine coronavirus PHEV share a close phylogenetic relationship, evidenced by high genomic homology, similar codon usage patterns and comparable tertiary structure in spike proteins. Inoculation of infectious OC43 viruses in organoids derived from porcine small and large intestine demonstrated that porcine intestinal organoids (pIOs) are highly susceptible to human coronavirus OC43 infection and support infectious virus production. Using transmission electron microscopy, we visualized OC43 viral particles in both intracellular and extracellular compartments, and observed abnormalities of multiple organelles in infected organoid cells. Robust OC43 infections in pIOs result in a significant reduction of organoids viability and widespread cell death. This study bears essential implications for better understanding the evolutionary origin of human coronavirus OC43, and provides a proof-of-concept for using pIOs as a model to investigate cross-species transmission of human coronavirus.
Collapse
Affiliation(s)
- Guige Xu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong 271018, China; Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Zhiwen Qiao
- State Key Laboratory of Crop Biology, College of Horticulture Science and Engineering, Shandong Agricultural University, Taian 271018, China
| | - Rick Schraauwen
- Department of Pathology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Amine Avan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Shijin Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong 271018, China.
| | - Pengfei Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
14
|
Kadekar D, Udrea AC, Bak SY, Christensen N, Gibbs K, Shen C, Bernardeau M. Cell-Free Culture Supernatant of Lactobacillus acidophilus AG01 and Bifidobacterium animalis subsp. lactis AG02 Reduces the Pathogenicity of NetB-Positive Clostridium perfringens in a Chicken Intestinal Epithelial Cell Line. Microorganisms 2024; 12:839. [PMID: 38674783 PMCID: PMC11052021 DOI: 10.3390/microorganisms12040839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/27/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The worldwide reduction in the use of antibiotics in animal feed is fueling the need for alternatives for the prevention and control of poultry intestinal diseases such as necrotic enteritis (NE), which is caused by Clostridium perfringens. This is the first report on the use of an intestinal epithelial chicken cell line (CHIC-8E11) to study the pathogenic traits of C. perfringens and to investigate the mode of action of cell-free supernatants (CFS) from probiotic Lactobacillus acidophilus AG01 and Bifidobacterium animalis subsp. lactis AG02 in reducing the pathogenicity of C. perfringens. The cell adhesion, permeability and cytotoxicity were assessed under challenge with four C. perfringens strains isolated from broiler NE episodes of differing geographical origin (CP1-UK; CP10-Sweden; 25037-CP01 and CP22-USA). All the C. perfringens strains could adhere to the CHIC-8E11 cells, with varying affinity (0.05-0.48% adhesion across the strains). The CFS from one out of two strains (CP22) increased the cell permeability (+4.5-fold vs. the control, p < 0.01), as measured by the fluorescein isothiocyanate-dextran (FD4) content, with NetB toxin implicated in this effect. The CFS from all the strains was cytotoxic against the CHIC-8E11 cells in a dose- and strain-dependent manner (cytotoxicity 23-62% across the strains when dosed at 50 µL/mL, as assessed by the MTT cell viability assay). Pre-treatment of the cells with CFS from B. animalis subsp. lactis AG02 but not L. acidophilus AG01 reduced the cell adhesion of three out of four C. perfringens strains (by 77-85% vs. the control, p < 0.001) and reduced the negative effect of two NetB-positive strains on the cell permeability. The CFS of both probiotics alleviated the cytotoxicity of all the C. perfringens strains, which was dependent on the dose. The results confirm the suitability of the CHIC-8E11 cell line for the study of host-pathogen cell interactions in the context of NE caused by C. perfringens and reveal a beneficial mode of action of B. animalis subsp. lactis AG02 in reducing C. perfringens cell adhesion and, together with L. acidophilus AG01, in reducing C. perfringens cytotoxicity.
Collapse
Affiliation(s)
- Darshana Kadekar
- Gut Immunology Lab, R&D, Health & Biosciences, IFF, 8220 Brabrand, Denmark (A.C.U.)
| | | | - Steffen Yde Bak
- IFF Advanced Analysis, R&D, ET, IFF, 8220 Brabrand, Denmark; (S.Y.B.); (N.C.)
| | - Niels Christensen
- IFF Advanced Analysis, R&D, ET, IFF, 8220 Brabrand, Denmark; (S.Y.B.); (N.C.)
| | - Kirsty Gibbs
- Danisco Animal Nutrition, IFF, 2342 BH Oegstgeest, The Netherlands;
| | - Chong Shen
- Gut Immunology Lab, R&D, Health & Biosciences, IFF, 8220 Brabrand, Denmark (A.C.U.)
| | - Marion Bernardeau
- Danisco Animal Nutrition, IFF, 2342 BH Oegstgeest, The Netherlands;
- Agro-Food Department, Normandy University, UNICAEN, ABTE, 14000 Caen, France
| |
Collapse
|
15
|
Hellman S, Martin F, Tydén E, Sellin ME, Norman A, Hjertner B, Svedberg P, Fossum C. Equine enteroid-derived monolayers recapitulate key features of parasitic intestinal nematode infection. Vet Res 2024; 55:25. [PMID: 38414039 PMCID: PMC10900620 DOI: 10.1186/s13567-024-01266-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/03/2024] [Indexed: 02/29/2024] Open
Abstract
Stem cell-derived organoid cultures have emerged as attractive experimental models for infection biology research regarding various types of gastro-intestinal pathogens and host species. However, the large size of infectious nematode larvae and the closed structure of 3-dimensional organoids often hinder studies of the natural route of infection. To enable easy administration to the apical surface of the epithelium, organoids from the equine small intestine, i.e. enteroids, were used in the present study to establish epithelial monolayer cultures. These monolayers were functionally tested by stimulation with IL-4 and IL-13, and/or exposure to infectious stage larvae of the equine nematodes Parascaris univalens, cyathostominae and/or Strongylus vulgaris. Effects were recorded using transcriptional analysis combined with histochemistry, immunofluorescence-, live-cell- and scanning electron microscopy. These analyses revealed heterogeneous monolayers containing both immature and differentiated cells including tuft cells and mucus-producing goblet cells. Stimulation with IL-4/IL-13 increased tuft- and goblet cell differentiation as demonstrated by the expression of DCLK1 and MUC2. In these cytokine-primed monolayers, the expression of MUC2 was further promoted by co-culture with P. univalens. Moreover, live-cell imaging revealed morphological alterations of the epithelial cells following exposure to larvae even in the absence of cytokine stimulation. Thus, the present work describes the design, characterization and usability of an experimental model representing the equine nematode-infected small intestinal epithelium. The presence of tuft cells and goblet cells whose mucus production is affected by Th2 cytokines and/or the presence of larvae opens up for mechanistic studies of the physical interactions between nematodes and the equine intestinal mucosa.
Collapse
Affiliation(s)
- Stina Hellman
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, SLU, P.O. Box 7028, 750 07, Uppsala, Sweden.
| | - Frida Martin
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, SLU, P.O. Box 7028, 750 07, Uppsala, Sweden
| | - Eva Tydén
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, SLU, P.O. Box 7028, 750 07, Uppsala, Sweden
| | - Mikael E Sellin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Albin Norman
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, SLU, P.O. Box 7028, 750 07, Uppsala, Sweden
| | - Bernt Hjertner
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, SLU, P.O. Box 7028, 750 07, Uppsala, Sweden
| | - Pia Svedberg
- Vidilab AB, P.O. Box 33, 745 21, Enköping, Sweden
| | - Caroline Fossum
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, SLU, P.O. Box 7028, 750 07, Uppsala, Sweden
| |
Collapse
|
16
|
Gabriel V, Zdyrski C, Sahoo DK, Ralston A, Wickham H, Bourgois-Mochel A, Ahmed B, Merodio MM, Paukner K, Piñeyro P, Kopper J, Rowe EW, Smith JD, Meyerholz D, Kol A, Viall A, Elbadawy M, Mochel JP, Allenspach K. Adult Animal Stem Cell-Derived Organoids in Biomedical Research and the One Health Paradigm. Int J Mol Sci 2024; 25:701. [PMID: 38255775 PMCID: PMC10815683 DOI: 10.3390/ijms25020701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Preclinical biomedical research is limited by the predictiveness of in vivo and in vitro models. While in vivo models offer the most complex system for experimentation, they are also limited by ethical, financial, and experimental constraints. In vitro models are simplified models that do not offer the same complexity as living animals but do offer financial affordability and more experimental freedom; therefore, they are commonly used. Traditional 2D cell lines cannot fully simulate the complexity of the epithelium of healthy organs and limit scientific progress. The One Health Initiative was established to consolidate human, animal, and environmental health while also tackling complex and multifactorial medical problems. Reverse translational research allows for the sharing of knowledge between clinical research in veterinary and human medicine. Recently, organoid technology has been developed to mimic the original organ's epithelial microstructure and function more reliably. While human and murine organoids are available, numerous other organoids have been derived from traditional veterinary animals and exotic species in the last decade. With these additional organoid models, species previously excluded from in vitro research are becoming accessible, therefore unlocking potential translational and reverse translational applications of animals with unique adaptations that overcome common problems in veterinary and human medicine.
Collapse
Affiliation(s)
- Vojtech Gabriel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | | | - Dipak K. Sahoo
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
| | - Abigail Ralston
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
| | - Hannah Wickham
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | - Agnes Bourgois-Mochel
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
| | - Basant Ahmed
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | - Maria M. Merodio
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
| | - Karel Paukner
- Atherosclerosis Research Laboratory, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic;
| | - Pablo Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (P.P.); (J.D.S.)
| | - Jamie Kopper
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
| | - Eric W. Rowe
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
| | - Jodi D. Smith
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (P.P.); (J.D.S.)
| | - David Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA;
| | - Amir Kol
- Department of Pathology, University of California, Davis, CA 94143, USA; (A.K.); (A.V.)
| | - Austin Viall
- Department of Pathology, University of California, Davis, CA 94143, USA; (A.K.); (A.V.)
| | - Mohamed Elbadawy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30530, USA;
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (H.W.); (B.A.); (J.P.M.)
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30530, USA;
| | - Karin Allenspach
- 3D Health Solutions Inc., Ames, IA 50010, USA; (C.Z.); (A.R.); (M.M.M.)
