1
|
Rossen ND, Touhara KK, Castro J, Harrington AM, Caraballo SG, Deng F, Li Y, Brierley SM, Julius D. Population imaging of enterochromaffin cell activity reveals regulation by somatostatin. Proc Natl Acad Sci U S A 2025; 122:e2501525122. [PMID: 40327690 DOI: 10.1073/pnas.2501525122] [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: 01/21/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Sensory enteroendocrine cells in the intestinal epithelium detect and relay information about the luminal environment to other cells within and outside the gut. Serotonergic enterochromaffin (EC) cells are a subset of enteroendocrine cells that detect noxious stimuli within the gut lumen, such as chemical irritants and microbial byproducts, and transduce this information to sensory nerve fibers to elicit defensive responses such as nausea and visceral pain. While much has recently been learned about the pharmacological and biophysical characteristics of EC cells, a more broadscale investigation of their properties has been hindered by their relatively low prevalence and sparse anatomical distribution within the gut epithelium. Here, we introduce a method for large-scale parallel analysis of individual EC cell activity within a physiologically relevant epithelial context. Using this approach, we identify somatostatin-28 as a potent inhibitor of both basal and stimulus-evoked serotonin release from EC cells and delineate the signaling pathway that underlies this modulatory response. Our analysis suggests that targeting this inhibitory signaling pathway may offer therapeutic avenues for treating gastrointestinal disorders associated with EC cell function and dysregulated serotonin signaling. Together with the ongoing development of specific biosensors, this platform provides a template for the efficient characterization of other rare sensory cell types and their pharmacological modulators.
Collapse
Affiliation(s)
- Nathan D Rossen
- Department of Physiology, University of California, San Francisco, CA 94158
- Tetrad Graduate Program, Graduate Division, University of California, San Francisco, CA 94158
| | - Kouki K Touhara
- Department of Physiology, University of California, San Francisco, CA 94158
| | - Joel Castro
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Andrea M Harrington
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Sonia Garcia Caraballo
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Fei Deng
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, Beijing 100871, China
- International Data Group / McGovern Institute for Brain Research at Peking University, Beijing 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, Beijing 100871, China
- International Data Group / McGovern Institute for Brain Research at Peking University, Beijing 100871, China
| | - Stuart M Brierley
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - David Julius
- Department of Physiology, University of California, San Francisco, CA 94158
| |
Collapse
|
2
|
Quacquarelli F, Davila S, Taelman J, Guiu J, Antfolk M. Enhanced small intestinal organoid-derived epithelial cell adhesion and growth in organ-on-a-chip devices. RSC Adv 2025; 15:3693-3703. [PMID: 39911548 PMCID: PMC11795259 DOI: 10.1039/d4ra08290g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
Organ-on-a-chip devices are predominately made of the polymer polymethylsiloxane (PDMS), exhibiting several attractive properties e.g., transparency, gas permeability, and biocompatibility. However, the attachment of cells to this polymer has proven challenging, especially for delicate primary cells e.g., small intestinal organoid-derived epithelial cells. Hence, a need to functionalize and coat the surface has arisen to render it more hydrophilic and improve its ability to support cell adhesion and growth. While previous research has demonstrated some successful results in culturing primary cells, no comprehensive and comparative protocol has been proposed. Here, we provide a protocol for enhanced small intestinal organoid-derived epithelial cell adhesion and growth on PDMS and plastics, assessing both PDMS surface functionalization, adhesion protein coating as well as medium selection. We assess PDMS functionalization using (3-aminopropyl)trimethoxysilane (APTMS) or polyethyleneimine-glutaraldehyde (PEIGA), and adhesion protein coating using various Laminins, Collagen I, Matrigel, or mixtures thereof. Finally, we assess the use of two different medium compositions including growth factors EGF, Noggin and R-spondin1 (ENR medium) alone or combined with the two small molecules CHIR99021 and valproic acid (CV medium). We envision that our results will be useful for further attempts in emulating the small intestine using plastic- or PDMS-based devices for organs-on-a-chip development.
Collapse
Affiliation(s)
| | - Sergio Davila
- Department of Biomedical Engineering, Lund University Lund Sweden
| | - Jasin Taelman
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL L'Hospitalet de Llobregat Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C] L'Hospitalet de Llobregat Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL L'Hospitalet de Llobregat Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C] L'Hospitalet de Llobregat Spain
| | - Maria Antfolk
- Department of Biomedical Engineering, Lund University Lund Sweden
| |
Collapse
|
3
|
Masete KV, Günzel D, Schulzke JD, Epple HJ, Hering NA. Matrix-free human 2D organoids recapitulate duodenal barrier and transport properties. BMC Biol 2025; 23:2. [PMID: 39757172 DOI: 10.1186/s12915-024-02105-7] [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: 08/05/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Traditionally, transformed cell line monolayers have been the standard model for studying epithelial barrier and transport function. Recently, intestinal organoids were proposed as superior in recapitulating the intestine. Typically, 3D organoids are digested and seeded as monolayers on gelatinous matrix pre-coated surfaces for anchorage. As this coat could potentially act as a diffusion barrier, we aimed to generate robust human duodenum-derived organoid monolayers that do not need a gelatinous matrix for anchorage to improve upon existing models to study epithelial transport and barrier function. RESULTS We characterized these monolayers phenotypically regarding polarization, tight junction formation and cellular composition, and functionally regarding uptake of nutrients, ion transport and cytokine-induced barrier dysfunction. The organoid monolayers recapitulated the duodenum phenotypically as well as functionally regarding glucose and short-chain fatty acid uptake. Tumour necrosis factor-alpha induced paracellular transport of 4-kDa Dextran and transcytosis of 44-kDa horseradish peroxidase. Notably, forskolin-stimulated chloride secretion was consistently lower when organoid monolayers were seeded on a layer of basement membrane extract (BME). CONCLUSIONS BME-free organoid monolayers represent an improved model for studying transcytotic, paracellular but especially transcellular transport. As BME is extracted from mice, our model furthers efforts to make organoid culture more animal-free.
Collapse
Affiliation(s)
- Kopano Valerie Masete
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Hans-Jörg Epple
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Antibiotic Stewardship Team, Medical Directorate, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Nina A Hering
- Department of General and Visceral Surgery, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, Berlin, 12203, Germany.
| |
Collapse
|
4
|
Larrañaga E, Marin-Riera M, Abad-Lázaro A, Bartolomé-Català D, Otero A, Fernández-Majada V, Batlle E, Sharpe J, Ojosnegros S, Comelles J, Martinez E. Long-range organization of intestinal 2D-crypts using exogenous Wnt3a micropatterning. Nat Commun 2025; 16:382. [PMID: 39753580 PMCID: PMC11698991 DOI: 10.1038/s41467-024-55651-7] [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: 11/10/2023] [Accepted: 12/19/2024] [Indexed: 01/06/2025] Open
Abstract
Intestinal epithelial cells are segregated into proliferative crypts and differentiated regions. This organization relies on specific signals, including Wnt3a, which regulates cell proliferation within crypts, and Eph/Ephrin, which dictates cell positioning along the crypt-villus axis. However, studying how the spatial distributions of these signals influences crypt-villus organization is challenging both in vitro and in vivo. Here we show that micropatterns of Wnt3a can govern the size, shape and long-range organization of crypts in vitro. By adjusting the spacing between Wnt3a ligand patterns at the microscale over large surfaces, we override endogenous Wnt3a to precisely control the distribution and long-range order of crypt-like regions in primary epithelial monolayers. Additionally, an agent-based model integrating Wnt3a/BMP feedback and Eph/Ephrin repulsion effectively replicates experimental tissue compartmentalization, crypt size, shape, and organization. This combined experimental and computational approach offers a framework to study how signaling pathways help organize intestinal epithelial tissue.
Collapse
Affiliation(s)
- Enara Larrañaga
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | | | - Aina Abad-Lázaro
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - David Bartolomé-Català
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Aitor Otero
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Vanesa Fernández-Majada
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - James Sharpe
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Samuel Ojosnegros
- Bioengineering in Reproductive Health, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Jordi Comelles
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Department of Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain.
| | - Elena Martinez
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Department of Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain.
- Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
5
|
Micati D, Hlavca S, Chan WH, Abud HE. Harnessing 3D models to uncover the mechanisms driving infectious and inflammatory disease in the intestine. BMC Biol 2024; 22:300. [PMID: 39736603 DOI: 10.1186/s12915-024-02092-9] [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: 06/16/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
Representative models of intestinal diseases are transforming our knowledge of the molecular mechanisms of disease, facilitating effective drug screening and avenues for personalised medicine. Despite the emergence of 3D in vitro intestinal organoid culture systems that replicate the genetic and functional characteristics of the epithelial tissue of origin, there are still challenges in reproducing the human physiological tissue environment in a format that enables functional readouts. Here, we describe the latest platforms engineered to investigate environmental tissue impacts, host-microbe interactions and enable drug discovery. This highlights the potential to revolutionise knowledge on the impact of intestinal infection and inflammation and enable personalised disease modelling and clinical translation.
Collapse
Affiliation(s)
- Diana Micati
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Sara Hlavca
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Wing Hei Chan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
6
|
Wang J, Chen Z, Zhao P, Wang Y, Chen J, Lin Q. PDGFR-α shRNA-polyplex for uveal melanoma treatment via EMT mediated vasculogenic mimicry interfering. J Nanobiotechnology 2024; 22:797. [PMID: 39726008 DOI: 10.1186/s12951-024-03077-0] [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: 09/05/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Up to 50% of individuals with uveal melanoma (UM), a frequent cancer of the eye, pass away from metastases. One of the major challenges in treating UM is the role of receptor tyrosine kinases (RTKs), which mediate the epithelial-mesenchymal transition (EMT) of tumors. RTKs are involved in binding multiple growth factors, leading to angiogenesis and vasculogenic mimicry (VM) phenomena. Currently, most anti-angiogenic drugs have shown a tendency to increase the VM of tumors in clinical trials, resulting in limited efficacy. The existing gap in UM treatment lies in the lack of effective strategies to target RTK-mediated EMT and VM. While some approaches have been attempted, there is still a need for novel therapeutic interventions that can specifically interfere with these processes. This research employed the gene vector PEI-g-PEG to interfere with the platelet derived growth factor-alpha receptor (PDGFR-α)-mediated EMT process, thereby retarding the growth of UM. The cell experiments demonstrated that the gene polyplex exhibited favorable cell uptake and lysosome escape properties, effectively suppressing the expression of PDGFR-α protein and EMT marker proteins and the occurrence of VM phenomenon. In vivo animal studies also inhibited the growth of UM, and PAS assays showed that the treatment reduced the generation of VM in tumor tissue. This study broadens the application of PEI-g-PEG while interfering with the RTK-mediated tumor EMT process with the help of RNAi technology, providing a new idea for tumor reduction research.
