1
|
Schuster T, Amoah A, Vollmer A, Marka G, Niemann J, Saçma M, Sakk V, Soller K, Vogel M, Grigoryan A, Wlaschek M, Scharffetter-Kochanek K, Mulaw M, Geiger H. Quantitative determination of the spatial distribution of components in single cells with CellDetail. Nat Commun 2024; 15:10250. [PMID: 39592623 PMCID: PMC11599593 DOI: 10.1038/s41467-024-54638-8] [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: 08/03/2023] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
The distribution of biomolecules within cells changes upon aging and diseases. To quantitatively determine the spatial distribution of components inside cells, we built the user-friendly open-source 3D-cell-image analysis platform Cell Detection and Analysis of Intensity Lounge (CellDetail). The algorithm within CellDetail is based on the concept of the dipole moment. CellDetail provides quantitative values for the distribution of the polarity proteins Cdc42 and Tubulin in young and aged hematopoietic stem cells (HSCs). Septin proteins form networks within cells that are critical for cell compartmentalization. We uncover a reduced level of organization of the Septin network within aged HSCs and within senescent human fibroblasts. Changes in the Septin network structure might therefore be a common feature of aging. The level of organization of the network of Septin proteins in aged HSCs can be restored to a youthful level by pharmacological attenuation of the activity of the small RhoGTPase Cdc42.
Collapse
Affiliation(s)
- Tanja Schuster
- Institute of Molecular Medicine, Ulm University, Ulm, Germany.
| | - Amanda Amoah
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
- Terry Fox Laboratory, BC Cancer Research Centre, Vancouver, BC, Canada
| | | | - Gina Marka
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Julian Niemann
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Mehmet Saçma
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Vadim Sakk
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Karin Soller
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Mona Vogel
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Ani Grigoryan
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany
| | | | - Medhanie Mulaw
- Unit for Single-Cell Genomics, Ulm University, Ulm, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Ulm, Germany.
| |
Collapse
|
2
|
Pasquier N, Jaulin F, Peglion F. Inverted apicobasal polarity in health and disease. J Cell Sci 2024; 137:jcs261659. [PMID: 38465512 PMCID: PMC10984280 DOI: 10.1242/jcs.261659] [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: 03/12/2024] Open
Abstract
Apicobasal epithelial polarity controls the functional properties of most organs. Thus, there has been extensive research on the molecular intricacies governing the establishment and maintenance of cell polarity. Whereas loss of apicobasal polarity is a well-documented phenomenon associated with multiple diseases, less is known regarding another type of apicobasal polarity alteration - the inversion of polarity. In this Review, we provide a unifying definition of inverted polarity and discuss multiple scenarios in mammalian systems and human health and disease in which apical and basolateral membrane domains are interchanged. This includes mammalian embryo implantation, monogenic diseases and dissemination of cancer cell clusters. For each example, the functional consequences of polarity inversion are assessed, revealing shared outcomes, including modifications in immune surveillance, altered drug sensitivity and changes in adhesions to neighboring cells. Finally, we highlight the molecular alterations associated with inverted apicobasal polarity and provide a molecular framework to connect these changes with the core cell polarity machinery and to explain roles of polarity inversion in health and disease. Based on the current state of the field, failure to respond to extracellular matrix (ECM) cues, increased cellular contractility and membrane trafficking defects are likely to account for most cases of inverted apicobasal polarity.
Collapse
Affiliation(s)
- Nicolas Pasquier
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
- Cell Adhesion and Cancer lab, University of Turku, FI-20520 Turku, Finland
| | - Fanny Jaulin
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
| | - Florent Peglion
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
| |
Collapse
|
3
|
Kang TH, Lee SI. Establishment of a chicken intestinal organoid culture system to assess deoxynivalenol-induced damage of the intestinal barrier function. J Anim Sci Biotechnol 2024; 15:30. [PMID: 38369477 PMCID: PMC10874546 DOI: 10.1186/s40104-023-00976-4] [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: 08/03/2023] [Accepted: 12/12/2023] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Deoxynivalenol (DON) is a mycotoxin that has received recognition worldwide because of its ability to cause growth delay, nutrient malabsorption, weight loss, emesis, and a reduction of feed intake in livestock. Since DON-contaminated feedstuff is absorbed in the gastrointestinal tract, we used chicken organoids to assess the DON-induced dysfunction of the small intestine. RESULTS We established a culture system using chicken organoids and characterized the organoids at passages 1 and 10. We confirmed the mRNA expression levels of various cell markers in the organoids, such as KI67, leucine-rich repeat containing G protein-coupled receptor 5 (Lgr5), mucin 2 (MUC2), chromogranin A (CHGA), cytokeratin 19 (CK19), lysozyme (LYZ), and microtubule-associated doublecortin-like kinase 1 (DCLK1), and compared the results to those of the small intestine. Our results showed that the organoids displayed functional similarities in permeability compared to the small intestine. DON damaged the tight junctions of the organoids, which resulted in increased permeability. CONCLUSIONS Our organoid culture displayed topological, genetic, and functional similarities with the small intestine cells. Based on these similarities, we confirmed that DON causes small intestine dysfunction. Chicken organoids offer a practical model for the research of harmful substances.
Collapse
Affiliation(s)
- Tae Hong Kang
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-Sangbuk-Do, 37224, Republic of Korea
| | - Sang In Lee
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeong-Sangbuk-Do, 37224, Republic of Korea.