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA; (D.K.S.); (A.B.-M.); (J.K.)
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30530, USA;
| |
Collapse
|
17
|
Alberge J, Mussard E, Al-Ayoubi C, Lencina C, Marrauld C, Cauquil L, Achard CS, Mateos I, Alassane-Kpembi I, Oswald IP, Soler L, Combes S, Beaumont M. Butyrate reduces epithelial barrier dysfunction induced by the foodborne mycotoxin deoxynivalenol in cell monolayers derived from pig jejunum organoids. Gut Microbes 2024; 16:2430424. [PMID: 39572558 PMCID: PMC11587856 DOI: 10.1080/19490976.2024.2430424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/30/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
The foodborne mycotoxin deoxynivalenol (DON) produced by Fusarium species threats animal and human health through disruption of the intestinal barrier. Targeting the gut microbiota and its products appears as a promising strategy to mitigate DON intestinal toxicity. In this study, we investigated whether the bacterial metabolite butyrate could alleviate epithelial barrier disruption induced by DON. We used a model of cell monolayers derived from porcine jejunum organoids allowing to reproduce the cellular complexity of the intestinal epithelium. Our results show that DON dose-dependently disrupted the epithelial barrier integrity, reduced epithelial differentiation, and altered innate immune defenses. Butyrate attenuated the DON-induced increase in paracellular permeability. Butyrate also prevented epithelial barrier dysfunction triggered by anisomycin, a ribosome inhibitor like DON. Moreover, butyrate partially counteracted the effects of DON on tight junctions (TJP1, OCLN), innate epithelial defenses (PTGS2, CD14, TLR4, TLR5), and absorptive cell functions (CA2, VIL1, NHE3, CFTR). In contrast, butyrate did not prevent the toxic effects of DON on mitochondrial metabolism, proliferation and goblet cell functions. Taken together, our results demonstrate that the bacterial metabolite butyrate is able to reduce DON-induced epithelial barrier disruption.
Collapse
Affiliation(s)
- Julie Alberge
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Eloïse Mussard
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
- Lallemand Animal Nutrition, Blagnac Cedex, France
| | - Carine Al-Ayoubi
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, Toulouse, France
| | - Corinne Lencina
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | | | - Laurent Cauquil
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | | | - Ivan Mateos
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
- Lallemand Animal Nutrition, Blagnac Cedex, France
- Departamento de Producción Animal, Universidad de León, León, Spain
| | - Imourana Alassane-Kpembi
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, Toulouse, France
- Centre de recherche en infectiologie porcine et avicole (CRIPA), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Isabelle P. Oswald
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, Toulouse, France
| | - Laura Soler
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, Toulouse, France
| | - Sylvie Combes
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Martin Beaumont
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| |
Collapse
|
18
|
Kawasaki M, Dykstra GD, McConnel CS, Burbick CR, Ambrosini YM. Adult Bovine-Derived Small and Large Intestinal Organoids: In Vitro Development and Maintenance. J Tissue Eng Regen Med 2023; 2023:3095002. [PMID: 38873240 PMCID: PMC11175594 DOI: 10.1155/2023/3095002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Recent progress in bovine intestinal organoid research has expanded opportunities for creating improved in vitro models to study intestinal physiology and pathology. However, the establishment of a culture condition capable of generating organoids from all segments of the cattle intestine has remained elusive. Although previous research has described the development of bovine jejunal, ileal, and colonic organoids, this study marks the first report of successful bovine duodenal and rectal organoid development. Maintenance of these organoids through serial passages and cryopreservation was achieved, with higher success rates observed in large intestinal organoids compared to their small intestinal counterparts. A novel approach involving the use of biopsy forceps during initial tissue sampling streamlined the subsequent tissue processing, simplifying the procedure compared to previously established protocols in cattle. Additionally, our study introduced a more cost-effective culture medium based on Advanced DMEM/F12, diverging from frequently used commercially available organoid culture media. This enhancement improves accessibility to organoid technology by reducing culture costs. Crucially, the derived organoids from jejunum, ileum, colon and rectum faithfully preserved the structural, cellular, and genetic characteristics of in vivo intestinal tissue. This research underscores the significant potential of adult bovine intestinal organoids as a physiologically and morphologically relevant in vitro model. Such organoids provide a renewable and sustainable resource for a broad spectrum of studies, encompassing investigations into normal intestinal physiology in cattle and the intricate host-pathogen interactions of clinically and economically significant enteric pathogens.
Collapse
Affiliation(s)
- Minae Kawasaki
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Gerald D Dykstra
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Craig S McConnel
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Claire R Burbick
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Yoko M Ambrosini
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| |
Collapse
|
19
|
Jolly A, Fernández B, Mundo SL, Elguezabal N. Modeling Paratuberculosis in Laboratory Animals, Cells, or Tissues: A Focus on Their Applications for Pathogenesis, Diagnosis, Vaccines, and Therapy Studies. Animals (Basel) 2023; 13:3553. [PMID: 38003170 PMCID: PMC10668694 DOI: 10.3390/ani13223553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Paratuberculosis is a chronic granulomatous enteritis caused by Mycobacterium avium subsp. Paratuberculosis that affects a wide variety of domestic and wild animals. It is considered one of the diseases with the highest economic impact on the ruminant industry. Despite many efforts and intensive research, paratuberculosis control still remains controversial, and the existing diagnostic and immunoprophylactic tools have great limitations. Thus, models play a crucial role in understanding the pathogenesis of infection and disease, and in testing novel vaccine candidates. Ruminant animal models can be restricted by several reasons, related to space requirements, the cost of the animals, and the maintenance of the facilities. Therefore, we review the potential and limitations of the different experimental approaches currently used in paratuberculosis research, focusing on laboratory animals and cell-based models. The aim of this review is to offer a vision of the models that have been used, and what has been achieved or discovered with each one, so that the reader can choose the best model to answer their scientific questions and prove their hypotheses. Also, we bring forward new approaches that we consider worth exploring in the near future.
Collapse
Affiliation(s)
- Ana Jolly
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
| | - Bárbara Fernández
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
- Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
- Instituto de Investigación y Tecnología en Reproducción Animal (INITRA), Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
| | - Silvia Leonor Mundo
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
- Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
- Instituto de Investigación y Tecnología en Reproducción Animal (INITRA), Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
| | - Natalia Elguezabal
- Departamento de Sanidad Animal, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario-Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| |
Collapse
|
20
|
Xu Z, Xu X, Yang B, Mi Y, Wang J. 3D sheep rumen epithelial structures driven from single cells in vitro. Vet Res 2023; 54:104. [PMID: 37946298 PMCID: PMC10636852 DOI: 10.1186/s13567-023-01234-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/22/2023] [Indexed: 11/12/2023] Open
Abstract
Ruminants play a vital economic role as livestock, providing high-quality protein for humans. At present, 3D-cultured ruminant abomasum and intestinal organoids have been successfully established to study host and pathogen interaction. The rumen is a unique digestive organ of ruminants that occupies 70% of the volume of the digestive tract and its microbiota can decompose lignocellulose to support animal growth. Here we report a method for culturing rumen epithelial organoids. We found that single rumen epithelial cells form self-organized 3D structures representative of typical stratified squamous epithelium, which is similar to rumen epithelium. EGF, Noggin, Wnt3a, IGF-1, and FGF-10 significantly enhanced the seeding efficiency of organoids. Moreover, the inclusion of CHIR-99021, A83-01, SB202190, and Y-27632 is crucial for organoid formation and maintenance. Importantly, we demonstrate that rumen epithelial cells retain their ability to form organoids after passage, cryopreservation, and resuscitation. The rumen epithelial organoids express rumen cell type-specific genes, uptake fatty acids, and generate 2D cultures. In summary, our data demonstrate that it is feasible to establish organoids from single rumen epithelial cells, which is a novel in vitro system that may reduce the use of experimental animals.
Collapse
Affiliation(s)
- Zebang Xu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Xinxin Xu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Bin Yang
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, 310023, Zhejiang, China
| | - Yuling Mi
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiakun Wang
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China.
| |
Collapse
|
21
|
Nishihara K, Wood KM, Guan LL, Steele MA. Cultivation of enteroids from fresh and cryopreserved bovine duodenal tissues. JDS COMMUNICATIONS 2023; 4:518-523. [PMID: 38045896 PMCID: PMC10692292 DOI: 10.3168/jdsc.2023-0379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/15/2023] [Indexed: 12/05/2023]
Abstract
This study aimed to develop a method for intestinal tissue cryopreservation and resuscitation for enteroid cultivation. Two different types of tissues, fresh duodenal tissues (n = 3, from Angus steers) and duodenal tissues cryopreserved in 90% fetal bovine serum (FBS) and 10% dimethyl sulfoxide (DMSO; n = 3, from Holstein calves), were collected to develop enteroids. Crypts were isolated using 2 mM EDTA/phosphate-buffered saline from both fresh and cryopreserved tissues and embedded in basement membrane extract. Embedded crypts were seeded in a 24-well plate and cultured in IntestiCult Organoid Growth Medium (Mouse) with inhibitors cocktail and Primocin. The upper opening of crypts became sealed, and crypts formed sphere structures (i.e., enteroids) within 24 h. Primary (passage 0) enteroids showed budding crypt domains from d 3 of cultivation at the earliest. After 7 d of cultivation, enteroids were passaged in a new 24-well plate. Fragments from passaged d 7 enteroids also formed sphere structures within 24 h after seeding and showed budding crypt domains from d 3 of cultivation at the earliest. The area of enteroids was measured in each animal during d 1 to 7 in passage 0 and 1, and the area of enteroids derived from both tissues increased during d 1 to 7 in passage 0 and 1. The area increased from d 1 to 7 of cultivation, and the area of passage 1 was greater than that of passage 0. F-actin staining using phalloidin revealed that brush border microvilli were distributed on the luminal side of the enteroids. In conclusion, a cryopreserved solution consisting of FBS and DMSO is useful for cryopreservation and resuscitation of bovine intestine for enteroid cultivation. This method allows researchers to investigate intestinal function and health in the laboratory using enteroids derived from fresh and cryopreserved tissues collected from cattle.