Collapse
Affiliation(s)
- Jiahao Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhirong Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Peiyi Zhao
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yajia Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiang Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Quankui Lin
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
7
|
Bhattacharjee A, Rudolph S, Kaplan DL. Thermoplastic Molding of Silk-Curcumin Sustainable Composite Materials with Antibacterial Properties. ACS APPLIED BIO MATERIALS 2024; 7:8272-8280. [PMID: 39601507 DOI: 10.1021/acsabm.4c00950] [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] [Indexed: 11/29/2024]
Abstract
Silk fibroin (SF) is a natural protein generated from the Bombyx mori silkworm cocoons. It is useful for many different material applications. Versatile aqueous process engineering options can be used to support the morphological and structural modifications of silk materials related to tailored physical, chemical, and biological properties. Conventional solution-based processing methods, while effective, present process control limitations, thus, thermoplastic molding of regenerated SF-based composites was pursued to fabricate dense, functionalized plastics consisting of silk and curcumin. Curcumin, the active compound in turmeric (Curcuma longa) was incorporated into SF during the high-temperature processing, with the objective to investigate composite thermoplastics with enhanced biological properties from the curcumin due to the protective role of silk during processing. The results showed that a significantly higher amount of curcumin (∼25-fold) could be added into thermoplastic molded silk materials compared with the solution route, attributed to the hydrophobicity and low solubility of curcumin in solution-based routes. The curcumin-incorporated silk thermoplastics provided stability in acidic environments like the human gut, and slow curcumin (∼2% over 8 days) release from the materials. The protective silk-curcumin materials supported improved cytocompatibility with immortalized human colorectal adenocarcinoma (Caco-2) cells at high doses. The intestinal epithelial barrier integrity based on zonula occluden 1 (ZO-1) testing showed that the higher amount of curcumin in the thermoplastic molded silk had no negative effects on the intestinal barrier. The functionalized silk-based plastics also displayed microwave stability and antibacterial efficacy against both Gram-positive S. aureus and Gram-negative E. coli. These silk-based sustainable plastics, functionalized with curcumin, offer potential utility for a range of consumer and medical devices.
Collapse
Affiliation(s)
- Arjak Bhattacharjee
- Department of Biomedical Engineering Tufts University, Medford, Massachusetts 02155, United States
- Department of Materials and Metallurgical Engineering, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, United States
| | - Sara Rudolph
- Department of Biomedical Engineering Tufts University, Medford, Massachusetts 02155, United States
| | - David L Kaplan
- Department of Biomedical Engineering Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
8
|
Hofer M, Duque-Correa MA, Lutolf MP. Patterned gastrointestinal monolayers with bilateral access as observable models of parasite gut infection. Nat Biomed Eng 2024:10.1038/s41551-024-01313-4. [PMID: 39633029 PMCID: PMC7617323 DOI: 10.1038/s41551-024-01313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
Organoids for modelling the physiology and pathology of gastrointestinal tissues are constrained by a poorly accessible lumen. Here we report the development and applicability of bilaterally accessible organoid-derived patterned epithelial monolayers that allow the independent manipulation of their apical and basal sides. We constructed gastric, small-intestinal, caecal and colonic epithelial models that faithfully reproduced their respective tissue geometries and that exhibited stem cell regionalization and transcriptional resemblance to in vivo epithelia. The models' enhanced observability allowed single-cell tracking and studies of the motility of cells in immersion culture and at the air-liquid interface. Models mimicking infection of the caecal epithelium by the parasite Trichuris muris allowed us to live image syncytial tunnel formation. The enhanced observability of bilaterally accessible organoid-derived gastrointestinal tissue will facilitate the study of the dynamics of epithelial cells and their interactions with pathogens.
Collapse
Affiliation(s)
- Moritz Hofer
- Laboratory of Stem Cell Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Maria A Duque-Correa
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
| |
Collapse
|
9
|
Zhang Y, Qi F, Chen P, Liu BF, Li Y. Spatially defined microenvironment for engineering organoids. BIOPHYSICS REVIEWS 2024; 5:041302. [PMID: 39679203 PMCID: PMC11646138 DOI: 10.1063/5.0198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
In the intricately defined spatial microenvironment, a single fertilized egg remarkably develops into a conserved and well-organized multicellular organism. This observation leads us to hypothesize that stem cells or other seed cell types have the potential to construct fully structured and functional tissues or organs, provided the spatial cues are appropriately configured. Current organoid technology, however, largely depends on spontaneous growth and self-organization, lacking systematic guided intervention. As a result, the structures replicated in vitro often emerge in a disordered and sparse manner during growth phases. Although existing organoids have made significant contributions in many aspects, such as advancing our understanding of development and pathogenesis, aiding personalized drug selection, as well as expediting drug development, their potential in creating large-scale implantable tissue or organ constructs, and constructing multicomponent microphysiological systems, together with functioning at metabolic levels remains underutilized. Recent discoveries have demonstrated that the spatial definition of growth factors not only induces directional growth and migration of organoids but also leads to the formation of assembloids with multiple regional identities. This opens new avenues for the innovative engineering of higher-order organoids. Concurrently, the spatial organization of other microenvironmental cues, such as physical stresses, mechanical loads, and material composition, has been minimally explored. This review delves into the burgeoning field of organoid engineering with a focus on potential spatial microenvironmental control. It offers insight into the molecular principles, expected outcomes, and potential applications, envisioning a future perspective in this domain.
Collapse
Affiliation(s)
- Yilan Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
10
|
Walther N, Anantakrishnan S, Graham TGW, Dailey GM, Tjian R, Darzacq X. Automated live-cell single-molecule tracking in enteroid monolayers reveals transcription factor dynamics probing lineage-determining function. Cell Rep 2024; 43:114914. [PMID: 39480809 DOI: 10.1016/j.celrep.2024.114914] [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: 04/22/2024] [Revised: 08/29/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Lineage transcription factors (TFs) provide one regulatory level of differentiation crucial for the generation and maintenance of healthy tissues. To probe TF function by measuring their dynamics during adult intestinal homeostasis, we established HILO-illumination-based live-cell single-molecule tracking (SMT) in mouse small intestinal enteroid monolayers recapitulating tissue differentiation hierarchies in vitro. To increase the throughput, capture cellular features, and correlate morphological characteristics with diffusion parameters, we developed an automated imaging and analysis pipeline, broadly applicable to two-dimensional culture systems. Studying two absorptive lineage-determining TFs, we found an expression level-independent contrasting diffusive behavior: while Hes1, key determinant of absorptive lineage commitment, displays a large cell-to-cell variability and an average fraction of DNA-bound molecules of ∼32%, Hnf4g, conferring enterocyte identity, exhibits more uniform dynamics and a bound fraction of ∼56%. Our results suggest that TF diffusive behavior could indicate the progression of differentiation and modulate early versus late differentiation within a lineage.
Collapse
Affiliation(s)
- Nike Walther
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Sathvik Anantakrishnan
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA; Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Thomas G W Graham
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gina M Dailey
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Robert Tjian
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, Berkeley, CA 94720, USA.
| | - Xavier Darzacq
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
11
|
Cabel CR, Guzman BA, Alizadeh E, Li S, Holberg C, Wichaidit C, Cusanovich DA, Paek AL, Thatcher GRJ, Doorslaer KV, Nargi RS, Sutton RE, Suryadevara N, Crowe JE, Carnahan RH, Campos SK, Thorne CA. Cell-based high-content approach for SARS-CoV-2 neutralization identifies unique monoclonal antibodies and PI3K pathway inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616743. [PMID: 39416139 PMCID: PMC11483034 DOI: 10.1101/2024.10.04.616743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The sudden rise of the SARS-CoV-2 virus and the delay in the development of effective therapeutics to mitigate it made evident a need for ways to screen for compounds that can block infection and prevent further pathogenesis and spread. Yet, identifying effective drugs efficacious against viral infection and replication with minimal toxicity for the patient can be difficult. Monoclonal antibodies were shown to be effective, yet as the SARS-CoV-2 mutated, these antibodies became ineffective. Small molecule antivirals were identified using pseudovirus constructs to recapitulate infection in non-human cells, such as Vero E6 cells. However, the impact was limited due to poor translation of these compounds in the clinical setting. This is partly due to the lack of similarity of screening platforms to the in vivo physiology of the patient and partly because drugs effective in vitro showed dose-limiting toxicities. In this study, we performed two high-throughput screens in human lung adenocarcinoma cells with authentic SARS-CoV-2 virus to identify both monoclonal antibodies that neutralize the virus and clinically useful kinase inhibitors to block the virus and prioritize minimal host toxicity. Using high-content imaging combined with single-cell and multidimensional analysis, we identified antibodies and kinase inhibitors that reduce virus infection without affecting the host. Our screening technique uncovered novel antibodies and overlooked kinase inhibitors (i.e. PIK3i, mTORi, multiple RTKi) that could be effective against SARS-CoV-2 virus. Further characterization of these molecules will streamline the repurposing of compounds for the treatment of future pandemics and uncover novel mechanisms viruses use to hijack and infect host cells.
Collapse
|
12
|
Žukauskaitė K, Li M, Horvath A, Jarmalaitė S, Stadlbauer V. Cellular and Microbial In Vitro Modelling of Gastrointestinal Cancer. Cancers (Basel) 2024; 16:3113. [PMID: 39272971 PMCID: PMC11394127 DOI: 10.3390/cancers16173113] [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: 07/12/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Human diseases are multifaceted, starting with alterations at the cellular level, damaging organs and their functions, and disturbing interactions and immune responses. In vitro systems offer clarity and standardisation, which are crucial for effectively modelling disease. These models aim not to replicate every disease aspect but to dissect specific ones with precision. Controlled environments allow researchers to isolate key variables, eliminate confounding factors and elucidate disease mechanisms more clearly. Technological progress has rapidly advanced model systems. Initially, 2D cell culture models explored fundamental cell interactions. The transition to 3D cell cultures and organoids enabled more life-like tissue architecture and enhanced intercellular interactions. Advanced bioreactor-based devices now recreate the physicochemical environments of specific organs, simulating features like perfusion and the gastrointestinal tract's mucus layer, enhancing physiological relevance. These systems have been simplified and adapted for high-throughput research, marking significant progress. This review focuses on in vitro systems for modelling gastrointestinal tract cancer and the side effects of cancer treatment. While cell cultures and in vivo models are invaluable, our main emphasis is on bioreactor-based in vitro modelling systems that include the gut microbiome.
Collapse
Affiliation(s)
- Kristina Žukauskaitė
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
| | - Melissa Li
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Biotech Campus Tulln, Fachhochschule Wiener Neustadt, 3430 Tulln, Austria
| | - Angela Horvath
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Center for Biomarker Research in Medicine (CBmed GmbH), 8010 Graz, Austria
| | - Sonata Jarmalaitė
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
- National Cancer Institute, 08406 Vilnius, Lithuania
| | - Vanessa Stadlbauer
- Department of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria
- Center for Biomarker Research in Medicine (CBmed GmbH), 8010 Graz, Austria
| |
Collapse
|
13
|
Almalla A, Alzain N, Elomaa L, Richter F, Scholz J, Lindner M, Siegmund B, Weinhart M. Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models. Cells 2024; 13:1080. [PMID: 38994934 PMCID: PMC11240340 DOI: 10.3390/cells13131080] [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: 05/29/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.