- Research Institute for Innovative Animal Science, Kyungpook National University, 37224, Sangju, Gyeong-Sangbuk-Do, Republic of Korea.
| |
Collapse
|
4
|
Chen B, Slocombe RF, Georgy SR. Advances in organoid technology for veterinary disease modeling. Front Vet Sci 2023; 10:1234628. [PMID: 37920327 PMCID: PMC10618422 DOI: 10.3389/fvets.2023.1234628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
Organoids are in vitro organ-like structures that faithfully recapitulate many characteristics of a specific organ. During the past decades, major progress has been accomplished in establishing three-dimensional (3D) culture systems toward stem cell-derived organoids. As a significant technological breakthrough, these amazing 3D organoid constructs bridge the conventional 2D in vitro models and in vivo animal models and provide an unprecedented opportunity to investigate the complexities of veterinary diseases ranging from their pathogenesis to the prevention, therapy, or even future organ replacement strategies. In this review, we briefly discuss several definitions used in organoid research and highlight the currently known achievements in modeling veterinary diseases, including infectious and inflammatory diseases, cancers, and metabolic diseases. The applications of organoid technology in veterinary disease modeling are still in their infancy stage but the future is promising.
Collapse
Affiliation(s)
| | | | - Smitha Rose Georgy
- Section of Anatomic Pathology, Melbourne Veterinary School, Faculty of Science, University of Melbourne, Werribee, VIC, Australia
| |
Collapse
|
5
|
Naturale VF, Pickett MA, Feldman JL. Persistent cell contacts enable E-cadherin/HMR-1- and PAR-3-based symmetry breaking within a developing C. elegans epithelium. Dev Cell 2023; 58:1830-1846.e12. [PMID: 37552986 PMCID: PMC10592304 DOI: 10.1016/j.devcel.2023.07.008] [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: 08/12/2022] [Revised: 05/10/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Tissue-wide patterning is essential to multicellular development, requiring cells to individually generate polarity axes and coordinate them in space and time with neighbors. Using the C. elegans intestinal epithelium, we identified a patterning mechanism that is informed by cell contact lifetime asymmetry and executed via the scaffolding protein PAR-3 and the transmembrane protein E-cadherin/HMR-1. Intestinal cells break symmetry as PAR-3 and HMR-1 recruit apical determinants into punctate "local polarity complexes" (LPCs) at homotypic contacts. LPCs undergo an HMR-1-based migration to a common midline, thereby establishing tissue-wide polarity. Thus, symmetry breaking results from PAR-3-dependent intracellular polarization coupled to HMR-1-based tissue-level communication, which occurs through a non-adhesive signaling role for HMR-1. Differential lifetimes between homotypic and heterotypic cell contacts are created by neighbor exchanges and oriented divisions, patterning where LPCs perdure and thereby breaking symmetry. These cues offer a logical and likely conserved framework for how epithelia without obvious molecular asymmetries can polarize.
Collapse
Affiliation(s)
| | - Melissa A Pickett
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Jessica L Feldman
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
Saleem W, Ren X, Van Den Broeck W, Nauwynck H. Changes in intestinal morphology, number of mucus-producing cells and expression of coronavirus receptors APN, DPP4, ACE2 and TMPRSS2 in pigs with aging. Vet Res 2023; 54:34. [PMID: 37055856 PMCID: PMC10100624 DOI: 10.1186/s13567-023-01169-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/01/2023] [Indexed: 04/15/2023] Open
Abstract
Porcine enteric viral infections cause high morbidity and mortality in young piglets (<3 weeks). Later, these rates decrease with age. This age-dependent infectivity remains largely unexplored. This study investigated the changes in intestinal morphology, number of mucus-producing cells and expression level of coronavirus receptors in three age groups of pigs. Villus height and crypt depth increased with age from 3 days to 3 months in duodenum and ileum but not in mid-jejunum, where the villus height decreased from 580 µm at 3 days to 430 µm at 3 months. Enterocyte length-to-width ratio increased from 3 days to 3 months in all intestinal regions. The number of mucus-producing cells increased with age in the intestinal villi and crypts. The Brunner's glands of the duodenum contained the highest concentration of mucus-producing cells. The expression of coronavirus receptor APN was highest in the small intestinal villi at all ages. DPP4 expression slightly decreased over time in jejunum and ileum; it was highest in the ileal villi of 3-day-old piglets (70.2% of cells). ACE2 and TMPRSS2 positive cells increased with age in jejunal and ileal crypts and were particularly dominant in the ileal crypts (> 45% of cells). Except for the expression of DPP4 in the jejunum and ileum of young pigs, the expression pattern of the selected coronavirus receptors was very different and not correlated with the age-dependent susceptibility to viral infections. In contrast, the number of mucus-producing cells increased over time and may play an essential role in protecting enteric mucosae against intestinal viruses.