Collapse
Affiliation(s)
- Koki Nishihara
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON, Canada N1G 1Y2
| | - Katie M. Wood
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON, Canada N1G 1Y2
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada T6G 2P5
| | - Michael A. Steele
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON, Canada N1G 1Y2
| |
Collapse
|
22
|
Liu Y, Tan J, Zhang N, Li W, Fu B. A Strainer-Based Platform for the Collection and Immunolabeling of Porcine Epidemic Diarrhea Virus-Infected Porcine Intestinal Organoid. Int J Mol Sci 2023; 24:15671. [PMID: 37958655 PMCID: PMC10650080 DOI: 10.3390/ijms242115671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
The development of organoid research has raised new requirements for this methodology. In a previous study, we demonstrated that an emerging protocol achieved the collection, loading, and programmed immunolabeling of mouse intestinal organoids based on a strainer platform. To uncover the applied potential of this novel methodology on organoids from other species, the strainer platform was utilized to characterize the porcine epidemic diarrhea virus (PEDV)-infected porcine intestinal organoid model. Based on a previous study, some steps were changed to improve the efficiency of the assay by simplifying the reagent addition procedure. In addition, we redefined the range of strainer sizes on porcine intestinal organoids, showing that strainers with pore sizes of 40 and 70 μm matched the above protocol well. Notably, the strainer platform was successfully used to label viral proteins, laying the foundation for its application in the visualization of viral infection models. In summary, the potential of the strainer platform for organoid technology was explored further. More extensive exploration of this platform will contribute to the development of organoid technology.
Collapse
Affiliation(s)
| | | | | | | | - Baoquan Fu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Public Health of Agriculture Ministry Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Y.L.); (J.T.); (N.Z.); (W.L.)
| |
Collapse
|
23
|
Chen B, Slocombe RF, Georgy SR. Advances in organoid technology for veterinary disease modeling. Front Vet Sci 2023; 10:1234628. [PMID: 37920327 PMCID: PMC10618422 DOI: 10.3389/fvets.2023.1234628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
Organoids are in vitro organ-like structures that faithfully recapitulate many characteristics of a specific organ. During the past decades, major progress has been accomplished in establishing three-dimensional (3D) culture systems toward stem cell-derived organoids. As a significant technological breakthrough, these amazing 3D organoid constructs bridge the conventional 2D in vitro models and in vivo animal models and provide an unprecedented opportunity to investigate the complexities of veterinary diseases ranging from their pathogenesis to the prevention, therapy, or even future organ replacement strategies. In this review, we briefly discuss several definitions used in organoid research and highlight the currently known achievements in modeling veterinary diseases, including infectious and inflammatory diseases, cancers, and metabolic diseases. The applications of organoid technology in veterinary disease modeling are still in their infancy stage but the future is promising.
Collapse
Affiliation(s)
| | | | - Smitha Rose Georgy
- Section of Anatomic Pathology, Melbourne Veterinary School, Faculty of Science, University of Melbourne, Werribee, VIC, Australia
| |
Collapse
|
24
|
Aizpurua O, Blijleven K, Trivedi U, Gilbert MTP, Alberdi A. Unravelling animal-microbiota evolution on a chip. Trends Microbiol 2023; 31:995-1002. [PMID: 37217368 DOI: 10.1016/j.tim.2023.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023]
Abstract
Whether and how microorganisms have shaped the evolution of their animal hosts is a major question in biology. Although many animal evolutionary processes appear to correlate with changes in their associated microbial communities, the mechanistic processes leading to these patterns and their causal relationships are still far from being resolved. Gut-on-a-chip models provide an innovative approach that expands beyond the potential of conventional microbiome profiling to study how different animals sense and react to microbes by comparing responses of animal intestinal tissue models to different microbial stimuli. This complementary knowledge can contribute to our understanding of how host genetic features facilitate or prevent different microbiomes from being assembled, and in doing so elucidate the role of host-microbiota interactions in animal evolution.
Collapse
Affiliation(s)
- Ostaizka Aizpurua
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Kees Blijleven
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Urvish Trivedi
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - M Thomas P Gilbert
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark; University Museum, NTNU, Trondheim, Norway
| | - Antton Alberdi
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Beaumont M, Lencina C, Bertide A, Gallo L, Barilly C, Marrauld C, Cauquil L, Samson A, Combes S. The Early Life Microbiota Is Not a Major Factor Underlying the Susceptibility to Postweaning Diarrhea in Piglets. Microbiol Spectr 2023; 11:e0069423. [PMID: 37358441 PMCID: PMC10433861 DOI: 10.1128/spectrum.00694-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/09/2023] [Indexed: 06/27/2023] Open
Abstract
Postweaning diarrhea (PWD) in piglets impair welfare, induce economic losses and lead to overuse of antibiotics. The early life gut microbiota was proposed to contribute to the susceptibility to PWD. The objective of our study was to evaluate in a large cohort of 116 piglets raised in 2 separate farms whether the gut microbiota composition and functions during the suckling period were associated with the later development of PWD. The fecal microbiota and metabolome were analyzed by 16S rRNA gene amplicon sequencing and nuclear magnetic based resonance at postnatal day 13 in male and female piglets. The later development of PWD was recorded for the same animals from weaning (day 21) to day 54. The gut microbiota structure and α-diversity during the suckling period were not associated with the later development of PWD. There was no significant difference in the relative abundances of bacterial taxa in suckling piglets that later developed PWD. The predicted functionality of the gut microbiota and the fecal metabolome signature during the suckling period were not linked to the later development of PWD. Trimethylamine was the bacterial metabolite which fecal concentration during the suckling period was the most strongly associated with the later development of PWD. However, experiments in piglet colon organoids showed that trimethylamine did not disrupt epithelial homeostasis and is thus not likely to predispose to PWD through this mechanism. In conclusion, our data suggest that the early life microbiota is not a major factor underlying the susceptibility to PWD in piglets. IMPORTANCE This study shows that the fecal microbiota composition and metabolic activity are similar in suckling piglets (13 days after birth) that either later develop post-weaning diarrhea (PWD) or not, which is a major threat for animal welfare that also causes important economic losses and antibiotic treatments in pig production. The aim of this work was to study a large cohort of piglets raised in separates environments, which is a major factor influencing the early life microbiota. One of the main findings is that, although the fecal concentration of trimethylamine in suckling piglets was associated with the later development of PWD, this gut microbiota-derived metabolite did not disrupt the epithelial homeostasis in organoids derived from the pig colon. Overall, this study suggests that the gut microbiota during the suckling period is not a major factor underlying the susceptibility of piglets to PWD.
Collapse
Affiliation(s)
- Martin Beaumont
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Corinne Lencina
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Allan Bertide
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Lise Gallo
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Céline Barilly
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | | | - Laurent Cauquil
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | | | - Sylvie Combes
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| |
Collapse
|
26
|
Lacroix-Lamandé S, Bernardi O, Pezier T, Barilleau E, Burlaud-Gaillard J, Gagneux A, Velge P, Wiedemann A. Differential Salmonella Typhimurium intracellular replication and host cell responses in caecal and ileal organoids derived from chicken. Vet Res 2023; 54:63. [PMID: 37525204 PMCID: PMC10391861 DOI: 10.1186/s13567-023-01189-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/20/2023] [Indexed: 08/02/2023] Open
Abstract
Chicken infection with Salmonella Typhimurium is an important source of foodborne human diseases. Salmonella colonizes the avian intestinal tract and more particularly the caecum, without causing symptoms. This thus poses a challenge for the prevention of foodborne transmission. Until now, studies on the interaction of Salmonella with the avian gut intestine have been limited by the absence of in vitro intestinal culture models. Here, we established intestinal crypt-derived chicken organoids to better decipher the impact of Salmonella intracellular replication on avian intestinal epithelium. Using a 3D organoid model, we observed a significantly higher replication rate of the intracellular bacteria in caecal organoids than in ileal organoids. Our model thus recreates intracellular environment, allowing Salmonella replication of avian epithelium according to the intestinal segment. Moreover, an inhibition of the cellular proliferation was observed in infected ileal and caecal organoids compared to uninfected organoids. This appears with a higher effect in ileal organoids, as well as a higher cytokine and signaling molecule response in infected ileal organoids at 3 h post-infection (hpi) than in caecal organoids that could explain the lower replication rate of Salmonella observed later at 24 hpi. To conclude, this study demonstrates that the 3D organoid is a model allowing to decipher the intracellular impact of Salmonella on the intestinal epithelium cell response and illustrates the importance of the gut segment used to purify stem cells and derive organoids to specifically study epithelial cell -Salmonella interaction.
Collapse
Affiliation(s)
| | | | - Tiffany Pezier
- INRAE, Université de Tours, ISP, 37380, Nouzilly, France
| | | | - Julien Burlaud-Gaillard
- Plateforme IBiSA de Microscopie Électronique, Université de Tours et CHRU de Tours, Tours, France
| | - Anissa Gagneux
- INRAE, Université de Tours, ISP, 37380, Nouzilly, France
| | - Philippe Velge
- INRAE, Université de Tours, ISP, 37380, Nouzilly, France
| | - Agnès Wiedemann
- INRAE, Université de Tours, ISP, 37380, Nouzilly, France.
- IRSD, Institut de Recherche en Santé Digestive, ENVT, INRAE, INSERM, Université́ de Toulouse, UPS, Toulouse, France.
| |
Collapse
|
27
|
Ferreira M, Sousa V, Oliveira B, Canadas-Sousa A, Abreu H, Dias J, Kiron V, Valente LMP. An in-depth characterisation of European seabass intestinal segments for assessing the impact of an algae-based functional diet on intestinal health. Sci Rep 2023; 13:11686. [PMID: 37468554 DOI: 10.1038/s41598-023-38826-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023] Open
Abstract
Sustainable farming of fish species depends on emerging new feed ingredients, which can alter the features of the digestive tract and influence animals' overall health. Recent research has shown that functional feeds hold great potential for enhancing fish robustness by evoking appropriate responses at the intestine level. However, there is a lack of extensive and accurate descriptions of the morphology of the gastrointestinal tract of most farmed fish. We have characterised the intestine of European seabass thoroughly, by targeting four segments - anterior, mid, posterior and rectum. Results indicated that the anterior segment is mostly associated with absorption-related features; this segment has the largest absorptive area, the longest villi, and the highest number of neutral goblet cells (GC). The posterior segment and rectum have distinct histomorphometric features, but both seem to be important for immunity, displaying the highest count of acid GC and the highest expression of immune-related genes. The strongest proliferating cell nuclear antigen (PCNA) signal was observed in the anterior intestine and rectum, with PCNA+ cells appearing at the base of the villi and the corresponding villi branches. We have also evaluated the impact of a novel feed supplemented with a macro- and microalgae blend and found that there were no differences in terms of growth. However, the alterations observed in the mid intestine of fish fed the blend, such as thickening of the submucosa and lamina propria, an increased number of leucocytes, and higher expression of immune- and oxidative stress-related genes, suggest that algae may have an immunomodulatory effect. In the current article, we have described the morphology and expression patterns of the intestine segments of European seabass in detail and have presented a comprehensive report of the indices and methods used for the semi-quantitative and quantitative histomorphometric assessments, thereby providing useful information for future studies that aim to maintain intestinal health through dietary interventions.