Collapse
Affiliation(s)
- Ahed Almalla
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Nadra Alzain
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
- Department of Gastroenterology, Infectious Diseases and Rheumatology (Including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Fiona Richter
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Johanna Scholz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Marcus Lindner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology (Including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstr. 3A, 30167 Hannover, Germany
| |
Collapse
|
14
|
Han W, Lu G, Zhao S, Wang R, Zhang H, Liu K, Nie Y, Dong J. Rapid, Efficient, and Universally Applicable Genetic Engineering of Intestinal Organoid with a Sequential Monolayer to Three-Dimensional Strategy. Stem Cells Int 2024; 2024:2005845. [PMID: 38882597 PMCID: PMC11178405 DOI: 10.1155/2024/2005845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/14/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
Genetically modified intestinal organoids are being explored as potential surrogates of immortalized cell lines and gene-engineered animals. However, genetic manipulation of intestinal organoids is time-consuming, and the efficiency is far beyond satisfactory. To ensure the yield of the genetically modified organoids, large quantity of starting materials is required, and the procedure usually takes more than 10 days. Two major obstacles that restrict the genetic delivery efficiency are the three-dimensional culture condition and that the genetic delivery is carried out in cell suspensions. In the present study, we introduce a novel highly efficient strategy for building genetically modified intestinal organoids in which genetic delivery was performed in freshly established monolayer primary intestinal epithelial cells under two-dimensional conditions and subsequentially transformed into three-dimensional organoids. The total procedure can be finished within 10 hr while displaying much higher efficiency than the traditional methods. Furthermore, this strategy allowed for the selection of transgenic cells in monolayer conditions before establishing high-purity genetically modified intestinal organoids.
Collapse
Affiliation(s)
- Weili Han
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
| | - Guofang Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
| | - Sheng Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
| | - Rui Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
- Department of Psychiatry The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China
| | - Haohao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
| | - Kun Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
| | - Jiaqiang Dong
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases Xijing Hospital of Digestive Diseases Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
15
|
Barnett AM, Mullaney JA, McNabb WC, Roy NC. Culture media and format alter cellular composition and barrier integrity of porcine colonoid-derived monolayers. Tissue Barriers 2024; 12:2222632. [PMID: 37340938 DOI: 10.1080/21688370.2023.2222632] [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: 03/01/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023] Open
Abstract
Intestinal organoid technology has revolutionized our approach to in vitro cell culture due in part to their three-dimensional structures being more like the native tissue from which they were derived with respect to cellular composition and architecture. For this reason, organoids are becoming the new gold standard for undertaking intestinal epithelial cell research. Unfortunately, their otherwise advantageous three-dimensional geometry prevents easy access to the apical epithelium, which is a major limitation when studying interactions between dietary or microbial components and host tissues. To overcome this problem, we developed porcine colonoid-derived monolayers cultured on both permeable Transwell inserts and tissue culture treated polystyrene plates. We found that seeding density and culture format altered the expression of genes encoding markers of specific cell types (stem cells, colonocytes, goblets, and enteroendocrine cells), and barrier maturation (tight junctions). Additionally, we found that changes to the formulation of the culture medium altered the cellular composition of colonoids and of monolayers derived from them, resulting in cultures with an increasingly differentiated phenotype that was similar to that of their tissue of origin.
Collapse
Affiliation(s)
- Alicia M Barnett
- AgResearch Ltd, Grasslands Research Centre, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Jane A Mullaney
- AgResearch Ltd, Grasslands Research Centre, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Liggins Institute, The High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Liggins Institute, The High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Liggins Institute, The High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition, The University of Otago, Dunedin, New Zealand
| |
Collapse
|
16
|
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
|
17
|
Chu JTS, Lamers MM. Organoids in virology. NPJ VIRUSES 2024; 2:5. [PMID: 40295690 PMCID: PMC11721363 DOI: 10.1038/s44298-024-00017-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 04/30/2025]
Abstract
To adequately prepare against imminent disease outbreaks from diverse and ever-changing viral pathogens, improved experimental models that can accurately recapitulate host-virus responses and disease pathogenesis in human are essential. Organoid platforms have emerged in recent years as amenable in vitro tools that can bridge the limitations of traditional 2D cell lines and animal models for viral disease research. We highlight in this review the key insights that have contributed by organoid models to virus research, the limitations that exist in current platforms, and outline novel approaches that are being applied to address these shortcomings.
Collapse
Affiliation(s)
- Julie T S Chu
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Mart M Lamers
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
18
|
Zhao J, Wang X, Zhu J, Chukwudi C, Finebaum A, Zhang J, Yang S, He S, Saeidi N. PhaseFIT: live-organoid phase-fluorescent image transformation via generative AI. LIGHT, SCIENCE & APPLICATIONS 2023; 12:297. [PMID: 38097545 PMCID: PMC10721831 DOI: 10.1038/s41377-023-01296-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/02/2023] [Accepted: 09/24/2023] [Indexed: 12/17/2023]
Abstract
Organoid models have provided a powerful platform for mechanistic investigations into fundamental biological processes involved in the development and function of organs. Despite the potential for image-based phenotypic quantification of organoids, their complex 3D structure, and the time-consuming and labor-intensive nature of immunofluorescent staining present significant challenges. In this work, we developed a virtual painting system, PhaseFIT (phase-fluorescent image transformation) utilizing customized and morphologically rich 2.5D intestinal organoids, which generate virtual fluorescent images for phenotypic quantification via accessible and low-cost organoid phase images. This system is driven by a novel segmentation-informed deep generative model that specializes in segmenting overlap and proximity between objects. The model enables an annotation-free digital transformation from phase-contrast to multi-channel fluorescent images. The virtual painting results of nuclei, secretory cell markers, and stem cells demonstrate that PhaseFIT outperforms the existing deep learning-based stain transformation models by generating fine-grained visual content. We further validated the efficiency and accuracy of PhaseFIT to quantify the impacts of three compounds on crypt formation, cell population, and cell stemness. PhaseFIT is the first deep learning-enabled virtual painting system focused on live organoids, enabling large-scale, informative, and efficient organoid phenotypic quantification. PhaseFIT would enable the use of organoids in high-throughput drug screening applications.
Collapse
Affiliation(s)
- Junhan Zhao
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Surgery, Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Xiyue Wang
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Junyou Zhu
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Surgery, Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
- Shriners Hospital for Children-Boston, Boston, MA, 02114, USA
| | - Chijioke Chukwudi
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Surgery, Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
- Shriners Hospital for Children-Boston, Boston, MA, 02114, USA
| | - Andrew Finebaum
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jun Zhang
- Tencent AI Lab, Shenzhen, Guangdong, 518057, China
| | - Sen Yang
- Tencent AI Lab, Shenzhen, Guangdong, 518057, China
| | - Shijie He
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Surgery, Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA.
- Shriners Hospital for Children-Boston, Boston, MA, 02114, USA.
| | - Nima Saeidi
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Surgery, Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA.
- Shriners Hospital for Children-Boston, Boston, MA, 02114, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
19
|
Okolo O, Honzel E, Britton WR, Yu VX, Flashner S, Martin C, Nakagawa H, Parikh AS. Experimental Modeling of Host-Bacterial Interactions in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:5810. [PMID: 38136355 PMCID: PMC10742111 DOI: 10.3390/cancers15245810] [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: 11/07/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The microscopic species colonizing the human body, collectively referred to as the microbiome, play a crucial role in the maintenance of tissue homeostasis, immunity, and the development of disease. There is evidence to suggest associations between alterations in the microbiome and the development of head and neck squamous cell carcinomas (HNSCC). The use of two-dimensional (2D) modeling systems has made significant strides in uncovering the role of microbes in carcinogenesis; however, direct mechanistic links remain in their infancy. Patient-derived three-dimensional (3D) HNSCC organoid and organotypic models have recently been described. Compared to 2D models, 3D organoid culture systems effectively capture the genetic and epigenetic features of parent tissue in a patient-specific manner and may offer a more nuanced understanding of the role of host-microbe responses in carcinogenesis. This review provides a topical literature review assessing the current state of the field investigating the role of the microbiome in HNSCC; including in vivo and in vitro modeling methods that may be used to characterize microbiome-epithelial interactions.
Collapse
Affiliation(s)
- Ogoegbunam Okolo
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Columbia Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA;
| | - Emily Honzel
- Columbia Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA;
| | - William R. Britton
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Columbia Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA;
| | - Victoria X. Yu
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Department of Otolaryngology-Head and Neck Surgery, Columbia University, New York, NY 10027, USA
| | - Samuel Flashner
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
| | - Cecilia Martin
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Organoid and Cell Culture Core, Columbia University Digestive and Liver Diseases Research Center, Columbia University, New York, NY 10027, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Organoid and Cell Culture Core, Columbia University Digestive and Liver Diseases Research Center, Columbia University, New York, NY 10027, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Anuraag S. Parikh
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Department of Otolaryngology-Head and Neck Surgery, Columbia University, New York, NY 10027, USA
| |
Collapse
|
20
|
Bornholdt J, Müller CV, Nielsen MJ, Strickertsson J, Rago D, Chen Y, Maciag G, Skov J, Wellejus A, Schweiger PJ, Hansen SL, Broholm C, Gögenur I, Maimets M, Sloth S, Hendel J, Baker A, Sandelin A, Jensen KB. Detecting host responses to microbial stimulation using primary epithelial organoids. Gut Microbes 2023; 15:2281012. [PMID: 37992398 PMCID: PMC10730191 DOI: 10.1080/19490976.2023.2281012] [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: 05/11/2023] [Accepted: 11/05/2023] [Indexed: 11/24/2023] Open
Abstract
The intestinal epithelium is constantly exposed to microbes residing in the lumen. Traditionally, the response to microbial interactions has been studied in cell lines derived from cancerous tissues, e.g. Caco-2. It is, however, unclear how the responses in these cancer cell lines reflect the responses of a normal epithelium and whether there might be microbial strain-specific effects. To address these questions, we derived organoids from the small intestine from a cohort of healthy individuals. Culturing intestinal epithelium on a flat laminin matrix induced their differentiation, facilitating analysis of microbial responses via the apical membrane normally exposed to the luminal content. Here, it was evident that the healthy epithelium across multiple individuals (n = 9) demonstrates robust acute both common and strain-specific responses to a range of probiotic bacterial strains (BB-12Ⓡ, LGGⓇ, DSM33361, and Bif195). Importantly, parallel experiments using the Caco-2 cell line provide no acute response. Collectively, we demonstrate that primary epithelial cells maintained as organoids represent a valuable resource for assessing interactions between the epithelium and luminal microbes across individuals, and that these models are likely to contribute to a better understanding of host microbe interactions.