Collapse
Affiliation(s)
- Waqar Saleem
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium.
| | - Xiaolei Ren
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| |
Collapse
|
7
|
Liu Y, Zhang S, Deng H, Chen A, Chai L. Lead and copper influenced bile acid metabolism by changing intestinal microbiota and activating farnesoid X receptor in Bufo gargarizans. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 863:160849. [PMID: 36521604 DOI: 10.1016/j.scitotenv.2022.160849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
Lead (Pb) and copper (Cu) are ubiquitous metal contaminants and can pose a threat to ecosystem and human health. Bile acids have recently received considerable attention for their role in the maintenance of health. However, there were few studies on whether Pb and Cu affect bile acid metabolism in amphibians. In this study, a combination approach of histological analysis, targeted metabolomics, 16S rDNA sequencing and qPCR was used to explore the impacts of Pb, Cu and their mixture (Mix) on bile acid in Bufo gargarizans tadpoles. The results showed that Pb, Cu, and Mix resulted in intestinal damage and altered the bile acid profiles. Specifically, Pb and Mix exposure decreased total bile acid concentrations while increased toxic bile acid levels; in contrast, Cu exposure increased total bile acid levels. And hydrophilic bile acids were reduced in all treated tadpoles. Moreover, Pb and/or Cu changed the composition of intestinal microbiota, especially Clostridia, Bacteroides and Eubacterium involved in bile acid biotransformation. qPCR revealed that the decreased total bile acid concentrations in Pb- and Mix-treated tadpoles were most likely attributed to the activation of intestinal farnesoid X receptor (Fxr), which suppressed bile acid synthesis and reabsorption. While activated fxr in the Cu treatment group may be a regulatory mechanism in response to increased bile excretion, which is a detoxification route of tadpoles under Cu stress. Collectively, Pb, Cu and Mix changed bile acid profiles by affecting intestinal microbial composition and activating Fxr signaling. This study provided insight into the impacts of Pb and Cu on bile acid metabolism and contributed to the assessment of the potential ecotoxicity of heavy metals on amphibians.
Collapse
Affiliation(s)
- Yutian Liu
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Siliang Zhang
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Hongzhang Deng
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Aixia Chen
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an 710054, China.
| |
Collapse
|
8
|
Kakni P, Jutten B, Teixeira Oliveira Carvalho D, Penders J, Truckenmüller R, Habibovic P, Giselbrecht S. Hypoxia-tolerant apical-out intestinal organoids to model host-microbiome interactions. J Tissue Eng 2023; 14:20417314221149208. [PMID: 36699634 PMCID: PMC9869231 DOI: 10.1177/20417314221149208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/16/2022] [Indexed: 01/20/2023] Open
Abstract
Microbiome is an integral part of the gut and is essential for its proper function. Imbalances of the microbiota can be devastating and have been linked with several gastrointestinal conditions. Current gastrointestinal models do not fully reflect the in vivo situation. Thus, it is important to establish more advanced in vitro models to study host-microbiome/pathogen interactions. Here, we developed for the first time an apical-out human small intestinal organoid model in hypoxia, where the apical surface is directly accessible and exposed to a hypoxic environment. These organoids mimic the intestinal cell composition, structure and functions and provide easy access to the apical surface. Co-cultures with the anaerobic strains Lactobacillus casei and Bifidobacterium longum showed successful colonization and probiotic benefits on the organoids. These novel hypoxia-tolerant apical-out small intestinal organoids will pave the way for unraveling unknown mechanisms related to host-microbiome interactions and serve as a tool to develop microbiome-related probiotics and therapeutics.
Collapse
Affiliation(s)
- Panagiota Kakni
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Barry Jutten
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Daniel Teixeira Oliveira Carvalho
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM School of Nutrition and Translational Research in Metabolism, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands,Stefan Giselbrecht, Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, Maastricht 6229 ER, The Netherlands.
| |
Collapse
|
9
|
Kakni P, López-Iglesias C, Truckenmüller R, Habibović P, Giselbrecht S. PSC-derived intestinal organoids with apical-out orientation as a tool to study nutrient uptake, drug absorption and metabolism. Front Mol Biosci 2023; 10:1102209. [PMID: 36743212 PMCID: PMC9889654 DOI: 10.3389/fmolb.2023.1102209] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Intestinal organoids recapitulate many features of the in vivo gastrointestinal tract and have revolutionized in vitro studies of intestinal function and disease. However, the restricted accessibility of the apical surface of the organoids facing the central lumen (apical-in) limits studies related to nutrient uptake and drug absorption and metabolism. Here, we demonstrate that pluripotent stem cell (PSC)-derived intestinal organoids with reversed epithelial polarity (apical-out) can successfully recapitulate tissue-specific functions. In particular, these apical-out organoids show strong epithelial barrier formation with all the major junctional complexes, nutrient transport and active lipid metabolism. Furthermore, the organoids express drug-metabolizing enzymes and relevant apical and basolateral transporters. The scalable and robust generation of functional, apical-out intestinal organoids lays the foundation for a completely new range of organoid-based high-throughput/high-content in vitro applications in the fields of nutrition, metabolism and drug discovery.