Collapse
Affiliation(s)
- Mariana Ferreira
- CIIMAR/CIMAR-LA, Centro Interdisciplinar de Investigação Marinha e Ambiental, 4450-208, Matosinhos, Portugal
- ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade Do Porto, 4050-313, Porto, Portugal
| | - Vera Sousa
- CIIMAR/CIMAR-LA, Centro Interdisciplinar de Investigação Marinha e Ambiental, 4450-208, Matosinhos, Portugal
- ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade Do Porto, 4050-313, Porto, Portugal
| | - Beatriz Oliveira
- CIIMAR/CIMAR-LA, Centro Interdisciplinar de Investigação Marinha e Ambiental, 4450-208, Matosinhos, Portugal
- ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade Do Porto, 4050-313, Porto, Portugal
| | - Ana Canadas-Sousa
- ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade Do Porto, 4050-313, Porto, Portugal
- EUVG, Escola Universitária Vasco da Gama, Quinta de S. Jorge, Estrada da Conraria, Castelo Viegas, 3040-714, Coimbra, Portugal
| | - H Abreu
- ALGAplus, Production and Trading of Seaweed and Derived Products Ltd, 3830-196, Ílhavo, Portugal
| | - J Dias
- SPAROS Lda., 8700-221, Olhão, Portugal
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, 8049, Bodø, Norway
| | - Luisa M P Valente
- CIIMAR/CIMAR-LA, Centro Interdisciplinar de Investigação Marinha e Ambiental, 4450-208, Matosinhos, Portugal. *
- ICBAS, Instituto de Ciências Biomédicas de Abel Salazar, Universidade Do Porto, 4050-313, Porto, Portugal. *
| |
Collapse
|
28
|
Sciascia QL, Metges CC. Review: Methods and biomarkers to investigate intestinal function and health in pigs. Animal 2023; 17 Suppl 3:100860. [PMID: 37316380 DOI: 10.1016/j.animal.2023.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/16/2023] Open
Abstract
Society is becoming increasingly critical of animal husbandry due to its environmental impact and issues involving animal health and welfare including scientific experiments conducted on farm animals. This opens up two new fields of scientific research, the development of non- or minimally invasive (1) methods and techniques using faeces, urine, breath or saliva sampling to replace existing invasive models, and (2) biomarkers reflecting a disease or malfunction of an organ that may predict the future outcome of a pig's health, performance or sustainability. To date, there is a paucity of non- or minimally invasive methods and biomarkers investigating gastrointestinal function and health in pigs. This review describes recent literature pertaining to parameters that assess gastrointestinal functionality and health, tools currently used to investigate them, and the development or the potential to develop new non- and minimally invasive methods and/or biomarkers in pigs. Methods described within this review are those that characterise gastrointestinal mass such as the citrulline generation test, intestinal protein synthesis rate, first pass splanchnic nutrient uptake and techniques describing intestinal proliferation, barrier function and transit rate, and microbial composition and metabolism. An important consideration is gut health, and several molecules with the potential to act as biomarkers of compromised gut health in pigs are reported. Many of these methods to investigate gut functionality and health are considered 'gold standards' but are invasive. Thus, in pigs, there is a need to develop and validate non-invasive methods and biomarkers that meet the principles of the 3 R guidelines, which aim to reduce and refine animal experimentation and replace animals where possible.
Collapse
Affiliation(s)
- Q L Sciascia
- Research Institute for Farm Animal Biology, Institute of Nutritional Physiology "Oskar Kellner", Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - C C Metges
- Research Institute for Farm Animal Biology, Institute of Nutritional Physiology "Oskar Kellner", Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
29
|
Lee SA, Lee HJ, Gu NY, Park YR, Kim EJ, Kang SJ, Hyun BH, Yang DK. Evaluation of porcine intestinal organoids as an in vitro model for mammalian orthoreovirus 3 infection. J Vet Sci 2023; 24:e53. [PMID: 37532298 PMCID: PMC10404702 DOI: 10.4142/jvs.23017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Mammalian orthoreovirus type 3 (MRV3), which is responsible for gastroenteritis in many mammalian species including pigs, has been isolated from piglets with severe diarrhea. However, the use of pig-derived cells as an infection model for swine-MRV3 has rarely been studied. OBJECTIVES This study aims to establish porcine intestinal organoids (PIOs) and examine their susceptibility as an in vitro model for intestinal MRV3 infection. METHODS PIOs were isolated and established from the jejunum of a miniature pig. Established PIOs were characterized using polymerase chain reaction (PCR) and immunofluorescence assays (IFAs) to confirm the expression of small intestine-specific genes and proteins, such as Lgr5, LYZI, Mucin-2, ChgA, and Villin. The monolayered PIOs and three-dimensional (3D) PIOs, obtained through their distribution to expose the apical surface, were infected with MRV3 for 2 h, washed with Dulbecco's phosphate-buffered saline, and observed. Viral infection was confirmed using PCR and IFA. We performed quantitative real-time reverse transcription-PCR to assess changes in viral copy numbers and gene expressions linked to intestinal epithelial genes and antiviral activity. RESULTS The established PIOs have molecular characteristics of intestinal organoids. Infected PIOs showed delayed proliferation with disruption of structures. In addition, infection with MRV3 altered the gene expression linked to intestinal epithelial cells and antiviral activity, and these effects were observed in both 2D and 3D models. Furthermore, viral copy numbers in the supernatant of both models increased in a time-dependent manner. CONCLUSIONS We suggest that PIOs can be an in vitro model to study the infection mechanism of MRV3 in detail, facilitating pharmaceutical development.
Collapse
Affiliation(s)
- Se-A Lee
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Hye Jeong Lee
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Na-Yeon Gu
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Yu-Ri Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Eun-Ju Kim
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Seok-Jin Kang
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Bang-Hun Hyun
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Dong-Kun Yang
- Viral Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Korea.
| |
Collapse
|
30
|
Tian J, Li Y, Bao X, Yang F, Tang X, Jiang Q, Yin Y, Yao K. Early weaning causes small intestinal atrophy by inhibiting the activity of intestinal stem cells: involvement of Wnt/β-catenin signaling. Stem Cell Res Ther 2023; 14:65. [PMID: 37020258 PMCID: PMC10077674 DOI: 10.1186/s13287-023-03293-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/17/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Early weaning and shorter breastfeeding duration are applied by a proportion of young mothers, especially in the social spheres of poverty-stricken areas. Early childhood is a critical period for intestinal development, which is driven by intestinal stem cells (ISCs). However, how early weaning practice affects the function of ISCs to mediate intestinal development remains unclear. METHODS We established an excellent early weaning mice model that has significant intestinal atrophy and growth arrest symptoms to explore the responses of ISCs to early weaning. The primary and passaged intestinal organoids from the suckling or early weaning mice were cultured to explore the underlying mechanism of early weaning affecting the ISCs. RESULTS Early weaning depressed the self-renewal of ISCs and attenuated the activity of ISCs-driven intestinal epithelial regeneration and crypt expansion in vivo and ex-vivo. Further results showed that early weaning retarded the differentiation of ISCs into transit-amplifying cells and Paneth cells, and accelerated the apoptosis of villous epithelial cells, jointly leading to intestinal epithelial atrophy. Mechanistically, early weaning inhibited Wnt signaling in ISCs, while an exogenous Wnt amplifier restored ISCs' function in ex-vivo. CONCLUSION Our findings indicate that early weaning depresses the activity of ISCs via attenuating Wnt/β-catenin signaling and triggers the proinflammatory cytokines TNF-α, IL-1β, IL-6, and IL-17 in jejunum, thereby impeding ISCs-driven epithelial regeneration and intestinal growth, which may provide a basal theory for the development of infant nutrients targeting stem cells to alleviate early weaning-induced intestinal problems.
Collapse
Affiliation(s)
- Junquan Tian
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
| | - Yuying Li
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
| | - Xuetai Bao
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
| | - Fan Yang
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
| | - Xiongzhuo Tang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410000, Hunan, China
| | - Qian Jiang
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410000, Hunan, China.
| | - Yulong Yin
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410000, Hunan, China
| | - Kang Yao
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China.
- University of Chinese Academy of Sciences, Beijing, 100008, China.
| |
Collapse
|
31
|
Penning LC, van den Boom R. Companion animal organoid technology to advance veterinary regenerative medicine. Front Vet Sci 2023; 10:1032835. [PMID: 37008367 PMCID: PMC10063859 DOI: 10.3389/fvets.2023.1032835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
First year medical and veterinary students are made very aware that drugs can have very different effects in various species or even in breeds of one specific species. On the other hand, the “One Medicine” concept implies that therapeutic and technical approaches are exchangeable between man and animals. These opposing views on the (dis)similarities between human and veterinary medicine are magnified in regenerative medicine. Regenerative medicine promises to stimulate the body's own regenerative capacity via activation of stem cells and/or the application of instructive biomaterials. Although the potential is enormous, so are the hurdles that need to be overcome before large scale clinical implementation is realistic. It is in the advancement of regenerative medicine that veterinary regenerative medicine can play an instrumental and crucial role. This review describes the discovery of (adult) stem cells in domesticated animals, mainly cats and dogs. The promise of cell-mediated regenerative veterinary medicine is compared to the actual achievements, and this will lead to a set of unanswered questions (controversies, research gaps, potential developments in relation to fundamental, pre-clinical, and clinical research). For veterinary regenerative medicine to have impact, either for human medicine and/or for domesticated animals, answering these questions is pivotal.