Collapse
Affiliation(s)
- Jette Bornholdt
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Human Health Research, Chr. Hansen AS, Hørsholm, Denmark
| | - Christina V. Müller
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Maria Juul Nielsen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Daria Rago
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yun Chen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Human Health Research, Chr. Hansen AS, Hørsholm, Denmark
| | - Grzegorz Maciag
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan Skov
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| | - Anja Wellejus
- Human Health Research, Chr. Hansen AS, Hørsholm, Denmark
| | - Pawel J. Schweiger
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| | - Stine L. Hansen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| | | | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Koge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martti Maimets
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| | - Stine Sloth
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Hendel
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Adam Baker
- Human Health Research, Chr. Hansen AS, Hørsholm, Denmark
| | - Albin Sandelin
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim B. Jensen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
21
|
Matsuoka Y, Tsujimoto Y. Housing conditions affect enterocyte death mode and turnover rate in mouse small intestine. Sci Rep 2023; 13:20423. [PMID: 37993588 PMCID: PMC10665386 DOI: 10.1038/s41598-023-47660-1] [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: 03/24/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023] Open
Abstract
Small intestinal enterocytes are continuously renewed. Shedding/death of enterocytes involves receptor-interacting protein kinase 1 (RIPK1)-dependent (but RIPK3-independent) necrotic death, but the regulatory mechanism of the processes is not fully understood. Here, we show that mouse housing conditions, such as the type of bedding material and the presence or absence of a Shepherd Shack, affect enterocyte turnover rate and determine whether enterocyte shedding/death is RIPK1-independent or -dependent. Mice housed with ALPHA-dri (αDri, hard paper chip) bedding material without a Shepherd Shack had a higher, largely RIPK1-dependent enterocyte turnover rate and higher blood corticosterone levels, suggesting the involvement of minor stress, whereas mice housed with αDri plus a Shepherd Shack or with Soft Chip had a lower, RIPK1-independent turnover rate and lower blood corticosterone levels. Corticosterone administration to a small intestine culture derived from mice housed with αDri plus a Shepherd Shack or with Soft Chip increased enterocyte shedding/death and turnover. By using kinase inhibitors and knockout mice, we showed that the switch from RIPK1-independent to RIPK1-dependent enterocyte shedding/death and turnover involves suppression of TANK-binding kinase 1. Our results demonstrate that housing conditions may cause minor stress, which alters the mode of enterocyte shedding/death and enterocyte turnover rate in mice.
Collapse
Affiliation(s)
- Yosuke Matsuoka
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan.
- Department of Molecular and Cellular Biology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan.
| | - Yoshihide Tsujimoto
- Department of Molecular and Cellular Biology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan.
| |
Collapse
|
22
|
Cui C, Li L, Wu L, Wang X, Zheng Y, Wang F, Wei H, Peng J. Paneth cells in farm animals: current status and future direction. J Anim Sci Biotechnol 2023; 14:118. [PMID: 37582766 PMCID: PMC10426113 DOI: 10.1186/s40104-023-00905-5] [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: 04/21/2023] [Accepted: 06/04/2023] [Indexed: 08/17/2023] Open
Abstract
A healthy intestine plays an important role in the growth and development of farm animals. In small intestine, Paneth cells are well known for their regulation of intestinal microbiota and intestinal stem cells (ISCs). Although there has been a lot of studies and reviews on human and murine Paneth cells under intestinal homeostasis or disorders, little is known about Paneth cells in farm animals. Most farm animals possess Paneth cells in their small intestine, as identified by various staining methods, and Paneth cells of various livestock species exhibit noticeable differences in cell shape, granule number, and intestinal distribution. Paneth cells in farm animals and their antimicrobial peptides (AMPs) are susceptible to multiple factors such as dietary nutrients and intestinal infection. Thus, the comprehensive understanding of Paneth cells in different livestock species will contribute to the improvement of intestinal health. This review first summarizes the current status of Paneth cells in pig, cattle, sheep, horse, chicken and rabbit, and points out future directions for the investigation of Paneth cells in the reviewed animals.
Collapse
Affiliation(s)
- Chenbin Cui
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lindeng Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lin Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xinru Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yao Zheng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Fangke Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 400700, China.
| |
Collapse
|
23
|
Moraitis I, Guiu J, Rubert J. Gut microbiota controlling radiation-induced enteritis and intestinal regeneration. Trends Endocrinol Metab 2023:S1043-2760(23)00108-X. [PMID: 37336645 DOI: 10.1016/j.tem.2023.05.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/21/2023]
Abstract
Cancer remains the second leading cause of mortality, with nearly 10 million deaths worldwide in 2020. In many cases, radiotherapy is used for its anticancer effects. However, radiation causes healthy tissue toxicity as a side effect. In intra-abdominal and pelvic malignancies, the healthy bowel is inevitably included in the radiation field, causing radiation-induced enteritis and dramatically affecting the gut microbiome. This condition is associated with significant morbidity and mortality that impairs cancer patients' and survivors' quality of life. This Review provides a critical overview of the main drivers in modulating the gut microenvironment in homeostasis, disease, and injury, focusing on gut microbial metabolites and microorganisms that influence epithelial regeneration upon radiation injury.
Collapse
Affiliation(s)
- Ilias Moraitis
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain; Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet de Llobregat, Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain; Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet de Llobregat, Spain.
| | - Josep Rubert
- Division of Human Nutrition and Health, Wageningen University & Research, Stippeneng 4, Wageningen, 6708, WE, Netherlands; Food Quality and Design, Wageningen University & Research, Bornse Weilanden 9, Wageningen, 6708, WG, Netherlands.
| |
Collapse
|
24
|
Li Y, Niu HM, Guo YX, Ma XK, Hu MX, Han JZ, Qin YM. Crypt-like patterned electrospun nanofibrous membrane and probiotics promote intestinal epithelium models close to tissues. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12602-4. [PMID: 37266585 DOI: 10.1007/s00253-023-12602-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/14/2023] [Accepted: 05/19/2023] [Indexed: 06/03/2023]
Abstract
In vitro intestinal epithelium models have drawn great attention to investigating intestinal biology in recent years. However, the difficulty to maintain the normal physiological status of primary intestinal epithelium in vitro limits the applications. Here, we designed patterned electrospun polylactic acid (PLA) nanofibrous membranes with crypt-like topography and mimic ECM fibrous network to support crypt culture and construct in vitro intestinal epithelium models. The patterned electrospun PLA nanofibrous membranes modified with Matrigels at 0 °C showed high biocompatibility and promoted cell growth and proliferation. The constructed duodenum epithelium models and colon epithelium models on the patterned electrospun PLA nanofibrous membranes expressed the typical differentiation markers of intestinal epithelia and the gene expression levels were close to the original tissues, especially with the help of probiotics. The constructed intestinal epithelium models could be used to assess probiotic adhesion and colonization, which were verified to show significant differences with the Caco-2 cell models due to the different cell types. These findings provide new insights and a better understanding of the roles of biophysical, biochemical, and biological signals in the construction of in vitro intestinal epithelium models as well as the potential applications of these models in the study of host-gut microbes interactions. KEY POINTS: • Patterned electrospun scaffold has crypt-like topography and ECM nanofibrous network. • Matrigels at 0°C modify scaffolds more effectively than at 37°C. • Synergy of biomimic scaffold and probiotics makes in vitro model close to tissue.
Collapse
Affiliation(s)
- Yue Li
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Hong-Mei Niu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Ya-Xin Guo
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Xue-Ke Ma
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Meng-Xin Hu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| | - Jian-Zhong Han
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yu-Mei Qin
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| |
Collapse
|
25
|
Snyder KB, Hunter CJ. Bugs and the barrier: A review of the gut microbiome and intestinal barrier in necrotizing enterocolitis. Semin Pediatr Surg 2023; 32:151310. [PMID: 37290337 DOI: 10.1016/j.sempedsurg.2023.151310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease that affects premature neonates. It frequently results in significant morbidity and mortality for those affected. Years of research into the pathophysiology of NEC have revealed it to be a variable and multifactorial disease. However, there are risk factors associated with NEC including low birth weight, prematurity, intestinal immaturity, alterations in microbial colonization, and history of rapid or formula based enteral feeds (Fig. 1).1-3 An accepted generalization of the pathogenesis of NEC includes a hyperresponsive immune reaction to insults such as ischemia, starting formula feeds, or alterations in the microbiome with pathologic bacterial colonization and translocation. This reaction causes a hyperinflammatory response disrupting the normal intestinal barrier, allowing abnormal bacterial translocation and ultimately sepsis.1,2,4 This review will focus specifically on the interactions with the microbiome and intestinal barrier function in NEC.
Collapse
Affiliation(s)
- K Brooke Snyder
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States
| | - Catherine J Hunter
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States.
| |
Collapse
|
26
|
Huang X, Nero T, Weerasekera R, Matej KH, Hinbest A, Jiang Z, Lee RF, Wu L, Chak C, Nijjer J, Gibaldi I, Yang H, Gamble N, Ng WL, Malaker SA, Sumigray K, Olson R, Yan J. Vibrio cholerae biofilms use modular adhesins with glycan-targeting and nonspecific surface binding domains for colonization. Nat Commun 2023; 14:2104. [PMID: 37055389 PMCID: PMC10102183 DOI: 10.1038/s41467-023-37660-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/24/2023] [Indexed: 04/15/2023] Open
Abstract
Bacterial biofilms are formed on environmental surfaces and host tissues, and facilitate host colonization and antibiotic resistance by human pathogens. Bacteria often express multiple adhesive proteins (adhesins), but it is often unclear whether adhesins have specialized or redundant roles. Here, we show how the model biofilm-forming organism Vibrio cholerae uses two adhesins with overlapping but distinct functions to achieve robust adhesion to diverse surfaces. Both biofilm-specific adhesins Bap1 and RbmC function as a "double-sided tape": they share a β-propeller domain that binds to the biofilm matrix exopolysaccharide, but have distinct environment-facing domains. Bap1 adheres to lipids and abiotic surfaces, while RbmC mainly mediates binding to host surfaces. Furthermore, both adhesins contribute to adhesion in an enteroid monolayer colonization model. We expect that similar modular domains may be utilized by other pathogens, and this line of research can potentially lead to new biofilm-removal strategies and biofilm-inspired adhesives.
Collapse
Affiliation(s)
- Xin Huang
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Thomas Nero
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Ranjuna Weerasekera
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, CT, USA
| | - Katherine H Matej
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Alex Hinbest
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, CT, USA
| | - Zhaowei Jiang
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Rebecca F Lee
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Longjun Wu
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Department of Ocean Science and Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong SAR, Guangzhou, Hong Kong SAR
| | - Cecilia Chak
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Japinder Nijjer
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Isabella Gibaldi
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, CT, USA
| | - Hang Yang
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, CT, USA
| | - Nathan Gamble
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, CT, USA
| | - Wai-Leung Ng
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Stacy A Malaker
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Kaelyn Sumigray
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Rich Olson
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, CT, USA.
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA.
- Quantitative Biology Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
27
|
Cohen-Kedar S, Shaham Barda E, Rabinowitz KM, Keizer D, Abu-Taha H, Schwartz S, Kaboub K, Baram L, Sadot E, White I, Wasserberg N, Wolff-Bar M, Levy-Barda A, Dotan I. Human intestinal epithelial cells can internalize luminal fungi via LC3-associated phagocytosis. Front Immunol 2023; 14:1142492. [PMID: 36969163 PMCID: PMC10030769 DOI: 10.3389/fimmu.2023.1142492] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Intestinal epithelial cells (IECs) are the first to encounter luminal microorganisms and actively participate in intestinal immunity. We reported that IECs express the β-glucan receptor Dectin-1, and respond to commensal fungi and β-glucans. In phagocytes, Dectin-1 mediates LC3-associated phagocytosis (LAP) utilizing autophagy components to process extracellular cargo. Dectin-1 can mediate phagocytosis of β-glucan-containing particles by non-phagocytic cells. We aimed to determine whether human IECs phagocytose β-glucan-containing fungal particles via LAP. METHODS Colonic (n=18) and ileal (n=4) organoids from individuals undergoing bowel resection were grown as monolayers. Fluorescent-dye conjugated zymosan (β-glucan particle), heat-killed- and UV inactivated C. albicans were applied to differentiated organoids and to human IEC lines. Confocal microscopy was used for live imaging and immuno-fluorescence. Quantification of phagocytosis was carried out with a fluorescence plate-reader. RESULTS zymosan and C. albicans particles were phagocytosed by monolayers of human colonic and ileal organoids and IEC lines. LAP was identified by LC3 and Rubicon recruitment to phagosomes and lysosomal processing of internalized particles was demonstrated by co-localization with lysosomal dyes and LAMP2. Phagocytosis was significantly diminished by blockade of Dectin-1, actin polymerization and NAPDH oxidases. CONCLUSIONS Our results show that human IECs sense luminal fungal particles and internalize them via LAP. This novel mechanism of luminal sampling suggests that IECs may contribute to the maintenance of mucosal tolerance towards commensal fungi.