Collapse
Affiliation(s)
- Panagiota Kakni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Maastricht, Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, Maastricht, Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Maastricht, Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Maastricht, Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Maastricht, Netherlands,*Correspondence: Stefan Giselbrecht,
| |
Collapse
|
10
|
Chew W, Lim YP, Lim WS, Chambers ES, Frost G, Wong SH, Ali Y. Gut-muscle crosstalk. A perspective on influence of microbes on muscle function. Front Med (Lausanne) 2023; 9:1065365. [PMID: 36698827 PMCID: PMC9868714 DOI: 10.3389/fmed.2022.1065365] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Our gastrointestinal system functions to digest and absorb ingested food, but it is also home to trillions of microbes that change across time, nutrition, lifestyle, and disease conditions. Largely commensals, these microbes are gaining prominence with regards to how they collectively affect the function of important metabolic organs, from the adipose tissues to the endocrine pancreas to the skeletal muscle. Muscle, as the biggest utilizer of ingested glucose and an important reservoir of body proteins, is intricately linked with homeostasis, and with important anabolic and catabolic functions, respectively. Herein, we provide a brief overview of how gut microbiota may influence muscle health and how various microbes may in turn be altered during certain muscle disease states. Specifically, we discuss recent experimental and clinical evidence in support for a role of gut-muscle crosstalk and include suggested underpinning molecular mechanisms that facilitate this crosstalk in health and diseased conditions. We end with a brief perspective on how exercise and pharmacological interventions may interface with the gut-muscle axis to improve muscle mass and function.
Collapse
Affiliation(s)
- Weixuan Chew
- Nutrition, Metabolism and Health Programme, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore,Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Yen Peng Lim
- Institute of Geriatrics and Active Aging, Tan Tock Seng Hospital, Singapore, Singapore,Department of Nutrition and Dietetics, Tan Tock Seng Hospital, National Healthcare Group, Singapore, Singapore
| | - Wee Shiong Lim
- Nutrition, Metabolism and Health Programme, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore,Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore,Institute of Geriatrics and Active Aging, Tan Tock Seng Hospital, Singapore, Singapore
| | - Edward S. Chambers
- Section for Nutrition Research, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Gary Frost
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
| | - Sunny Hei Wong
- Nutrition, Metabolism and Health Programme, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore,Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore,Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, National Healthcare Group, Singapore, Singapore
| | - Yusuf Ali
- Nutrition, Metabolism and Health Programme, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore,Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore,Singapore General Hospital, Singapore Eye Research Institute (SERI), Singapore, Singapore,Clinical Research Unit, Khoo Teck Puat Hospital, National Healthcare Group, Singapore, Singapore,*Correspondence: Yusuf Ali ✉
| |
Collapse
|
11
|
Naturale VF, Pickett MA, Feldman JL. Context matters: Lessons in epithelial polarity from the Caenorhabditis elegans intestine and other tissues. Curr Top Dev Biol 2023; 154:37-71. [PMID: 37100523 DOI: 10.1016/bs.ctdb.2023.02.007] [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: 04/28/2023]
Abstract
Epithelia are tissues with diverse morphologies and functions across metazoans, ranging from vast cell sheets encasing internal organs to internal tubes facilitating nutrient uptake, all of which require establishment of apical-basolateral polarity axes. While all epithelia tend to polarize the same components, how these components are deployed to drive polarization is largely context-dependent and likely shaped by tissue-specific differences in development and ultimate functions of polarizing primordia. The nematode Caenorhabditis elegans (C. elegans) offers exceptional imaging and genetic tools and possesses unique epithelia with well-described origins and roles, making it an excellent model to investigate polarity mechanisms. In this review, we highlight the interplay between epithelial polarization, development, and function by describing symmetry breaking and polarity establishment in a particularly well-characterized epithelium, the C. elegans intestine. We compare intestinal polarization to polarity programs in two other C. elegans epithelia, the pharynx and epidermis, correlating divergent mechanisms with tissue-specific differences in geometry, embryonic environment, and function. Together, we emphasize the importance of investigating polarization mechanisms against the backdrop of tissue-specific contexts, while also underscoring the benefits of cross-tissue comparisons of polarity.
Collapse
Affiliation(s)
- Victor F Naturale
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Melissa A Pickett
- Department of Biology, Stanford University, Stanford, CA, United States; Department of Biological Sciences, San José State University, San José, CA, United States
| | - Jessica L Feldman
- Department of Biology, Stanford University, Stanford, CA, United States.
| |
Collapse
|
12
|
Wang Q, Guo F, Jin Y, Ma Y. Applications of human organoids in the personalized treatment for digestive diseases. Signal Transduct Target Ther 2022; 7:336. [PMID: 36167824 PMCID: PMC9513303 DOI: 10.1038/s41392-022-01194-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/09/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Digestive system diseases arise primarily through the interplay of genetic and environmental influences; there is an urgent need in elucidating the pathogenic mechanisms of these diseases and deploy personalized treatments. Traditional and long-established model systems rarely reproduce either tissue complexity or human physiology faithfully; these shortcomings underscore the need for better models. Organoids represent a promising research model, helping us gain a more profound understanding of the digestive organs; this model can also be used to provide patients with precise and individualized treatment and to build rapid in vitro test models for drug screening or gene/cell therapy, linking basic research with clinical treatment. Over the past few decades, the use of organoids has led to an advanced understanding of the composition of each digestive organ and has facilitated disease modeling, chemotherapy dose prediction, CRISPR-Cas9 genetic intervention, high-throughput drug screening, and identification of SARS-CoV-2 targets, pathogenic infection. However, the existing organoids of the digestive system mainly include the epithelial system. In order to reveal the pathogenic mechanism of digestive diseases, it is necessary to establish a completer and more physiological organoid model. Combining organoids and advanced techniques to test individualized treatments of different formulations is a promising approach that requires further exploration. This review highlights the advancements in the field of organoid technology from the perspectives of disease modeling and personalized therapy.