Collapse
|
32
|
Shakya R, Jiménez-Meléndez A, Robertson LJ, Myrmel M. Bovine Enteroids as an In Vitro Model for Infection with Bovine Coronavirus. Viruses 2023; 15:635. [PMID: 36992344 PMCID: PMC10054012 DOI: 10.3390/v15030635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Bovine coronavirus (BCoV) is one of the major viral pathogens of cattle, responsible for economic losses and causing a substantial impact on animal welfare. Several in vitro 2D models have been used to investigate BCoV infection and its pathogenesis. However, 3D enteroids are likely to be a better model with which to investigate host-pathogen interactions. This study established bovine enteroids as an in vitro replication system for BCoV, and we compared the expression of selected genes during the BCoV infection of the enteroids with the expression previously described in HCT-8 cells. The enteroids were successfully established from bovine ileum and permissive to BCoV, as shown by a seven-fold increase in viral RNA after 72 h. Immunostaining of differentiation markers showed a mixed population of differentiated cells. Gene expression ratios at 72 h showed that pro-inflammatory responses such as IL-8 and IL-1A remained unchanged in response to BCoV infection. Expression of other immune genes, including CXCL-3, MMP13, and TNF-α, was significantly downregulated. This study shows that the bovine enteroids had a differentiated cell population and were permissive to BCoV. Further studies are necessary for a comparative analysis to determine whether enteroids are suitable in vitro models to study host responses during BCoV infection.
Collapse
Affiliation(s)
| | | | | | - Mette Myrmel
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), 1430 Ås, Norway
| |
Collapse
|
33
|
Nash TJ, Morris KM, Mabbott NA, Vervelde L. Temporal transcriptome profiling of floating apical out chicken enteroids suggest stability and reproducibility. Vet Res 2023; 54:12. [PMID: 36793124 PMCID: PMC9933378 DOI: 10.1186/s13567-023-01144-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
Enteroids are miniature self-organising three-dimensional (3D) tissue cultures which replicate much of the complexity of the intestinal epithelium. We recently developed an apical-out leukocyte-containing chicken enteroid model providing a novel physiologically relevant in vitro tool to explore host-pathogen interactions in the avian gut. However, the replicate consistency and culture stability have not yet been fully explored at the transcript level. In addition, causes for the inability to passage apical-out enteroids were not determined. Here we report the transcriptional profiling of chicken embryonic intestinal villi and chicken enteroid cultures using bulk RNA-seq. Comparison of the transcriptomes of biological and technical replicate enteroid cultures confirmed their high level of reproducibility. Detailed analysis of cell subpopulation and function markers revealed that the mature enteroids differentiate from late embryonic intestinal villi to recapitulate many digestive, immune and gut-barrier functions present in the avian intestine. These transcriptomic results demonstrate that the chicken enteroid cultures are highly reproducible, and within the first week of culture they morphologically mature to appear similar to the in vivo intestine, therefore representing a physiologically-relevant in vitro model of the chicken intestine.
Collapse
Affiliation(s)
- Tessa J. Nash
- grid.4305.20000 0004 1936 7988Division of Immunology, The Roslin Institute, R(D)SVS, University of Edinburgh, Midlothian Edinburgh, UK
| | - Katrina M. Morris
- grid.4305.20000 0004 1936 7988Division of Immunology, The Roslin Institute, R(D)SVS, University of Edinburgh, Midlothian Edinburgh, UK
| | - Neil A. Mabbott
- grid.4305.20000 0004 1936 7988Division of Immunology, The Roslin Institute, R(D)SVS, University of Edinburgh, Midlothian Edinburgh, UK
| | - Lonneke Vervelde
- Division of Immunology, The Roslin Institute, R(D)SVS, University of Edinburgh, Midlothian, Edinburgh, UK.
| |
Collapse
|
34
|
Stewart AS, Schaaf CR, Veerasammy B, Freund JM, Gonzalez LM. Culture of equine intestinal epithelial stem cells after delayed tissue storage for future applications. BMC Vet Res 2022; 18:445. [PMID: 36564773 PMCID: PMC9783463 DOI: 10.1186/s12917-022-03552-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Equine intestinal epithelial stem cells (ISCs) serve as potential targets to treat horses with severe intestinal injury. The ability to isolate and store ISCs from intestinal biopsies creates an opportunity for both in vitro experiments to study ISC dynamics in a variety of intestinal diseases, and, in the future, utilize these cells as a possible therapy. If biopsies could be successfully stored prior to processing for ISCs, this would increase the availability of sample repositories for future experimental and therapeutic use. However, delayed culture of equine ISCs following prolonged sample storage has not been described. The objective of this study was to describe the isolation and culture of equine ISCs following delayed tissue storage. Small intestinal full thickness biopsies were collected post euthanasia. Fresh tissue was immediately processed or stored at 4 °C for 24, 48 and 72 h (H) before processing. Intestinal stem cells (crypts) were dissociated and cultured. Size, growth efficiency and proliferation potential were compared between resultant enteroids ("mini-guts") derived from each storage timepoint. In a separate study, growth efficiency of cryopreserved crypts was compared to cryopreserved enteroid fragments to investigate prolonged storage techniques. RESULTS Intestinal crypts were successfully isolated and cultured from all timepoints. At 72H post initial collection, the intestine was friable with epithelial sloughing; resultant dissociation yielded more partial crypts. Enteroids grown from crypts isolated at 72H were smaller with less proliferative potential (bud units, (median 6.5, 3.75-14.25)) than control (median 25, 15-28, p < 0.0001). No statistical differences were noted from tissues stored for 24H compared to control. Following cryopreservation, growth efficiency improved when cells were stored as enteroid fragments (median 81.6%, 66.2-109) compared to crypts (median 21.2%, 20-21.5, p = 0.01). The main limitations included a small sample size and lack of additional functional assays on enteroids. CONCLUSIONS Equine ISCs can be isolated and cultured after prolonged tissue storage. Resultant enteroids had minimal differences even after 24-48H of whole tissue storage. This suggests that ISCs could be isolated for several days from samples properly stored after procedures, including surgery or necropsy, and used to create ISC repositories for study or therapy of equine intestinal diseases.
Collapse
Affiliation(s)
- Amy Stieler Stewart
- grid.40803.3f0000 0001 2173 6074Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607 USA
| | - Cecilia R. Schaaf
- grid.40803.3f0000 0001 2173 6074Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607 USA
| | - Brittany Veerasammy
- grid.40803.3f0000 0001 2173 6074Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607 USA
| | - John M. Freund
- grid.40803.3f0000 0001 2173 6074Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607 USA
| | - Liara M. Gonzalez
- grid.40803.3f0000 0001 2173 6074Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607 USA
| |
Collapse
|
35
|
Park KW, Yang H, Lee MG, Ock SA, Wi H, Lee P, Hwang IS, Yoo JG, Park CK, Lee BR. Establishment of intestinal organoids from small intestine of growing cattle (12 months old). JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:1105-1116. [PMID: 36812001 PMCID: PMC9890329 DOI: 10.5187/jast.2022.e70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022]
Abstract
Recently, we reported the robust in vitro three-dimensional (3D) expansion of intestinal organoids derived from adult bovine (> 24 months) samples. The present study aimed to establish an in vitro 3D system for the cultivation of intestinal organoids derived from growing cattle (12 months old) for practical use as a potential alternative to in vivo systems for various purposes. However, very few studies on the functional characterization and 3D expansion of adult stem cells from livestock species compared to those from other species are available. In this study, intestinal crypts, including intestinal stem cells, from the small intestines (ileum and jejunum) of growing cattle were isolated and long-term 3D cultures were successfully established using a scaffold-based method. Furthermore, we generated an apical-out intestinal organoid derived from growing cattle. Interestingly, intestinal organoids derived from the ileum, but not the jejunum, could be expanded without losing the ability to recapitulate crypts, and these organoids specifically expressed several specific markers of intestinal stem cells and the intestinal epithelium. Furthermore, these organoids exhibited key functionality with regard to high permeability for compounds up to 4 kDa in size (e.g., fluorescein isothiocyanate [FITC]-dextran), indicating that apical-out intestinal organoids are better than other models. Collectively, these results indicate the establishment of growing cattle-derived intestinal organoids and subsequent generation of apical-out intestinal organoids. These organoids may be valuable tools and potential alternatives to in vivo systems for examining host-pathogen interactions involving epithelial cells, such as enteric virus infection and nutrient absorption, and may be used for various purposes.
Collapse
Affiliation(s)
- Kang Won Park
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea
| | - Hyeon Yang
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea
| | - Min Gook Lee
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea
| | - Sun A Ock
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea
| | - Hayeon Wi
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea
| | - Poongyeon Lee
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea
| | - In-Sul Hwang
- Columbia Center for Translational
Immunology, Columbia University Irving Medical Center, Columbia
University, New York, NY 10032, USA
| | - Jae Gyu Yoo
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea
| | - Choon-Keun Park
- College of Animal Life Sciences, Kangwon
National University, Chuncheon 24341, Korea
| | - Bo Ram Lee
- Animal Biotechnology Division, National
Institute of Animal Science, Rural Development Administration,
Wanju 55365, Korea,Corresponding author: Bo Ram Lee,
Animal Biotechnology Division, National Institute of Animal Science, Rural
Development Administration, Wanju 55365, Korea. Tel: +82-63-238-7259, E-mail:
| |
Collapse
|
36
|
Mussard E, Lencina C, Gallo L, Barilly C, Poli M, Feve K, Albin M, Cauquil L, Knudsen C, Achard C, Devailly G, Soler L, Combes S, Beaumont M. The phenotype of the gut region is more stably retained than developmental stage in piglet intestinal organoids. Front Cell Dev Biol 2022; 10:983031. [PMID: 36105361 PMCID: PMC9465596 DOI: 10.3389/fcell.2022.983031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal organoids are innovative in vitro tools to study the digestive epithelium. The objective of this study was to generate jejunum and colon organoids from suckling and weaned piglets in order to determine the extent to which organoids retain a location-specific and a developmental stage-specific phenotype. Organoids were studied at three time points by gene expression profiling for comparison with the transcriptomic patterns observed in crypts in vivo. In addition, the gut microbiota and the metabolome were analyzed to characterize the luminal environment of epithelial cells at the origin of organoids. The location-specific expression of 60 genes differentially expressed between jejunum and colon crypts from suckling piglets was partially retained (48%) in the derived organoids at all time point. The regional expression of these genes was independent of luminal signals since the major differences in microbiota and metabolome observed in vivo between the jejunum and the colon were not reproduced in vitro. In contrast, the regional expression of other genes was erased in organoids. Moreover, the developmental stage-specific expression of 30 genes differentially expressed between the jejunum crypts of suckling and weaned piglets was not stably retained in the derived organoids. Differentiation of organoids was necessary to observe the regional expression of certain genes while it was not sufficient to reproduce developmental stage-specific expression patterns. In conclusion, piglet intestinal organoids retained a location-specific phenotype while the characteristics of developmental stage were erased in vitro. Reproducing more closely the luminal environment might help to increase the physiological relevance of intestinal organoids.