Collapse
Affiliation(s)
- Sarit Cohen-Kedar
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Efrat Shaham Barda
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Keren Masha Rabinowitz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Danielle Keizer
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Hanan Abu-Taha
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shoshana Schwartz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Kawsar Kaboub
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Liran Baram
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Sadot
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Ian White
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Wasserberg
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Meirav Wolff-Bar
- Department of Pathology, Rabin Medical Center, Petah-Tikva, Israel
| | | | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Liebe H, Schlegel C, Cai X, Golubkova A, Loerke C, Leiva T, Hunter CJ. Apical-Out Enteroids as an Innovative Model for Necrotizing Enterocolitis. J Surg Res 2023; 283:1106-1116. [PMID: 36915002 PMCID: PMC10014931 DOI: 10.1016/j.jss.2022.11.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 11/15/2022] [Accepted: 11/20/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Necrotizing enterocolitis (NEC) is a gastrointestinal disease of premature neonates. We previously validated a NEC enteroid model derived from human infant intestinal tissue. Typical enteroid configuration is basolateral-out (BO) without direct access to the luminal (apical) surface. Apical access is necessary to allow physiologic comparison of pathogen interaction with the intestinal epithelial barrier. We hypothesize that apical-out (AO) enteroids will provide a relevant NEC model to study this relationship. METHODS Following the institutional review board approval (#11610-11611), neonatal intestinal tissue was collected from surgical specimens. Stem cells were collected; enteroids were generated and grown to maturity in BO conformation then everted to AO. Enteroids were untreated or treated for 24 h with 100 μg/mL lipopolysaccharide and hypoxia. Protein and gene expression were analyzed for inflammatory markers, tight junction (TJ) proteins and permeability characteristic of NEC. RESULTS Apical TJ protein zonula occludens-1 and basolateral protein β-catenin immunofluorescence confirmed AO configuration. Treated AO enteroids had significantly increased messenger RNA (P = 0.001) and protein levels (P < 0.0001) of tumor necrosis factor-α compared to controls. Corrected total cell fluorescence of toll-like receptor 4 was significantly increased in treated AO enteroids compared to control (P = 0.002). Occludin was found to have significantly decreased messenger RNA in treated AO enteroids (P = 0.003). Expression of other TJ proteins claudins-1, -4 and zonula occludens-1 was significantly decreased in treated AO enteroids (P < 0.05). CONCLUSIONS AO enteroids present an innovative model for NEC with increased inflammation and gut barrier restructuring. This model allows for a biologically relevant investigation of the interaction between the pathogen and the intestinal epithelial barrier in NEC.
Collapse
Affiliation(s)
- Heather Liebe
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma.
| | - Camille Schlegel
- The University of Oklahoma Health Sciences Center, Department of Surgery, Oklahoma City, Oklahoma
| | - Xue Cai
- The University of Oklahoma Health Sciences Center, Department of Surgery, Oklahoma City, Oklahoma
| | - Alena Golubkova
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma
| | | | - Tyler Leiva
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma
| | - Catherine J Hunter
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma
| |
Collapse
|
29
|
Schamberger B, Ziege R, Anselme K, Ben Amar M, Bykowski M, Castro APG, Cipitria A, Coles RA, Dimova R, Eder M, Ehrig S, Escudero LM, Evans ME, Fernandes PR, Fratzl P, Geris L, Gierlinger N, Hannezo E, Iglič A, Kirkensgaard JJK, Kollmannsberger P, Kowalewska Ł, Kurniawan NA, Papantoniou I, Pieuchot L, Pires THV, Renner LD, Sageman-Furnas AO, Schröder-Turk GE, Sengupta A, Sharma VR, Tagua A, Tomba C, Trepat X, Waters SL, Yeo EF, Roschger A, Bidan CM, Dunlop JWC. Curvature in Biological Systems: Its Quantification, Emergence, and Implications across the Scales. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206110. [PMID: 36461812 DOI: 10.1002/adma.202206110] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Surface curvature both emerges from, and influences the behavior of, living objects at length scales ranging from cell membranes to single cells to tissues and organs. The relevance of surface curvature in biology is supported by numerous experimental and theoretical investigations in recent years. In this review, first, a brief introduction to the key ideas of surface curvature in the context of biological systems is given and the challenges that arise when measuring surface curvature are discussed. Giving an overview of the emergence of curvature in biological systems, its significance at different length scales becomes apparent. On the other hand, summarizing current findings also shows that both single cells and entire cell sheets, tissues or organisms respond to curvature by modulating their shape and their migration behavior. Finally, the interplay between the distribution of morphogens or micro-organisms and the emergence of curvature across length scales is addressed with examples demonstrating these key mechanistic principles of morphogenesis. Overall, this review highlights that curved interfaces are not merely a passive by-product of the chemical, biological, and mechanical processes but that curvature acts also as a signal that co-determines these processes.
Collapse
Affiliation(s)
- Barbara Schamberger
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| | - Ricardo Ziege
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Karine Anselme
- IS2M (CNRS - UMR 7361), Université de Haute-Alsace, F-68100, Mulhouse, France
- Université de Strasbourg, F-67081, Strasbourg, France
| | - Martine Ben Amar
- Department of Physics, Laboratoire de Physique de l'Ecole Normale Supérieure, 24 rue Lhomond, 75005, Paris, France
| | - Michał Bykowski
- Department of Plant Anatomy and Cytology, Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
| | - André P G Castro
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisboa, Portugal
- ESTS, Instituto Politécnico de Setúbal, 2914-761, Setúbal, Portugal
| | - Amaia Cipitria
- IS2M (CNRS - UMR 7361), Université de Haute-Alsace, F-68100, Mulhouse, France
- Group of Bioengineering in Regeneration and Cancer, Biodonostia Health Research Institute, 20014, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, 48009, Bilbao, Spain
| | - Rhoslyn A Coles
- Cluster of Excellence, Matters of Activity, Humboldt-Universität zu Berlin, 10178, Berlin, Germany
| | - Rumiana Dimova
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Michaela Eder
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Sebastian Ehrig
- Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- Berlin Institute for Medical Systems Biology, 10115, Berlin, Germany
| | - Luis M Escudero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Universidad de Sevilla, 41013, Seville, Spain
- Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Myfanwy E Evans
- Institute for Mathematics, University of Potsdam, 14476, Potsdam, Germany
| | - Paulo R Fernandes
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Peter Fratzl
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In Silico Medicine, University of Liège, 4000, Liège, Belgium
| | - Notburga Gierlinger
- Institute of Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna (Boku), 1190, Vienna, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria, 3400, Klosterneuburg, Austria
| | - Aleš Iglič
- Laboratory of Physics, Faculty of Electrical engineering, University of Ljubljana, Tržaška 25, SI-1000, Ljubljana, Slovenia
| | - Jacob J K Kirkensgaard
- Condensed Matter Physics, Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100, København Ø, Denmark
- Ingredients and Dairy Technology, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg, Denmark
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, University of Würzburg, 97074, Würzburg, Germany
| | - Łucja Kowalewska
- Department of Plant Anatomy and Cytology, Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Ioannis Papantoniou
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, PB 813, 3000, Leuven, Belgium
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology (FORTH), Stadiou Str., 26504, Patras, Greece
| | - Laurent Pieuchot
- IS2M (CNRS - UMR 7361), Université de Haute-Alsace, F-68100, Mulhouse, France
- Université de Strasbourg, F-67081, Strasbourg, France
| | - Tiago H V Pires
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Lars D Renner
- Leibniz Institute of Polymer Research and the Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | | | - Gerd E Schröder-Turk
- School of Physics, Chemistry and Mathematics, Murdoch University, 90 South St, Murdoch, WA, 6150, Australia
- Department of Materials Physics, Research School of Physics, The Australian National University, Canberra, ACT, 2600, Australia
| | - Anupam Sengupta
- Physics of Living Matter, Department of Physics and Materials Science, University of Luxembourg, L-1511, Luxembourg City, Grand Duchy of Luxembourg
| | - Vikas R Sharma
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| | - Antonio Tagua
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Universidad de Sevilla, 41013, Seville, Spain
- Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Caterina Tomba
- Univ Lyon, CNRS, INSA Lyon, Ecole Centrale de Lyon, Université Claude Bernard Lyon 1, CPE Lyon, INL, UMR5270, 69622, Villeurbanne, France
| | - Xavier Trepat
- ICREA at the Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08028, Barcelona, Spain
| | - Sarah L Waters
- Mathematical Institute, University of Oxford, OX2 6GG, Oxford, UK
| | - Edwina F Yeo
- Mathematical Institute, University of Oxford, OX2 6GG, Oxford, UK
| | - Andreas Roschger
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| | - Cécile M Bidan
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476, Potsdam, Germany
| | - John W C Dunlop
- Department of the Chemistry and Physics of Materials, Paris-Lodron University of Salzburg, 5020, Salzburg, Austria
| |
Collapse
|
30
|
Castillo-Azofeifa D, Wald T, Reyes EA, Gallagher A, Schanin J, Vlachos S, Lamarche-Vane N, Bomidi C, Blutt S, Estes MK, Nystul T, Klein OD. A DLG1-ARHGAP31-CDC42 axis is essential for the intestinal stem cell response to fluctuating niche Wnt signaling. Cell Stem Cell 2023; 30:188-206.e6. [PMID: 36640764 PMCID: PMC9922544 DOI: 10.1016/j.stem.2022.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/13/2022] [Accepted: 12/12/2022] [Indexed: 01/15/2023]
Abstract
A central factor in the maintenance of tissue integrity is the response of stem cells to variations in the levels of niche signals. In the gut, intestinal stem cells (ISCs) depend on Wnt ligands for self-renewal and proliferation. Transient increases in Wnt signaling promote regeneration after injury or in inflammatory bowel diseases, whereas constitutive activation of this pathway leads to colorectal cancer. Here, we report that Discs large 1 (Dlg1), although dispensable for polarity and cellular turnover during intestinal homeostasis, is required for ISC survival in the context of increased Wnt signaling. RNA sequencing (RNA-seq) and genetic mouse models demonstrated that DLG1 regulates the cellular response to increased canonical Wnt ligands. This occurs via the transcriptional regulation of Arhgap31, a GTPase-activating protein that deactivates CDC42, an effector of the non-canonical Wnt pathway. These findings reveal a DLG1-ARHGAP31-CDC42 axis that is essential for the ISC response to increased niche Wnt signaling.