Collapse
Affiliation(s)
- Qinying Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fanying Guo
- School of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yutao Jin
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Wang H, Chen H, Lin Y, Wang G, Luo Y, Li X, Wang M, Huai M, Li L, Barri A. Butyrate Glycerides Protect against Intestinal Inflammation and Barrier Dysfunction in Mice. Nutrients 2022; 14:3991. [PMID: 36235644 PMCID: PMC9570839 DOI: 10.3390/nu14193991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 12/29/2022] Open
Abstract
This study investigates the attenuating effects of butyrate glycerides (BG) on intestinal inflammatory responses and barrier dysfunction induced by LPS stimulation. An initial dose-response test was carried out to identify the optimal dose of BG for further testing. The mice were given intragastric administration of BG at different doses followed by lipopolysaccharide (LPS) intraperitoneal injection. The small intestinal morphology and cytokine mRNA expression were measured. With 1.5 g/kg BW BG administration, it was possible to alleviate the injury of duodenal morphology, attenuate ileum villus height reduction and promote IL-10 mRNA expression. Therefore, the optimal dosage of 1.5 g/kg BW BG was selected for the main experiment. The ultrastructure image of jejunum and ileum epithelial cells, mRNA expression, the level of cytokine and immunofluorescence in the ileum were analyzed. The results showed that BG maintain the ileac brush border, tight junction structures and protein expression. BG attenuated the increased inflammatory cytokines, TLR4 and JNK mRNA expression. Taken together, 1.5 g/kg BW BG administration maintained intestinal barrier function and reduced intestinal and body inflammation responses induced by LPS in mice. The mechanism by which BG alleviated intestinal inflammatory response and maintained intestinal barrier function may be related to the JNK signaling pathway.
Collapse
Affiliation(s)
- Haidong Wang
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Haohan Chen
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yueying Lin
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Geng Wang
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanqiu Luo
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Li
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minqi Wang
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | | | - Lily Li
- BASF SEA Pte Ltd., Singapore 038987, Singapore
| | | |
Collapse
|
14
|
Kang TH, Kang KS, Lee SI. Deoxynivalenol Induces Apoptosis via FOXO3a-Signaling Pathway in Small-Intestinal Cells in Pig. TOXICS 2022; 10:toxics10090535. [PMID: 36136500 PMCID: PMC9503759 DOI: 10.3390/toxics10090535] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 05/12/2023]
Abstract
Deoxynivalenol (DON) is a mycotoxin that is found in feed ingredients derived from grains such as corn and wheat. Consumption of DON-contaminated feed has been shown to cause damage to the intestine, kidneys, and liver. However, the molecular mechanism by which DON exerts its effect in the small intestine is not completely understood. As a result, we profiled gene expression in intestinal epithelial cells treated with DON and examined the molecular function in vitro. We hypothesized that DON could induce apoptosis via the FOXO3a-signaling pathway in intestinal epithelial cells based on these findings. DON induced the apoptosis and the translocation of FOXO3a into the nucleus. Moreover, the inhibiting of FOXO3a alleviated the apoptosis and expression of apoptosis-related genes (TRAL, BCL-6, CASP8, and CASP3). ERK1/2 inhibitor treatment suppressed the translocation of FOXO3a into the nucleus. Our discovery suggests that DON induces apoptosis in intestinal epithelial cells through the FOXO3a-signaling pathway.
Collapse
Affiliation(s)
- Tae Hong Kang
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Korea
| | - Kyung Soo Kang
- Department of Bio Life Sciences, Shingu College, Seongnam-si 13174, Korea
| | - Sang In Lee
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Korea
- Correspondence: ; Tel.: +82-010-4183-5831
| |
Collapse
|
15
|
Apical-basal polarity and the control of epithelial form and function. Nat Rev Mol Cell Biol 2022; 23:559-577. [PMID: 35440694 DOI: 10.1038/s41580-022-00465-y] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 02/02/2023]
Abstract
Epithelial cells are the most common cell type in all animals, forming the sheets and tubes that compose most organs and tissues. Apical-basal polarity is essential for epithelial cell form and function, as it determines the localization of the adhesion molecules that hold the cells together laterally and the occluding junctions that act as barriers to paracellular diffusion. Polarity must also target the secretion of specific cargoes to the apical, lateral or basal membranes and organize the cytoskeleton and internal architecture of the cell. Apical-basal polarity in many cells is established by conserved polarity factors that define the apical (Crumbs, Stardust/PALS1, aPKC, PAR-6 and CDC42), junctional (PAR-3) and lateral (Scribble, DLG, LGL, Yurt and RhoGAP19D) domains, although recent evidence indicates that not all epithelia polarize by the same mechanism. Research has begun to reveal the dynamic interactions between polarity factors and how they contribute to polarity establishment and maintenance. Elucidating these mechanisms is essential to better understand the roles of apical-basal polarity in morphogenesis and how defects in polarity contribute to diseases such as cancer.