Collapse
Affiliation(s)
- Eloïse Mussard
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
- Lallemand Animal Nutrition, Blagnac Cedex, France
| | - Corinne Lencina
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Lise Gallo
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Céline Barilly
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Maryse Poli
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Katia Feve
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Mikael Albin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Laurent Cauquil
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | | | | | | | - Laura Soler
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Sylvie Combes
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Martin Beaumont
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
- *Correspondence: Martin Beaumont,
| |
Collapse
|
37
|
Nash T, Vervelde L. Advances, challenges and future applications of avian intestinal in vitro models. Avian Pathol 2022; 51:317-329. [PMID: 35638458 DOI: 10.1080/03079457.2022.2084363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
There is a rapidly growing interest in how the avian intestine is affected by dietary components and probiotic microorganisms, as well as its role in the spread of infectious diseases in both the developing and developed world. A paucity of physiologically relevant models has limited research in this essential field of poultry gut health and led to an over-reliance on the use of live birds for experiments. The intestine is characterized by a complex cellular composition with numerous functions, unique dynamic locations and interdependencies making this organ challenging to recreate in vitro. This review illustrates the in vitro tools that aim to recapitulate this intestinal environment; from the simplest cell lines, which mimic select features of the intestine but lack anatomical and physiological complexity, to the more recently developed complex 3D enteroids, which recreate more of the intestine's intricate microanatomy, heterogeneous cell populations and signalling gradients. We highlight the benefits and limitations of in vitro intestinal models and describe their current applications and future prospective utilizations in intestinal biology and pathology research. We also describe the scope to improve on the current systems to include, for example, microbiota and a dynamic mechanical environment, vital components which enable the intestine to develop and maintain homeostasis in vivo. As this review explains, no one model is perfect, but the key to choosing a model or combination of models is to carefully consider the purpose or scientific question.
Collapse
Affiliation(s)
- Tessa Nash
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, UK
| | - Lonneke Vervelde
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
38
|
Faber MN, Smith D, Price DRG, Steele P, Hildersley KA, Morrison LJ, Mabbott NA, Nisbet AJ, McNeilly TN. Development of Bovine Gastric Organoids as a Novel In Vitro Model to Study Host-Parasite Interactions in Gastrointestinal Nematode Infections. Front Cell Infect Microbiol 2022; 12:904606. [PMID: 35846775 PMCID: PMC9281477 DOI: 10.3389/fcimb.2022.904606] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/24/2022] [Indexed: 12/17/2022] Open
Abstract
Gastro-intestinal nematode (GIN) parasites are a major cause of production losses in grazing cattle, primarily through reduced growth rates in young animals. Control of these parasites relies heavily on anthelmintic drugs; however, with growing reports of resistance to currently available anthelmintics, alternative methods of control are required. A major hurdle in this work has been the lack of physiologically relevant in vitro infection models that has made studying precise interactions between the host and the GINs difficult. Such mechanistic insights into the infection process will be valuable for the development of novel targets for drugs, vaccines, or other interventions. Here we created bovine gastric epithelial organoids from abomasal gastric tissue and studied their application as in vitro models for understanding host invasion by GIN parasites. Transcriptomic analysis of gastric organoids across multiple passages and the corresponding abomasal tissue showed conserved expression of tissue-specific genes across samples, demonstrating that the organoids are representative of bovine gastric tissue from which they were derived. We also show that self-renewing and self-organising three-dimensional organoids can also be serially passaged, cryopreserved, and resuscitated. Using Ostertagia ostertagi, the most pathogenic gastric parasite in cattle in temperate regions, we show that cattle gastric organoids are biologically relevant models for studying GIN invasion in the bovine abomasum. Within 24 h of exposure, exsheathed larvae rapidly and repeatedly infiltrated the lumen of the organoids. Prior to invasion by the parasites, the abomasal organoids rapidly expanded, developing a ‘ballooning’ phenotype. Ballooning of the organoids could also be induced in response to exposure to parasite excretory/secretory products. In summary, we demonstrate the power of using abomasal organoids as a physiologically relevant in vitro model system to study interactions of O. ostertagi and other GIN with bovine gastrointestinal epithelium.
Collapse
Affiliation(s)
- Marc N. Faber
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
- *Correspondence: Marc N. Faber,
| | - David Smith
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
| | - Daniel R. G. Price
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
| | - Philip Steele
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
| | - Katie A. Hildersley
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
| | - Liam J. Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Penicuik, United Kingdom
| | - Neil A. Mabbott
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Penicuik, United Kingdom
| | - Alasdair J. Nisbet
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
| | - Tom N. McNeilly
- Moredun Research Institute, Pentlands Science Park, Penicuik, United Kingdom
| |
Collapse
|
39
|
Description of a Newly Isolated Blautia faecis Strain and Its Benefit in Mouse Models of Post-Influenza Secondary Enteric and Pulmonary Infections. Nutrients 2022; 14:nu14071478. [PMID: 35406091 PMCID: PMC9003314 DOI: 10.3390/nu14071478] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/28/2022] Open
Abstract
The expanding knowledge on the systemic influence of the human microbiome suggests that fecal samples are underexploited sources of new beneficial strains for extra-intestinal health. We have recently shown that acetate, a main circulating microbiota-derived molecule, reduces the deleterious effects of pulmonary Streptococcus pneumoniae and enteric Salmonella enterica serovar Typhimurium bacterial post-influenza superinfections. Considering the beneficial and broad effects of acetate, we intended to isolate a commensal strain, producing acetate and potentially exploitable in the context of respiratory infections. We designed successive steps to select intestinal commensals that are extremely oxygen-sensitive, cultivable after a freezing process, without a proinflammatory effect on IL-8 induction, and producing acetate. We have identified the Blautia faecis DSM33383 strain, which decreased the TNFα-induced production of IL-8 by the intestinal epithelial cell line HT-29. The beneficial effect of this bacterial strain was further studied in two preclinical models of post-influenza Streptococcus pneumoniae (S.p) and Salmonella enterica serovar Typhimurium (S.t) superinfection. The intragastrical administration of Blautia faecis DSM33383 led to protection in influenza-infected mice suffering from an S.p. and, to a lesser extent, from an S.t secondary infection. Altogether, this study showed that Blautia faecis DSM33383 could be a promising candidate for preventive management of respiratory infectious diseases.
Collapse
|
40
|
Smith D, Price DRG, Faber MN, Chapuis AF, McNeilly TN. Advancing animal health and disease research in the lab with three-dimensional cell culture systems. Vet Rec 2022; 191:e1528. [PMID: 35338777 DOI: 10.1002/vetr.1528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/31/2022] [Accepted: 02/11/2022] [Indexed: 11/08/2022]
Abstract
The development of three-dimensional cell culture systems representative of tissues from animals of veterinary interest is accelerating research that seeks to address specific questions tied to animal health. In terms of their relevance and complexity, these in vitro models can be seen as a midpoint between the more reductionist single-cell culture systems and complex live animals. Organoids in particular represent a significant development due to their organised multicellular structure that more closely represents in vivo tissues than any other cell culture technology previously developed. In this review, we provide an overview of the different three-dimensional cell culture systems available to veterinary researchers and give examples of their application in contexts relating to animal health.
Collapse
Affiliation(s)
- David Smith
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, UK
| | - Daniel R G Price
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, UK
| | - Marc N Faber
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, UK
| | - Ambre F Chapuis
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, UK
| | - Tom N McNeilly
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, UK
| |
Collapse
|
41
|
Marks H, Grześkowiak Ł, Martinez-Vallespin B, Dietz H, Zentek J. Porcine and Chicken Intestinal Epithelial Cell Models for Screening Phytogenic Feed Additives—Chances and Limitations in Use as Alternatives to Feeding Trials. Microorganisms 2022; 10:microorganisms10030629. [PMID: 35336204 PMCID: PMC8951747 DOI: 10.3390/microorganisms10030629] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 02/07/2023] Open
Abstract
Numerous bioactive plant additives have shown various positive effects in pigs and chickens. The demand for feed additives of natural origin has increased rapidly in recent years to support the health of farm animals and thus minimize the need for antibiotics and other drugs. Although only in vivo experiments can fully represent their effect on the organism, the establishment of reliable in vitro methods is becoming increasingly important in the goal of reducing the use of animals in experiments. The use of cell models requires strict control of the experimental conditions so that reliability and reproducibility can be achieved. In particular, the intestinal porcine epithelial cell line IPEC-J2 represents a promising model for the development of new additives. It offers the possibility to investigate antioxidative, antimicrobial, anti- or pro-proliferative and antiviral effects. However, the use of IPEC-J2 is limited due to its purely epithelial origin and some differences in its morphology and functionality compared to the in vivo situation. With regard to chickens, the development of a reliable intestinal epithelial cell model has attracted the attention of researchers in recent years. Although a promising model was presented lately, further studies are needed to enable the standardized use of a chicken cell line for testing phytogenic feed additives. Finally, co-cultivation of the currently available cell lines with other cell lines and the development of organoids will open up further application possibilities. Special emphasis was given to the IPEC-J2 cell model. Therefore, all publications that investigated plant derived compounds in this cell line were considered. The section on chicken cell lines is based on publications describing the development of chicken intestinal epithelial cell models.