Collapse
Affiliation(s)
- David Castillo-Azofeifa
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Regenerative Medicine, Genentech, Inc., South San Francisco, CA, USA
| | - Tomas Wald
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Efren A Reyes
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pharmaceutical Chemistry and TETRAD Program, University of California, San Francisco, San Francisco, CA, USA
| | - Aaron Gallagher
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Schanin
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie Vlachos
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| | - Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sarah Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Todd Nystul
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Matejčić M, Trepat X. Mechanobiological approaches to synthetic morphogenesis: learning by building. Trends Cell Biol 2023; 33:95-111. [PMID: 35879149 DOI: 10.1016/j.tcb.2022.06.013] [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: 04/26/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 01/25/2023]
Abstract
Tissue morphogenesis occurs in a complex physicochemical microenvironment with limited experimental accessibility. This often prevents a clear identification of the processes that govern the formation of a given functional shape. By applying state-of-the-art methods to minimal tissue systems, synthetic morphogenesis aims to engineer the discrete events that are necessary and sufficient to build specific tissue shapes. Here, we review recent advances in synthetic morphogenesis, highlighting how a combination of microfabrication and mechanobiology is fostering our understanding of how tissues are built.
Collapse
Affiliation(s)
- Marija Matejčić
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain; Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain.
| |
Collapse
|
32
|
Stevens AJ, Harris AR, Gerdts J, Kim KH, Trentesaux C, Ramirez JT, McKeithan WL, Fattahi F, Klein OD, Fletcher DA, Lim WA. Programming multicellular assembly with synthetic cell adhesion molecules. Nature 2023; 614:144-152. [PMID: 36509107 PMCID: PMC9892004 DOI: 10.1038/s41586-022-05622-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
Cell adhesion molecules are ubiquitous in multicellular organisms, specifying precise cell-cell interactions in processes as diverse as tissue development, immune cell trafficking and the wiring of the nervous system1-4. Here we show that a wide array of synthetic cell adhesion molecules can be generated by combining orthogonal extracellular interactions with intracellular domains from native adhesion molecules, such as cadherins and integrins. The resulting molecules yield customized cell-cell interactions with adhesion properties that are similar to native interactions. The identity of the intracellular domain of the synthetic cell adhesion molecules specifies interface morphology and mechanics, whereas diverse homotypic or heterotypic extracellular interaction domains independently specify the connectivity between cells. This toolkit of orthogonal adhesion molecules enables the rationally programmed assembly of multicellular architectures, as well as systematic remodelling of native tissues. The modularity of synthetic cell adhesion molecules provides fundamental insights into how distinct classes of cell-cell interfaces may have evolved. Overall, these tools offer powerful abilities for cell and tissue engineering and for systematically studying multicellular organization.
Collapse
Affiliation(s)
- Adam J Stevens
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
| | - Andrew R Harris
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Ontario, Canada
| | - Josiah Gerdts
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco, CA, USA
| | - Ki H Kim
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
| | - Coralie Trentesaux
- Program in Craniofacial Biology, University of California, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Jonathan T Ramirez
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Wesley L McKeithan
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Maze Therapeutics, San Francisco, CA, USA
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology, University of California, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel A Fletcher
- Center for Cellular Construction, University of California, San Francisco, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Wendell A Lim
- UCSF Cell Design Institute, University of California, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
- Center for Cellular Construction, University of California, San Francisco, CA, USA.
| |
Collapse
|
33
|
Ahn JS, Kang MJ, Seo Y, Kim HS. Intestinal organoids as advanced modeling platforms to study the role of host-microbiome interaction in homeostasis and disease. BMB Rep 2023; 56:15-23. [PMID: 36379514 PMCID: PMC9887104 DOI: 10.5483/bmbrep.2022-0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 01/09/2025] Open
Abstract
After birth, animals are colonized by a diverse community of microorganisms. The digestive tract is known to contain the largest number of microbiome in the body. With emergence of the gut-brain axis, the importance of gut microbiome and its metabolites in host health has been extensively studied in recent years. The establishment of organoid culture systems has contributed to studying intestinal pathophysiology by replacing current limited models. Owing to their architectural and functional complexity similar to a real organ, co-culture of intestinal organoids with gut microbiome can provide mechanistic insights into the detrimental role of pathobiont and the homeostatic function of commensal symbiont. Here organoid-based bacterial co-culture techniques for modeling host-microbe interactions are reviewed. This review also summarizes representative studies that explore impact of enteric microorganisms on intestinal organoids to provide a better understanding of host-microbe interaction in the context of homeostasis and disease. [BMB Reports 2023; 56(1): 15-23].
Collapse
Affiliation(s)
- Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
34
|
Ahn JS, Kang MJ, Seo Y, Kim HS. Intestinal organoids as advanced modeling platforms to study the role of host-microbiome interaction in homeostasis and disease. BMB Rep 2023; 56:15-23. [PMID: 36379514 PMCID: PMC9887104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 01/28/2023] Open
Abstract
After birth, animals are colonized by a diverse community of microorganisms. The digestive tract is known to contain the largest number of microbiome in the body. With emergence of the gut-brain axis, the importance of gut microbiome and its metabolites in host health has been extensively studied in recent years. The establishment of organoid culture systems has contributed to studying intestinal pathophysiology by replacing current limited models. Owing to their architectural and functional complexity similar to a real organ, co-culture of intestinal organoids with gut microbiome can provide mechanistic insights into the detrimental role of pathobiont and the homeostatic function of commensal symbiont. Here organoid-based bacterial co-culture techniques for modeling host-microbe interactions are reviewed. This review also summarizes representative studies that explore impact of enteric microorganisms on intestinal organoids to provide a better understanding of host-microbe interaction in the context of homeostasis and disease. [BMB Reports 2023; 56(1): 15-23].
Collapse
Affiliation(s)
- Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
35
|
Dalleywater W, Wheat F, Sculthorpe D, Hyland G, Ilyas M. In Vitro Culture and Histological Evaluation of 3D Organotypic Cultures. Methods Mol Biol 2023; 2650:155-170. [PMID: 37310631 DOI: 10.1007/978-1-0716-3076-1_13] [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] [Indexed: 06/14/2023]
Abstract
Organotypic cultures allow cells to grow in a system which mimics in vivo tissue organization. Here we describe a method for establishing 3D organotypic cultures (using intestine as an example system), followed by methods for demonstrating cell morphology and tissue architecture using histological techniques and molecular expression analysis using immunohistochemistry, though the system is also amenable to molecular expression analysis, such as by PCR, RNA sequencing, or FISH.
Collapse
Affiliation(s)
- William Dalleywater
- Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK.
- Department of Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham, UK.
| | - Francesca Wheat
- Department of Cellular Pathology, University Hospitals of Leicester NHS Trust, Nottingham, UK
| | - Declan Sculthorpe
- Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Georgina Hyland
- Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Mohammad Ilyas
- Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| |
Collapse
|
36
|
Jayawardena D, Priyamvada S, Kageyama T, White Z, Kumar A, Griggs TF, Majumder A, Akram R, Anbazhagan AN, Sano T, Dudeja PK. Loss of SLC26A3 Results in Colonic Mucosal Immune Dysregulation via Epithelial-Immune Cell Crosstalk. Cell Mol Gastroenterol Hepatol 2022; 15:903-919. [PMID: 36535508 PMCID: PMC9971172 DOI: 10.1016/j.jcmgh.2022.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Down-regulation of chloride transporter SLC26A3 or down-regulated in adenoma (DRA) in colonocytes has recently been linked to the pathogenesis of ulcerative colitis (UC). Because exaggerated immune responses are one of the hallmarks of UC, these current studies were undertaken to define the mechanisms by which loss of DRA relays signals to immune cells to increase susceptibility to inflammation. METHODS NanoString Immunology Panel, fluorescence assisted cell sorting, immunoblotting, immunofluorescence, and quantitative real-time polymerase chain reaction assays were used in wild-type and DRA knockout (KO) mice. Interleukin (IL)-33 blocking was used to determine specific changes in immune cells and co-housing/broad spectrum antibiotics administration, and ex vivo studies in colonoids were conducted to rule out the involvement of microbiota. Colonoid-derived monolayers from healthy and UC patient biopsies were analyzed for translatability. RESULTS There was a marked induction of Th2 (>2-fold), CD4+ Th2 cells (∼8-fold), RORγt+ Th17, and FOXP3+ regulatory T cells (Tregs). DRA KO colons also exhibited a robust induction of IL-33 (>8-fold). In vivo studies using blocking of IL-33 established that T2 immune dysregulation (alterations in ILC2, Th2, and GATA3+ iTregs) in response to loss of DRA was due to altered epithelial-immune cell crosstalk via IL-33. CONCLUSIONS Loss of DRA in colonocytes triggers the release of IL-33 to drive a type 2 immune response. These observations emphasize the critical importance of DRA in mucosal immune homeostasis and its implications in the pathogenesis of UC.
Collapse
Affiliation(s)
- Dulari Jayawardena
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Shubha Priyamvada
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Takahiro Kageyama
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - Anoop Kumar
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois
| | - Theodor F Griggs
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Apurba Majumder
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ramsha Akram
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | | | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - Pradeep K Dudeja
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Jesse Brown VA Medical Center, Chicago, Illinois.
| |
Collapse
|
37
|
Gao Y, Dong J, Qi S, Zhou X, Wu X, Wang W, Wen L, Fu W, Tang F. Establishment and characterization of adult human gastric epithelial progenitor‐like cell lines. Cell Prolif 2022:e13355. [PMID: 36331058 DOI: 10.1111/cpr.13355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/23/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yuan Gao
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery Third Hospital, Peking University Beijing China
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies Peking University Beijing China
- Beijing Advanced Innovation Center for Genomics Ministry of Education Key Laboratory of Cell Proliferation and Differentiation Beijing China
| | - Ji Dong
- Guangzhou Laboratory Guangzhou China
| | - Shuyue Qi
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery Third Hospital, Peking University Beijing China
- Beijing Advanced Innovation Center for Genomics Ministry of Education Key Laboratory of Cell Proliferation and Differentiation Beijing China
| | - Xin Zhou
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery Third Hospital, Peking University Beijing China
- Peking University Third Hospital Cancer Center Peking University Third Hospital Beijing China
| | - Xinglong Wu
- College of Animal Science and Technology Hebei Agricultural University Baoding Hebei China
| | - Wendong Wang
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery Third Hospital, Peking University Beijing China
- Peking University Third Hospital Cancer Center Peking University Third Hospital Beijing China
| | - Lu Wen
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery Third Hospital, Peking University Beijing China
- Beijing Advanced Innovation Center for Genomics Ministry of Education Key Laboratory of Cell Proliferation and Differentiation Beijing China
| | - Wei Fu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery Third Hospital, Peking University Beijing China
- Peking University Third Hospital Cancer Center Peking University Third Hospital Beijing China
| | - Fuchou Tang
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery Third Hospital, Peking University Beijing China
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies Peking University Beijing China
- Beijing Advanced Innovation Center for Genomics Ministry of Education Key Laboratory of Cell Proliferation and Differentiation Beijing China
| |
Collapse
|
38
|
Altay G, Abad‐Lázaro A, Gualda EJ, Folch J, Insa C, Tosi S, Hernando‐Momblona X, Batlle E, Loza‐Álvarez P, Fernández‐Majada V, Martinez E. Modeling Biochemical Gradients In Vitro to Control Cell Compartmentalization in a Microengineered 3D Model of the Intestinal Epithelium. Adv Healthc Mater 2022; 11:e2201172. [PMID: 36073021 PMCID: PMC11468757 DOI: 10.1002/adhm.202201172] [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: 06/16/2022] [Revised: 08/29/2022] [Indexed: 01/28/2023]
Abstract
Gradients of signaling pathways within the intestinal stem cell (ISC) niche are instrumental for cellular compartmentalization and tissue function, yet how are they sensed by the epithelium is still not fully understood. Here a new in vitro model of the small intestine based on primary epithelial cells (i), apically accessible (ii), with native tissue mechanical properties and controlled mesh size (iii), 3D villus-like architecture (iv), and precisely controlled biomolecular gradients of the ISC niche (v) is presented. Biochemical gradients are formed through hydrogel-based scaffolds by free diffusion from a source to a sink chamber. To confirm the establishment of spatiotemporally controlled gradients, light-sheet fluorescence microscopy and in-silico modeling are employed. The ISC niche biochemical gradients coming from the stroma and applied along the villus axis lead to the in vivo-like compartmentalization of the proliferative and differentiated cells, while changing the composition and concentration of the biochemical factors affects the cellular organization along the villus axis. This novel 3D in vitro intestinal model derived from organoids recapitulates both the villus-like architecture and the gradients of ISC biochemical factors, thus opening the possibility to study in vitro the nature of such gradients and the resulting cellular response.