Collapse
|
16
|
Gut microbiota alternation under the intestinal epithelium-specific knockout of mouse Piga gene. Sci Rep 2022; 12:10812. [PMID: 35752737 PMCID: PMC9233684 DOI: 10.1038/s41598-022-15150-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 04/22/2022] [Indexed: 11/08/2022] Open
Abstract
Crosstalk between the gut microbiota and intestinal epithelium shapes the gut environment and profoundly influences the intestinal immune homeostasis. Glycosylphosphatidylinositol anchored proteins (GPI – APs) contribute to a variety of gut-associated immune functions, including microbial surveillance and defense, and epithelial cell polarity. Properly polarised epithelial cells are essential for the establishment of the barrier function of gut epithelia. The Piga gene is one among seven genes that encode for an enzyme which is involved in the first step of GPI-anchor biosynthesis. This is the first study reporting a knockout of the intestinal epithelial cell-specific Piga gene (Piga-/-) and its association with the gut microbiota in mice using a whole metagenome shotgun-based sequencing approach. An overall reduced microbiota diversity has been observed in the Piga-/- group as compared to the control group (ANOVA p = 0.34). The taxonomic biomarkers, namely: Gammaproteobacteria (class), Enterobacterales (order), Enterobacteriaceae (family), Escherichia (genus), Proteus (genus) and Escherichia coli (species), increased more in the Piga-/- mice as compared to in the control group. Further, the pathogenic E. coli strains, namely E. coli O157:H7 str. EDL 933 (EHEC), E. coli CFT073 (UPEC) and E. coli 536 (UPEC), were found in the Piga-/- mice which also harbored virulence factor transporters. In addition, the taxa responsible for short chain fatty acid production were decreased in the Piga-/- group. The Piga-/- mice gut harbored an increased number of microbial functions responsible for the survival of pathogens in the inflamed gut environment. Our observations clearly indicate that the Piga-/- mice gut might have an overall enhancement in pathogenic behaviour and reduced capabilities beneficial to health.
Collapse
|
17
|
Abstract
We study microvilli of Caenorhabditis elegans larvae and mouse intestinal tissues by combining high-pressure freezing, cryo-focused ion-beam milling, cryo-electron tomography, and subtomogram averaging. We find that many radial nanometer bristles, referred to as nanobristles, project from the lateral surface of nematode and mouse microvilli. The C. elegans nanobristles are 37.5 nm long. We show that nanobristle formation requires a protocadherin family protein, CDH-8, in C. elegans. The loss of nanobristles in cdh-8 mutants slows down animal growth and ectopically increases the number of Y-shaped microvilli, the putative intermediate structures if microvilli split from their tips. Our results reveal a potential role of nanobristles in separating microvilli and suggest that microvilli division may help generate nascent microvilli with uniformity. Microvilli are actin-bundle-supported membrane protrusions essential for absorption, secretion, and sensation. Microvilli defects cause gastrointestinal disorders; however, mechanisms controlling microvilli formation and organization remain unresolved. Here, we study microvilli by vitrifying the Caenorhabditis elegans larvae and mouse intestinal tissues with high-pressure freezing, thinning them with cryo-focused ion-beam milling, followed by cryo-electron tomography and subtomogram averaging. We find that many radial nanometer bristles referred to as nanobristles project from the lateral surface of nematode and mouse microvilli. The C. elegans nanobristles are 37.5 nm long and 4.5 nm wide. Nanobristle formation requires a protocadherin family protein, CDH-8, in C. elegans. The loss of nanobristles in cdh-8 mutants slows down animal growth and ectopically increases the number of Y-shaped microvilli, the putative intermediate structures if microvilli split from tips. Our results reveal a potential role of nanobristles in separating microvilli and suggest that microvilli division may help generate nascent microvilli with uniformity.
Collapse
|
18
|
Wang J, Zhu Y, Tian S, Shi Q, Yang H, Wang J, Zhu W. Effects of Protein Restriction and Succedent Realimentation on Jejunal Function and Bacterial Composition of Different Colonic Niches in Weaned Piglets. Front Vet Sci 2022; 9:877130. [PMID: 35591867 PMCID: PMC9111176 DOI: 10.3389/fvets.2022.877130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 12/03/2022] Open
Abstract
Recent studies have proved that protein succedent realimentation could rescue the loss of growth performance in weaning piglets caused by a prior protein restriction. However, how the protein restriction and succedent realimentation influence the jejunal function and bacterial composition of different colonic niches microbiota in weaning piglets needs a further investigation. After protein succedent realimentation, we found that the treatment group (TRE) piglets had a higher IGF-1 content and IGF-1R gene expression level in jejunal mucosa than the control group (CON) piglets. The ZO-1 gene expression level was up-regulated in the jejunal mucosa of TRE piglets during protein restriction and succedent realimentation, while the jejunal permeability of TRE piglets was only decreased after protein succedent realimentation. In addition, we found that protein restriction and succedent realimentation increased the gene expression of Pept-1 and the fecal apparent digestibility of crude protein in TRE piglets, but decreased the fecal nitrogen content. After 16S rRNA MiSeq sequencing of bacteria in different colonic niches (mucosa and digesta), TRE piglets had a higher relative abundance of beneficial bacteria and a lower relative abundance of potential pathogens than CON piglets in different colonic niches after protein restriction and succedent realimentation. Our data showed that protein restriction and succedent realimentation decreased the concentrations of branch chain fatty acids and ammonia-N in the colon of TRE piglets. In addition, protein succedent realimentation increased the concentration of total short chain fatty acids in the colon of TRE piglets. All these findings demonstrated that the strategy of protein restriction and succedent realimentation is an effective way to improve intestinal health of weaning piglets, and provided new insights into the nutrition management of piglets during the weaning period.