Collapse
Affiliation(s)
- Hannah Marks
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
- Kaesler Research Institute, Kaesler Nutrition GmbH, Fischkai 1, 27572 Bremerhaven, Germany;
- Correspondence:
| | - Łukasz Grześkowiak
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
| | - Beatriz Martinez-Vallespin
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
| | - Heiko Dietz
- Kaesler Research Institute, Kaesler Nutrition GmbH, Fischkai 1, 27572 Bremerhaven, Germany;
| | - Jürgen Zentek
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany; (Ł.G.); (B.M.-V.); (J.Z.)
| |
Collapse
|
42
|
Paradowska M, Dunislawska A, Siwek M, Slawinska A. Avian Cell Culture Models to Study Immunomodulatory Properties of Bioactive Products. Animals (Basel) 2022; 12:670. [PMID: 35268238 PMCID: PMC8909239 DOI: 10.3390/ani12050670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/17/2022] [Accepted: 02/28/2022] [Indexed: 11/21/2022] Open
Abstract
Antimicrobial resistance is becoming a greater danger to both human and animal health, reducing the capacity to treat bacterial infections and increasing the risk of morbidity and mortality from resistant bacteria. Antimicrobial efficacy in the treatment of bacterial infections is still a major concern in both veterinary and human medicine. Antimicrobials can be replaced with bioactive products. Only a small number of plant species have been studied in respect to their bioactive compounds. More research is needed to characterize and evaluate the therapeutic properties of the plant extracts. Due to the more and more common phenomenon of antimicrobial resistance, poultry farming requires the use of natural alternatives to veterinary antibiotics that have an immunomodulatory effect. These include a variety of bioactive products, such as plant extracts, essential oils, probiotics, prebiotics, and synbiotics. This article presents several studies on bioactive products and their immunomodulatory effects tested in vitro and ex vivo using various avian cell culture models. Primary cell cultures that have been established to study the immune response in chickens include peripheral blood mononuclear cells (PBMCs), intestinal epithelial cells (IEC), and bone marrow-derived dendritic cells (BMDCs). Chicken lymphatic lines that can be used to study immune responses are mainly: chicken B cells infected with avian leukemia RAV-1 virus (DT40), macrophage-like cell line (HD11), and a spleen-derived macrophage cell line (MQ-NCSU). Ex vivo organ cultures combine in vitro and in vivo studies, as this model is based on fragments of organs or tissues grown in vitro. As such, it mimics the natural reactions of organisms, but under controlled conditions. Most ex vivo organ cultures of chickens are derived from the ileum and are used to model the interaction between the gastrointestinal tract and the microbiota. In conclusion, the use of in vitro and ex vivo models allows for numerous experimental replications in a short period, with little or no ethical constraints and limited confounding factors.
Collapse
Affiliation(s)
- Michelle Paradowska
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland; (A.D.); (M.S.)
| | - Aleksandra Dunislawska
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland; (A.D.); (M.S.)
| | - Maria Siwek
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland; (A.D.); (M.S.)
| | - Anna Slawinska
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland;
| |
Collapse
|
43
|
Sutton KM, Orr B, Hope J, Jensen SR, Vervelde L. Establishment of bovine 3D enteroid-derived 2D monolayers. Vet Res 2022; 53:15. [PMID: 35236416 PMCID: PMC8889782 DOI: 10.1186/s13567-022-01033-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/03/2022] [Indexed: 12/28/2022] Open
Abstract
Three-dimensional (3D) intestinal enteroids are powerful in vitro models for studying intestinal biology. However, due to their closed structure direct access to the apical surface is impeded, limiting high-throughput applications of exogenous compounds and pathogens. In this study, we describe a method for generating confluent 2D enteroids from single-cell suspensions of enzymatically-dissociated ileum-derived bovine 3D enteroids. Confluent monolayers were first achieved using IntestiCult media but to establish a defined, cost-effective culture media, we also developed a bovine enteroid monolayer (BEM) medium. The monolayers cultured in BEM media proliferated extensively and formed confluent cell layers on both Matrigel-coated plastic plates and transwell inserts by day 3 of culture. The 2D enteroids maintained the epithelial cell lineages found in 3D enteroids and ileum tissue. In addition, the monolayers formed a functional epithelial barrier based on the presence of the adherens and tight junction proteins, E-cadherin and ZO-1, and electrical resistance across the monolayer was measured from day 3 and maintained for up to 7 days in culture. The method described here will provide a useful model to study bovine epithelial cell biology with ease of access to the apical surface of epithelial cells and has potential to investigate host-pathogen interactions and screen bioactive compounds.
Collapse
Affiliation(s)
- Kate M Sutton
- Division of Infection and Immunity, The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| | - Brigid Orr
- Division of Infection and Immunity, The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Jayne Hope
- Division of Infection and Immunity, The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Stina R Jensen
- Novozymes A/S, Animal Health and Nutrition, 2800, Lyngby, Denmark
| | - Lonneke Vervelde
- Division of Infection and Immunity, The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| |
Collapse
|
44
|
Joo SS, Gu BH, Park YJ, Rim CY, Kim MJ, Kim SH, Cho JH, Kim HB, Kim M. Porcine Intestinal Apical-Out Organoid Model for Gut Function Study. Animals (Basel) 2022; 12:ani12030372. [PMID: 35158695 PMCID: PMC8833427 DOI: 10.3390/ani12030372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Pigs have been used in various animal model studies on the gastrointestinal tract (GIT) across both animal science and biomedical science fields. Recently, intestinal organoids have been used as a research tool for the GIT, and they have also been applied to farm animals, including pigs. However, to our knowledge, no functional studies of the porcine intestine using intestinal organoids have been conducted to date. In the present study, we developed two porcine intestinal organoid models (basal-out and apical-out organoids) and compared their characteristics. We also confirmed the possibility of conducting research related to intestinal functions, such as nutrient uptake and gut barrier function. The present study suggests that porcine intestinal organoids can be used as potential models for future GIT mechanism studies, such as host–microbe interactions, harmful ingredient tests, and nutritional research. Abstract Pig models provide valuable research information on farm animals, veterinary, and biomedical sciences. Experimental pig gut models are used in studies on physiology, nutrition, and diseases. Intestinal organoids are powerful tools for investigating intestinal functions such as nutrient uptake and gut barrier function. However, organoids have a basal-out structure and need to grow in the extracellular matrix, which causes difficulties in research on the intestinal apical membrane. We established porcine intestinal organoids from jejunum tissues and developed basal-out and apical-out organoids using different sub-culture methods. Staining and quantitative real-time PCR showed the difference in axis change of the membrane and gene expression of epithelial cell marker genes. To consider the possibility of using apical-out organoids for intestinal function, studies involving fatty acid uptake and disruption of the epithelial barrier were undertaken. Fluorescence fatty acid was more readily absorbed in apical-out organoids than in basal-out organoids within the same time. To determine whether apical-out organoids form a functional barrier, a fluorescent dextran diffusion assay was performed. Hence, we successfully developed porcine intestinal organoid culture systems and showed that the porcine apical-out organoid model is ideal for the investigation of the intestinal environment. It can be used in future studies related to the intestine across various research fields.
Collapse
Affiliation(s)
- Sang-Seok Joo
- Department of Animal Science, College of Natural Resources & Live Science, Pusan National University, Miryang 50463, Korea; (S.-S.J.); (Y.-J.P.); (C.-Y.R.)
| | - Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Mirayng 50463, Korea;
| | - Yei-Ju Park
- Department of Animal Science, College of Natural Resources & Live Science, Pusan National University, Miryang 50463, Korea; (S.-S.J.); (Y.-J.P.); (C.-Y.R.)
| | - Chae-Yun Rim
- Department of Animal Science, College of Natural Resources & Live Science, Pusan National University, Miryang 50463, Korea; (S.-S.J.); (Y.-J.P.); (C.-Y.R.)
| | - Min-Ji Kim
- Animal Nutrition and Physiology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea; (M.-J.K.); (S.-H.K.)
| | - Sang-Ho Kim
- Animal Nutrition and Physiology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea; (M.-J.K.); (S.-H.K.)
| | - Jin-Ho Cho
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Korea;
| | - Hyeun-Bum Kim
- Department of Animal Resources and Science, Dankook University, Cheonan 31116, Korea;
| | - Myunghoo Kim
- Department of Animal Science, College of Natural Resources & Live Science, Pusan National University, Miryang 50463, Korea; (S.-S.J.); (Y.-J.P.); (C.-Y.R.)
- Life and Industry Convergence Research Institute, Pusan National University, Mirayng 50463, Korea;
- Correspondence: ; Tel.: +82-55-350-5516; Fax: +82-55-350-5519
| |
Collapse
|
45
|
Intestinal Organoids: New Tools to Comprehend the Virulence of Bacterial Foodborne Pathogens. Foods 2022; 11:foods11010108. [PMID: 35010234 PMCID: PMC8750402 DOI: 10.3390/foods11010108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
Foodborne diseases cause high morbidity and mortality worldwide. Understanding the relationships between bacteria and epithelial cells throughout the infection process is essential to setting up preventive and therapeutic solutions. The extensive study of their pathophysiology has mostly been performed on transformed cell cultures that do not fully mirror the complex cell populations, the in vivo architectures, and the genetic profiles of native tissues. Following advances in primary cell culture techniques, organoids have been developed. Such technological breakthroughs have opened a new path in the study of microbial infectious diseases, and thus opened onto new strategies to control foodborne hazards. This review sheds new light on cellular messages from the host–foodborne pathogen crosstalk during in vitro organoid infection by the foodborne pathogenic bacteria with the highest health burden. Finally, future perspectives and current challenges are discussed to provide a better understanding of the potential applications of organoids in the investigation of foodborne infectious diseases.