Collapse
Affiliation(s)
- Gizem Altay
- Biomimetic Systems for Cell Engineering LaboratoryInstitute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 15‐21Barcelona08028Spain
- Institut de l'AuditionInstitut PasteurINSERMUniversité de ParisParis75012France
| | - Aina Abad‐Lázaro
- Biomimetic Systems for Cell Engineering LaboratoryInstitute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 15‐21Barcelona08028Spain
| | - Emilio J. Gualda
- SLN Research FacilityInstitute of Photonic Sciences (ICFO)Mediterranean Technology ParkAv. Carl Friedrich Gauss 3 CastelldefelsBarcelona08860Spain
| | - Jordi Folch
- Biomimetic Systems for Cell Engineering LaboratoryInstitute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 15‐21Barcelona08028Spain
| | - Claudia Insa
- Biomimetic Systems for Cell Engineering LaboratoryInstitute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 15‐21Barcelona08028Spain
| | - Sébastien Tosi
- Advanced Digital Microscopy Core Facility (ADMCF)Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 10‐12Barcelona08028Spain
| | - Xavier Hernando‐Momblona
- Colorectal Cancer LaboratoryInstitute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 10‐12Barcelona08028Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Barcelona08028Spain
| | - Eduard Batlle
- Colorectal Cancer LaboratoryInstitute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 10‐12Barcelona08028Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Barcelona08028Spain
- ICREAPasseig Lluís Companys 23Barcelona08010Spain
| | - Pablo Loza‐Álvarez
- SLN Research FacilityInstitute of Photonic Sciences (ICFO)Mediterranean Technology ParkAv. Carl Friedrich Gauss 3 CastelldefelsBarcelona08860Spain
| | - Vanesa Fernández‐Majada
- Biomimetic Systems for Cell Engineering LaboratoryInstitute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 15‐21Barcelona08028Spain
| | - Elena Martinez
- Biomimetic Systems for Cell Engineering LaboratoryInstitute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)Baldiri i Reixac 15‐21Barcelona08028Spain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBER‐BBN)Av. Monforte de Lemos 3‐5 Pabellón 11 Planta 0Madrid28029Spain
- Department of Electronics and Biomedical EngineeringUniversity of Barcelona (UB)Martí i Franquès 1Barcelona08028Spain
| |
Collapse
|
39
|
Rudolph SE, Longo BN, Tse MW, Houchin MR, Shokoufandeh MM, Chen Y, Kaplan DL. Crypt-Villus Scaffold Architecture for Bioengineering Functional Human Intestinal Epithelium. ACS Biomater Sci Eng 2022; 8:4942-4955. [PMID: 36191009 PMCID: PMC10379436 DOI: 10.1021/acsbiomaterials.2c00851] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Crypt-villus architecture in the small intestine is crucial for the structural integrity of the intestinal epithelium and maintenance of gut homeostasis. We utilized three-dimensional (3D) printing and inverse molding techniques to form three-dimensional (3D) spongy scaffold systems that resemble the intestinal crypt-villus microarchitecture. The scaffolds consist of silk fibroin protein with curved lumens with rows of protruding villi with invaginating crypts to generate the architecture. Intestinal cell (Caco-2, HT29-MTX) attachment and growth, as well as long-term culture support were demonstrated with cell polarization and tissue barrier properties compared to two-dimensional (2D) Transwell culture controls. Further, physiologically relevant oxygen gradients were generated in the 3D system. The various advantages of this system may be ascribed to the more physiologically relevant 3D environment, offering a system for the exploration of disease pathogenesis, host-microbiome interactions, and therapeutic discovery.
Collapse
Affiliation(s)
- Sara E Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Brooke N Longo
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Megan W Tse
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Megan R Houchin
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Mina M Shokoufandeh
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
40
|
Felsenthal N, Vignjevic DM. Stand by me: Fibroblasts regulation of the intestinal epithelium during development and homeostasis. Curr Opin Cell Biol 2022; 78:102116. [PMID: 35914344 DOI: 10.1016/j.ceb.2022.102116] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 01/31/2023]
Abstract
The epithelium of the small intestine is composed of a single layer of cells that line two functionally distinct compartments, the villi that project into the lumen of the gut and the crypts that descend into the underlying connective tissue. Stem cells are located in crypts, where they divide and give rise to transit-amplifying cells that differentiate into secretory and absorptive epithelial cells. Most differentiated cells travel upwards from the crypt towards the villus tip, where they shed into the lumen. While some of these cell behaviors are an intrinsic property of the epithelium, it is becoming evident that tight coordination between the epithelium and the underlying fibroblasts plays a critical role in tissue morphogenesis, stem-cell niche maintenance and regionalized gene expression along the crypt-villus axis. Here, we will review the current literature describing the interaction between epithelium and fibroblasts during crypt-villus axis development and intestinal epithelium renewal during homeostasis.
Collapse
Affiliation(s)
- Neta Felsenthal
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005 Paris, France.
| | | |
Collapse
|
41
|
Pond KW, Morris JM, Alkhimenok O, Varghese RP, Cabel CR, Ellis NA, Chakrabarti J, Zavros Y, Merchant JL, Thorne CA, Paek AL. Live-cell imaging in human colonic monolayers reveals ERK waves limit the stem cell compartment to maintain epithelial homeostasis. eLife 2022; 11:e78837. [PMID: 36094159 PMCID: PMC9499537 DOI: 10.7554/elife.78837] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/11/2022] [Indexed: 11/13/2022] Open
Abstract
The establishment and maintenance of different cellular compartments in tissues is a universal requirement across all metazoans. Maintaining the correct ratio of cell types in time and space allows tissues to form patterned compartments and perform complex functions. Patterning is especially evident in the human colon, where tissue homeostasis is maintained by stem cells in crypt structures that balance proliferation and differentiation. Here, we developed a human 2D patient derived organoid screening platform to study tissue patterning and kinase pathway dynamics in single cells. Using this system, we discovered that waves of ERK signaling induced by apoptotic cells play a critical role in maintaining tissue patterning and homeostasis. If ERK is activated acutely across all cells instead of in wave-like patterns, then tissue patterning and stem cells are lost. Conversely, if ERK activity is inhibited, then stem cells become unrestricted and expand dramatically. This work demonstrates that the colonic epithelium requires coordinated ERK signaling dynamics to maintain patterning and tissue homeostasis. Our work reveals how ERK can antagonize stem cells while supporting cell replacement and the function of the gut.
Collapse
Affiliation(s)
- Kelvin W Pond
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
- Department of Molecular and Cellular Biology, The University of ArizonaTucsonUnited States
- University of Arizona Cancer CenterTucsonUnited States
| | - Julia M Morris
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
| | - Olga Alkhimenok
- Department of Molecular and Cellular Biology, The University of ArizonaTucsonUnited States
| | - Reeba P Varghese
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
- Cancer Biology Graduate Interdisciplinary Program, University of ArizonaTucsonUnited States
| | - Carly R Cabel
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
- Cancer Biology Graduate Interdisciplinary Program, University of ArizonaTucsonUnited States
| | - Nathan A Ellis
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
- University of Arizona Cancer CenterTucsonUnited States
| | - Jayati Chakrabarti
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
| | - Yana Zavros
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
- University of Arizona Cancer CenterTucsonUnited States
| | | | - Curtis A Thorne
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
- University of Arizona Cancer CenterTucsonUnited States
| | - Andrew L Paek
- Department of Molecular and Cellular Biology, The University of ArizonaTucsonUnited States
| |
Collapse
|
42
|
Chen L, Jiao T, Liu W, Luo Y, Wang J, Guo X, Tong X, Lin Z, Sun C, Wang K, He Y, Zhang Y, Xu H, Wang J, Zuo J, Ding Q, He S, Gonzalez FJ, Xie C. Hepatic cytochrome P450 8B1 and cholic acid potentiate intestinal epithelial injury in colitis by suppressing intestinal stem cell renewal. Cell Stem Cell 2022; 29:1366-1381.e9. [PMID: 36055192 PMCID: PMC10673678 DOI: 10.1016/j.stem.2022.08.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 06/08/2022] [Accepted: 08/11/2022] [Indexed: 11/03/2022]
Abstract
Although disrupted bile acid (BA) homeostasis is implicated in inflammatory bowel disease (IBD), the role of hepatic BA metabolism in the pathogenesis of colitis is poorly understood. Here, we found that cholic acid (CA) levels were increased in patients and mice. Cytochrome P450 8B1 (CYP8B1), which synthesizes CA, was induced in livers of colitic mice. CA-treated or liver Cyp8b1-overexpressing mice developed more severe colitis with compromised repair of the mucosal barrier, whereas Cyp8b1-knockout mice were resistant to colitis. Mechanistically, CA inhibited peroxisome proliferator-activated receptor alpha (PPARα), resulting in impeded fatty acid oxidation (FAO) and impaired Lgr5+ intestinal stem cell (ISC) renewal. A PPARα agonist restored FAO and improved Lgr5+ ISC function. Activation of the farnesoid X receptor (FXR) suppressed liver CYP8B1 expression and ameliorated colitis in mice. This study reveals a connection between the hepatic CYP8B1-CA axis and colitis via regulating intestinal epithelial regeneration, suggesting that BA-based strategies might be beneficial in IBD treatment.