Collapse
Affiliation(s)
- Jue Wang
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Yizhi Zhu
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shiyi Tian
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qing Shi
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Huairong Yang
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jing Wang
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Jing Wang
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
19
|
Ao J, Liu Y, Tang W, Zhang J. Bisphenol S exposure induces intestinal inflammation: An integrated metabolomic and transcriptomic study. CHEMOSPHERE 2022; 292:133510. [PMID: 34979203 DOI: 10.1016/j.chemosphere.2021.133510] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 06/14/2023]
Abstract
As a typical substitute for bisphenol A (BPA), bisphenol S (BPS) is raising concerns due to the potential adverse effects on human health. Limit evidence is available to understand the toxicity of BPS to the digestive system, especially for intestine. In this study, we aimed to investigate the potential effects and underlying mechanisms of BPS exposure on human colon mucosal epithelial cells (NCM460). Our results showed that BPS exposure significantly increased the production of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ) and interleukin-17A (IL-17A). The tight junctions of the cells has been destroyed by BPS exposure, which was characterized by a down-regulation of the tight junction proteins (Claudin1 and zonula occluden 1 (ZO1)). A multi-omics study explored the underlying mechanisms based on the metabolomic and transcriptomic responses. A variety of neurotransmitters increased significantly after exposure to BPS. The top enriched pathway was "glutamatergic synapse", which was activated by BPS exposure, resulting in the up-regulation of l-glutamine. Links were observed among the altered metabolites, genes and cytokines. Our results indicate that exposure to BPS may disturb the balance of gut-brain axis, leading to the production of inflammatory cytokines and the destruction of tight junction in NCM460 cells. It provides new clue for the development of intestinal inflammation in terms of the environmental pollutants.
Collapse
Affiliation(s)
- Junjie Ao
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Yongjie Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Weifeng Tang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; School of Public Health, Shanghai Jiao Tong University, Shanghai, 200025, China
| |
Collapse
|
20
|
GLP1 Exerts Paracrine Activity in the Intestinal Lumen of Human Colon. Int J Mol Sci 2022; 23:ijms23073523. [PMID: 35408884 PMCID: PMC8998470 DOI: 10.3390/ijms23073523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
GLP1 produced in the upper part of the gut is released after food intake and acts by activating insulin secretion, but the role of GLP1 in the colon, where it is predominantly produced, remains unknown. Here we characterized the apical versus basolateral secretion of GLP1 and PYY and the paracrine mechanisms of action of these enterohormones in the human colon. We stimulated human colon tissue in different ex vivo models with meat peptone and we used immunofluorescence to study the presence of canonical and non-canonical receptors of GLP1. We found that PYY and GLP1 are secreted mainly at the gut lumen in unstimulated and stimulated conditions. We detected DPP4 activity and found that GLP1R and GCGR are widely expressed in the human colon epithelium. Unlike GLP1R, GCGR is not expressed in the lamina propria, but it is located in the crypts of Lieberkühn. We detected GLP1R expression in human colon cell culture models. We show that the apical secretion of PYY and GLP1 occurs in humans, and we provide evidence that GLP1 has a potential direct paracrine function through the expression of its receptors in the colon epithelium, opening new therapeutic perspectives in the use of enterohormones analogues in metabolic pathologies.
Collapse
|
21
|
Afzal MW, Duan K, Zhang Y, Gao Y, Qin B, Wang G, Lei L, Tang H, Guo Y. A rhodol-based fluorescent probe with a pair of hydrophilic and rotatable wings for sensitively monitoring intracellular polarity. Chem Asian J 2022; 17:e202200063. [PMID: 35191194 DOI: 10.1002/asia.202200063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/21/2022] [Indexed: 11/06/2022]
Abstract
Cell polarity, as a vital intracellular microenvironment characteristic, has immense effects on numerous pathological and biological processes. Therefore, the tracking of polarity variations is highly essential to explore the role and mechanism of the polarity in pathophysiological processes. Herein, we designed and synthesized a novel rhodol-based fluorescent probe RDS sensitive to polarity by introducing a bis(2-hydroxyethylthio)methyl group, like a pair of hydrophilic and rotatable wings, into the rhodol skeleton. This unique design makes RDS adopt the colorless and non-fluorescent spirocyclic form in low polarity medium while the colored and fluorescent ring-open form in high polarity system, resulting in a positive-correlation response of fluorescence intension to polarity. Importantly, RDS was successfully applied to monitor the polarity changes in living cells including cancer cells, healthy cells and senescent healthy cells, visualizing that the polarity of cancer cells is lower than that of healthy cells in which the more senescent ones have higher polarity.