Collapse
|
46
|
Beaumont M, Lencina C, Painteaux L, Viémon-Desplanque J, Phornlaphat O, Lambert W, Chalvon-Demersay T. A mix of functional amino acids and grape polyphenols promotes the growth of piglets, modulates the gut microbiota in vivo and regulates epithelial homeostasis in intestinal organoids. Amino Acids 2021; 54:1357-1369. [PMID: 34642825 DOI: 10.1007/s00726-021-03082-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022]
Abstract
Weaning is a challenging period for gut health in piglets. Previous studies showed that dietary supplementations with either amino acids or polyphenols promote piglet growth and intestinal functions, when administered separately. Thus, we hypothesized that a combination of amino acids and polyphenols could facilitate the weaning transition. Piglets received during the first two weeks after weaning a diet supplemented or not with a mix of a low dose (0.1%) of functional amino acids (L-arginine, L-leucine, L-valine, L-isoleucine, L-cystine) and 100 ppm of a polyphenol-rich extract from grape seeds and skins. The mix of amino acids and polyphenols improved growth and feed efficiency. These beneficial effects were associated with a lower microbiota diversity and a bloom of Lactobacillaceae in the jejunum content while the abundance of Proteobacteria was reduced in the caecum content. The mix of amino acids and polyphenols also increased the production by the caecum microbiota of short-chain fatty acids (butyrate, propionate) and of metabolites derived from amino acids (branched-chain fatty acids, valerate, putrescine) and from polyphenols (3-phenylpropionate). Experiments in piglet jejunum organoids revealed that the mix of amino acids and polyphenols upregulated the gene expression of epithelial differentiation markers while it reduced the gene expression of proliferation and innate immunity markers. In conclusion, the supplementation of a mix of amino acids and polyphenols is a promising nutritional strategy to manage gut health in piglets through the modulation of the gut microbiota and of the epithelial barrier.
Collapse
Affiliation(s)
- Martin Beaumont
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet-Tolosan, France.
| | - Corinne Lencina
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet-Tolosan, France
| | - Louise Painteaux
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet-Tolosan, France
| | | | - Orasin Phornlaphat
- BARC, Bangkok Animal Research Center Co., Ltd, 74/4 Mu 7 Tambon Naiklong Bangplakod, Phrasamutjedi,, Samut Prakan, 10290, Thailand
| | | | | |
Collapse
|
47
|
Smith D, Price DRG, Burrells A, Faber MN, Hildersley KA, Chintoan-Uta C, Chapuis AF, Stevens M, Stevenson K, Burgess STG, Innes EA, Nisbet AJ, McNeilly TN. The Development of Ovine Gastric and Intestinal Organoids for Studying Ruminant Host-Pathogen Interactions. Front Cell Infect Microbiol 2021; 11:733811. [PMID: 34568096 PMCID: PMC8456012 DOI: 10.3389/fcimb.2021.733811] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/13/2021] [Indexed: 01/10/2023] Open
Abstract
Gastrointestinal (GI) infections in sheep have significant implications for animal health, welfare and productivity, as well as being a source of zoonotic pathogens. Interactions between pathogens and epithelial cells at the mucosal surface play a key role in determining the outcome of GI infections; however, the inaccessibility of the GI tract in vivo significantly limits the ability to study such interactions in detail. We therefore developed ovine epithelial organoids representing physiologically important gastric and intestinal sites of infection, specifically the abomasum (analogous to the stomach in monogastrics) and ileum. We show that both abomasal and ileal organoids form self-organising three-dimensional structures with a single epithelial layer and a central lumen that are stable in culture over serial passage. We performed RNA-seq analysis on abomasal and ileal tissue from multiple animals and on organoids across multiple passages and show the transcript profile of both abomasal and ileal organoids cultured under identical conditions are reflective of the tissue from which they were derived and that the transcript profile in organoids is stable over at least five serial passages. In addition, we demonstrate that the organoids can be successfully cryopreserved and resuscitated, allowing long-term storage of organoid lines, thereby reducing the number of animals required as a source of tissue. We also report the first published observations of a helminth infecting gastric and intestinal organoids by challenge with the sheep parasitic nematode Teladorsagia circumcincta, demonstrating the utility of these organoids for pathogen co-culture experiments. Finally, the polarity in the abomasal and ileal organoids can be inverted to make the apical surface directly accessible to pathogens or their products, here shown by infection of apical-out organoids with the zoonotic enteric bacterial pathogen Salmonella enterica serovar Typhimurium. In summary, we report a simple and reliable in vitro culture system for generation and maintenance of small ruminant intestinal and gastric organoids. In line with 3Rs principals, use of such organoids will reduce and replace animals in host-pathogen research.
Collapse
Affiliation(s)
- David Smith
- Department of Disease Control, Moredun Research Institute, Midlothian, United Kingdom
| | - Daniel R G Price
- Department of Vaccines and Diagnostics, Moredun Research Institute, Midlothian, United Kingdom
| | - Alison Burrells
- Department of Disease Control, Moredun Research Institute, Midlothian, United Kingdom
| | - Marc N Faber
- Department of Disease Control, Moredun Research Institute, Midlothian, United Kingdom
| | - Katie A Hildersley
- Department of Disease Control, Moredun Research Institute, Midlothian, United Kingdom.,Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | | | - Ambre F Chapuis
- Department of Vaccines and Diagnostics, Moredun Research Institute, Midlothian, United Kingdom.,The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Mark Stevens
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Karen Stevenson
- Department of Vaccines and Diagnostics, Moredun Research Institute, Midlothian, United Kingdom
| | - Stewart T G Burgess
- Department of Vaccines and Diagnostics, Moredun Research Institute, Midlothian, United Kingdom
| | - Elisabeth A Innes
- Department of Disease Control, Moredun Research Institute, Midlothian, United Kingdom
| | - Alasdair J Nisbet
- Department of Vaccines and Diagnostics, Moredun Research Institute, Midlothian, United Kingdom
| | - Tom N McNeilly
- Department of Disease Control, Moredun Research Institute, Midlothian, United Kingdom
| |
Collapse
|
48
|
Sang Y, Miller LC, Nelli RK, Giménez-Lirola LG. Harness Organoid Models for Virological Studies in Animals: A Cross-Species Perspective. Front Microbiol 2021; 12:725074. [PMID: 34603253 PMCID: PMC8481363 DOI: 10.3389/fmicb.2021.725074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/27/2021] [Indexed: 02/02/2023] Open
Abstract
Animal models and cell culture in vitro are primarily used in virus and antiviral immune research. Whereas the limitation of these models to recapitulate the viral pathogenesis in humans has been made well aware, it is imperative to introduce more efficient systems to validate emerging viruses in both domestic and wild animals. Organoids ascribe to representative miniatures of organs (i.e., mini-organs), which are derived from three-dimensional culture of stem cells under respective differential conditions mimicking endogenous organogenetic niches. Organoids have broadened virological studies in the human context, particularly in recent uses for COVID19 research. This review examines the status and potential for cross-species applied organotypic culture in validating emerging animal, particularly zoonotic, viruses in domestic and wild animals.
Collapse
Affiliation(s)
- Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN, United States
| | - Laura C Miller
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA, United States
| | - Rahul K Nelli
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Luis Gabriel Giménez-Lirola
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
49
|
Establishment of a Newborn Lamb Gut-Loop Model to Evaluate New Methods of Enteric Disease Control and Reduce Experimental Animal Use. Vet Sci 2021; 8:vetsci8090170. [PMID: 34564564 PMCID: PMC8472880 DOI: 10.3390/vetsci8090170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Enteric infectious diseases are not all well controlled, which leads to animal suffering and sometimes death in the most severe cases, in addition to economic losses for farmers. Typical symptoms of enteric infections include watery diarrhea, stomach cramps or pain, dehydration, nausea, vomiting, fever and weight loss. Evaluation of new control methods against enteric infections requires the use of many animals. We aimed to develop a new method for an initial in vivo screen of promising compounds against neonatal diseases such as cryptosporidiosis while limiting experimental animal use. We therefore adapted an in vivo method of multiple consecutive but independent intestinal loops to newborn lambs delivered by cesarean section, in which endotoxin responsiveness is retained. This new method allowed for the screening of natural yeast fractions for their ability to stimulate immune responses and to limit early Cryptosporidium parvum development. This model may also be used to investigate host–pathogen interactions and immune responses in a neonatal controlled environment.
Collapse
|
50
|
Hoffmann P, Schnepel N, Langeheine M, Künnemann K, Grassl GA, Brehm R, Seeger B, Mazzuoli-Weber G, Breves G. Intestinal organoid-based 2D monolayers mimic physiological and pathophysiological properties of the pig intestine. PLoS One 2021; 16:e0256143. [PMID: 34424915 PMCID: PMC8382199 DOI: 10.1371/journal.pone.0256143] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
Gastrointestinal infectious diseases remain an important issue for human and animal health. Investigations on gastrointestinal infectious diseases are classically performed in laboratory animals leading to the problem that species-specific models are scarcely available, especially when it comes to farm animals. The 3R principles of Russel and Burch were achieved using intestinal organoids of porcine jejunum. These organoids seem to be a promising tool to generate species-specific in vitro models of intestinal epithelium. 3D Organoids were grown in an extracellular matrix and characterized by qPCR. Organoids were also seeded on permeable filter supports in order to generate 2D epithelial monolayers. The organoid-based 2D monolayers were characterized morphologically and were investigated regarding their potential to study physiological transport properties and pathophysiological processes. They showed a monolayer structure containing different cell types. Moreover, their functional activity was demonstrated by their increasing transepithelial electrical resistance over 18 days and by an active glucose transport and chloride secretion. Furthermore, the organoid-based 2D monolayers were also confronted with cholera toxin derived from Vibrio cholerae as a proof of concept. Incubation with cholera toxin led to an increase of short-circuit current indicating an enhanced epithelial chloride secretion, which is a typical characteristic of cholera infections. Taken this together, our model allows the investigation of physiological and pathophysiological mechanisms focusing on the small intestine of pigs. This is in line with the 3R principle and allows the reduction of classical animal experiments.
Collapse
Affiliation(s)
- Pascal Hoffmann
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
- * E-mail:
| | - Nadine Schnepel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marion Langeheine
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Katrin Künnemann
- Institute of Medical Microbiology and Hospital Epidemiology and German Center for Infection Research (DZIF), Partner Site Hannover, Hannover Medical School, Hannover, Germany
| | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology and German Center for Infection Research (DZIF), Partner Site Hannover, Hannover Medical School, Hannover, Germany
| | - Ralph Brehm
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Bettina Seeger
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gemma Mazzuoli-Weber
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gerhard Breves
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|