Collapse
Affiliation(s)
- Li Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Tingying Jiao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Weiwei Liu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China; Department of Laboratory Medicine and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200070, P.R. China
| | - Yuhong Luo
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jue Wang
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Xiao Tong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Zemin Lin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Chuying Sun
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
| | - Kanglong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Yifan He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Yuwei Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Hualing Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
| | - Jiawen Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, P.R. China
| | - Jianping Zuo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Shijun He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| |
Collapse
|
43
|
Guo Y, Raev S, Kick MK, Raque M, Saif LJ, Vlasova AN. Rotavirus C Replication in Porcine Intestinal Enteroids Reveals Roles for Cellular Cholesterol and Sialic Acids. Viruses 2022; 14:v14081825. [PMID: 36016447 PMCID: PMC9416568 DOI: 10.3390/v14081825] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
Rotaviruses (RVs) are a significant cause of severe diarrheal illness in infants and young animals, including pigs. Group C rotavirus (RVC) is an emerging pathogen increasingly reported in pigs and humans worldwide, and is currently recognized as the major cause of gastroenteritis in neonatal piglets that results in substantial economic losses to the pork industry. However, little is known about RVC pathogenesis due to the lack of a robust cell culture system, with the exception of the RVC Cowden strain. Here, we evaluated the permissiveness of porcine crypt-derived 3D and 2D intestinal enteroid (PIE) culture systems for RVC infection. Differentiated 3D and 2D PIEs were infected with porcine RVC (PRVC) Cowden G1P[1], PRVC104 G3P[18], and PRVC143 G6P[5] virulent strains, and the virus replication was measured by qRT-PCR. Our results demonstrated that all RVC strains replicated in 2D-PIEs poorly, while 3D-PIEs supported a higher level of replication, suggesting that RVC selectively infects terminally differentiated enterocytes, which were less abundant in the 2D vs. 3D PIE cultures. While cellular receptors for RVC are unknown, target cell surface carbohydrates, including histo-blood-group antigens (HBGAs) and sialic acids (SAs), are believed to play a role in cell attachment/entry. The evaluation of the selective binding of RVCs to different HBGAs revealed that PRVC Cowden G1P[1] replicated to the highest titers in the HBGA-A PIEs, while PRVC104 or PRVC143 achieved the highest titers in the HBGA-H PIEs. Further, contrasting outcomes were observed following sialidase treatment (resulting in terminal SA removal), which significantly enhanced Cowden and RVC143 replication, but inhibited the growth of PRVC104. These observations suggest that different RVC strains may recognize terminal (PRVC104) as well as internal (Cowden and RVC143) SAs on gangliosides. Finally, several cell culture additives, such as diethylaminoethyl (DEAE)-dextran, cholesterol, and bile extract, were tested to establish if they could enhance RVC replication. We observed that only DEAE-dextran significantly enhanced RVC attachment, but it had no effect on RVC replication. Additionally, the depletion of cellular cholesterol by MβCD inhibited Cowden replication, while the restoration of the cellular cholesterol partially reversed the MβCD effects. These results suggest that cellular cholesterol plays an important role in the replication of the PRVC strain tested. Overall, our study has established a novel robust and physiologically relevant system to investigate RVC pathogenesis. We also generated novel, experimentally derived evidence regarding the role of host glycans, DEAE, and cholesterol in RVC replication, which is critical for the development of control strategies.
Collapse
Affiliation(s)
- Yusheng Guo
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Sergei Raev
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Maryssa K. Kick
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Molly Raque
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Linda J. Saif
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
- Correspondence:
| |
Collapse
|
44
|
Abstract
The vertebrate intestine experiences a range of intrinsically generated and external forces during both development and adult homeostasis. It is increasingly understood how the coordination of these forces shapes the intestine through organ-scale folding and epithelial organization into crypt-villus compartments. Moreover, accumulating evidence shows that several cell types in the adult intestine can sense and respond to forces to regulate key cellular processes underlying adult intestinal functions and self-renewal. In this way, transduction of forces may direct both intestinal homeostasis as well as adaptation to external stimuli, such as food ingestion or injury. In this review, we will discuss recent insights from complementary model systems into the force-dependent mechanisms that establish and maintain the unique architecture of the intestine, as well as its homeostatic regulation and function throughout adult life.
Collapse
|
45
|
Song C, Broadie K. Dysregulation of BMP, Wnt, and Insulin Signaling in Fragile X Syndrome. Front Cell Dev Biol 2022; 10:934662. [PMID: 35880195 PMCID: PMC9307498 DOI: 10.3389/fcell.2022.934662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/06/2022] [Indexed: 01/21/2023] Open
Abstract
Drosophila models of neurological disease contribute tremendously to research progress due to the high conservation of human disease genes, the powerful and sophisticated genetic toolkit, and the rapid generation time. Fragile X syndrome (FXS) is the most prevalent heritable cause of intellectual disability and autism spectrum disorders, and the Drosophila FXS disease model has been critical for the genetic screening discovery of new intercellular secretion mechanisms. Here, we focus on the roles of three major signaling pathways: BMP, Wnt, and insulin-like peptides. We present Drosophila FXS model defects compared to mouse models in stem cells/embryos, the glutamatergic neuromuscular junction (NMJ) synapse model, and the developing adult brain. All three of these secreted signaling pathways are strikingly altered in FXS disease models, giving new mechanistic insights into impaired cellular outcomes and neurological phenotypes. Drosophila provides a powerful genetic screening platform to expand understanding of these secretory mechanisms and to test cellular roles in both peripheral and central nervous systems. The studies demonstrate the importance of exploring broad genetic interactions and unexpected regulatory mechanisms. We discuss a number of research avenues to pursue BMP, Wnt, and insulin signaling in future FXS investigations and the development of potential therapeutics.
Collapse
Affiliation(s)
- Chunzhu Song
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
| | - Kendal Broadie
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
- Department of Cell and Developmental Biology, School of Medicine, Vanderbilt University, Nashville, TN, United States
- Kennedy Center for Research on Human Development, Nashville, TN, United States
- Vanderbilt Brain Institute, School of Medicine, Vanderbilt University and Medical Center, Nashville, TN, United States
| |
Collapse
|
46
|
Wang Y, Sims CE, Allbritton NL. Human 2D Crypt Model for Assaying Intestinal Stem Cell Proliferation and Differentiation. Anal Chem 2022; 94:9345-9354. [PMID: 35736812 DOI: 10.1021/acs.analchem.2c00905] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Intestine is a common site of adverse drug effects in clinical trials; thus, improved in vitro models for preclinical screening of pharmaceutical compounds are sought. A planar, self-renewing human intestinal monolayer platform based on primary adult gastrointestinal stem cells, termed the 2D crypt model, has been developed to screen for the effects of various compounds on the intestinal epithelium. The 2D crypt platform is based on a standard 12-well plate format and consists of cell culture inserts with a collagen film overlaying an impermeable film patterned with an array of micron-scale holes. This two-chamber format enables a gradient of growth factors to be applied such that the tissue self-organizes into spatially segregated stem and differentiated cell compartments. The patterned monolayer mimics a gut epithelium in possessing a stem cell niche, migrating proliferative and differentiated cells. Once established, the 2D crypts replicate many aspects of in vivo physiology, including cell migration, maturation, and apoptotic cell death. The planar geometry of the system simplifies dosing, sampling, and imaging during assay. An immunofluorescence-based assay was established to quantitatively assess cell density, proliferation, migration, viability, and the abundance and localization of postmitotic lineages as a function of time. The model was used to perform a small-scale screen of compounds, including signaling molecules, endogenous hormones/cytokines, and microbial metabolites, on tissue homeostasis. Hit compounds that significantly impacted proliferation and/or differentiation were readily identified. The 2D crypt platform represents a significant innovation in the development of microphysiological systems for emulating the gut epithelium for compound screens.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | | | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
47
|
Yu T, Cazares O, Tang AD, Kim HY, Wald T, Verma A, Liu Q, Barcellos-Hoff MH, Floor SN, Jung HS, Brooks AN, Klein OD. SRSF1 governs progenitor-specific alternative splicing to maintain adult epithelial tissue homeostasis and renewal. Dev Cell 2022; 57:624-637.e4. [PMID: 35202586 PMCID: PMC8974236 DOI: 10.1016/j.devcel.2022.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/04/2021] [Accepted: 01/18/2022] [Indexed: 12/30/2022]
Abstract
Alternative splicing generates distinct mRNA variants and is essential for development, homeostasis, and renewal. Proteins of the serine/arginine (SR)-rich splicing factor family are major splicing regulators that are broadly required for organ development as well as cell and organism viability. However, how these proteins support adult organ function remains largely unknown. Here, we used the continuously growing mouse incisor as a model to dissect the functions of the prototypical SR family protein SRSF1 during tissue homeostasis and renewal. We identified an SRSF1-governed alternative splicing network that is specifically required for dental proliferation and survival of progenitors but dispensable for the viability of differentiated cells. We also observed a similar progenitor-specific role of SRSF1 in the small intestinal epithelium, indicating a conserved function of SRSF1 across adult epithelial tissues. Thus, our findings define a regulatory mechanism by which SRSF1 specifically controls progenitor-specific alternative splicing events to support adult tissue homeostasis and renewal.
Collapse
Affiliation(s)
- Tingsheng Yu
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Oscar Cazares
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alison D Tang
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hyun-Yi Kim
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Tomas Wald
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Adya Verma
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Qi Liu
- Department of Radiation Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Stephen N Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Angela N Brooks
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
48
|
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: 10] [Impact Index Per Article: 3.3] [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
|
49
|
Xi W, Saleh J, Yamada A, Tomba C, Mercier B, Janel S, Dang T, Soleilhac M, Djemat A, Wu H, Romagnolo B, Lafont F, Mège RM, Chen Y, Delacour D. Modulation of designer biomimetic matrices for optimized differentiated intestinal epithelial cultures. Biomaterials 2022; 282:121380. [DOI: 10.1016/j.biomaterials.2022.121380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 01/07/2022] [Accepted: 01/16/2022] [Indexed: 12/22/2022]
|
50
|
Ren LL, Zhou JY, Liang SJ, Wang XQ. Impaired intestinal stem cell activity in ETEC infection: enterotoxins, cyclic nucleotides, and Wnt signaling. Arch Toxicol 2022; 96:1213-1225. [PMID: 35226135 DOI: 10.1007/s00204-021-03213-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/21/2021] [Indexed: 12/25/2022]
Abstract
Enterotoxigenic Escherichia coli (ETEC) in humans and animals colonizes the intestine and thereafter secrets heat-stable enterotoxin (ST) with or without heat-labile enterotoxin (LT), which triggers massive fluid and electrolyte secretion into the gut lumen. The crosstalk between the cyclic nucleotide-dependent protein kinase/cystic fibrosis transmembrane conductance regulator (cAMP or cGMP/CFTR) pathway involved in ETEC-induced diarrhea channels, and the canonical Wnt/β-catenin signaling pathway leads to changes in intestinal stem cell (ISC) fates, which are strongly associated with developmental disorders caused by diarrhea. We review how alterations in enterotoxin-activated ion channel pathways and the canonical Wnt/β-catenin signaling pathway can explain inhibited intestinal epithelial activity, characterize alterations in the crosstalk of cyclic nucleotides, and predict harmful effects on ISCs in targeted therapy. Besides, we discuss current deficits in the understanding of enterotoxin-intestinal epithelial cell activity relationships that should be considered when interpreting sequelae of diarrhea.
Collapse
Affiliation(s)
- Lu-Lu Ren
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, 510642, China
| | - Jia-Yi Zhou
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, 510642, China
| | - Shao-Jie Liang
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, 510642, China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, 510642, China.
| |
Collapse
|