Collapse
Affiliation(s)
| | - Kaixuan Duan
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Yanhui Zhang
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Ying Gao
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Bo Qin
- Xi'an University of Posts and Telecommunications, School of Automation, CHINA
| | - Guangwei Wang
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Lin Lei
- Northwest University, College of Chemistry and Materials Science, CHINA
| | - Haoyang Tang
- Xi'an University of Posts and Telecommunications, School of Automation, CHINA
| | - Yuan Guo
- Northwest University, College of Chemistry and Materials Science, 1 Xuefu Road, Chang'an district, 710127, Xi'an, CHINA
| |
Collapse
|
22
|
Almasi S, Jasmin BJ. The multifunctional RNA-binding protein Staufen1: an emerging regulator of oncogenesis through its various roles in key cellular events. Cell Mol Life Sci 2021; 78:7145-7160. [PMID: 34633481 PMCID: PMC8629789 DOI: 10.1007/s00018-021-03965-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
The double-stranded multifunctional RNA-binding protein (dsRBP) Staufen was initially discovered in insects as a regulator of mRNA localization. Later, its mammalian orthologs have been described in different organisms, including humans. Two human orthologues of Staufen, named Staufen1 (STAU1) and Staufen2 (STAU2), share some structural and functional similarities. However, given their different spatio-temporal expression patterns, each of these orthologues plays distinct roles in cells. In the current review, we focus on the role of STAU1 in cell functions and cancer development. Since its discovery, STAU1 has mostly been studied for its involvement in various aspects of RNA metabolism. Given the pivotal role of RNA metabolism within cells, recent studies have explored the mechanistic impact of STAU1 in a wide variety of cell functions ranging from cell growth to cell death, as well as in various disease states. In particular, there has been increasing attention on the role of STAU1 in neuromuscular disorders, neurodegeneration, and cancer. Here, we provide an overview of the current knowledge on the role of STAU1 in RNA metabolism and cell functions. We also highlight the link between STAU1-mediated control of cellular functions and cancer development, progression, and treatment. Hence, our review emphasizes the potential of STAU1 as a novel biomarker and therapeutic target for cancer diagnosis and treatment, respectively.
Collapse
Affiliation(s)
- Shekoufeh Almasi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada.
| |
Collapse
|
23
|
Shen Y, Zhang N, Tian J, Xin G, Liu L, Sun X, Li B. Advanced approaches for improving bioavailability and controlled release of anthocyanins. J Control Release 2021; 341:285-299. [PMID: 34822910 DOI: 10.1016/j.jconrel.2021.11.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022]
Abstract
Anthocyanins are a group of phytochemicals responsible for the purple or red color of plants. Additionally, they are recognized to have health promoting functions including anti-cardiovascular, anti-thrombotic, anti-diabetic, antimicrobial, neuroprotective, and visual protective effect as well as anti-cancer activities. Thus, consumption of anthocyanin supplement or anthocyanin-rich foods has been recommended to prevent the risk of development of chronic diseases. However, the low stability and bioavailability of anthocyanins limit the efficacy and distribution of anthocyanins in human body. Thus, strategies to achieve target site-local delivery with good bioavailability and controlled release rate are necessary. This review introduced and discussed the latest advanced techniques of designing lipid-based, polysaccharide-based and protein-based complexes, nano-encapsulation and exosome to overcome the limitation of anthocyanins. The improved bioavailability and controlled release of anthocyanins have great significance for gastrointestinal tract absorption, transepithelial transportation and cellular uptake. The techniques of applying different biocompatible materials and modifying the solubility of anthocyanins complex could achieve target site-local delivery with negligible degradation and good bioavailability in human body.
Collapse
Affiliation(s)
- Yixiao Shen
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Ning Zhang
- College of Horticulture Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Horticulture Germplasm Excavation and Innovative Utilization Qinhuangdao, Hebei, China
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Guang Xin
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Ling Liu
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Xiyun Sun
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China.
| |
Collapse
|
24
|
Yan W, Xiao Y, Zhang Y, Tao Y, Cao Y, Liu K, Cai W, Wang Y. Monogenic mutations in four cases of neonatal-onset watery diarrhea and a mutation review in East Asia. Orphanet J Rare Dis 2021; 16:383. [PMID: 34503561 PMCID: PMC8427875 DOI: 10.1186/s13023-021-01995-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/25/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Infants with neonatal-onset diarrhea present with intractable diarrhea in the first few weeks of life. A monogenic mutation is one of the disease etiologies and the use of next-generation sequencing (NGS) has made it possible to screen patients for their mutations. MAIN BODY We retrospectively reviewed the clinical data of four children from unrelated families, who presented with neonatal-onset, chronic, watery, non-bloody diarrhea. After genetic whole-exome sequencing, novel mutations were identified in the EPCAM gene of two children. Congenital chloride diarrhea was diagnosed in one case, which was associated with an SLC26A3 mutation, in which the patient presented with watery diarrhea, malnutrition, and hypochloremic alkalosis. Patient 4 was diagnosed with microvillus inclusion disease and possessed novel compound heterozygous mutations in the MYO5B gene. A review of the genetic variants of SLC26A3 reported in East Asia revealed that c.269_270 dupAA (p.G91Kfs*3) is the most frequent SLC26A3 mutation in China, compared with c.2063-1 G > T in Japan and Korea. EPCAM and MYO5B genetic variants were only sporadically reported in East Asia. CONCLUSION This study expands our knowledge of the clinical manifestations and molecular genetics of neonatal-onset watery diarrhea. Early diagnosis could be achieved by genomic analysis in those infants whose histology features are not typical. The discovery of four novel mutations in the EPCAM gene and two novel mutations in the MYO5B gene provides further etiological evidence for the association of genetic mutations with neonatal-onset diarrhea. To date, c.269_270 dupAA is the most frequent SLC26A3 mutation in China.
Collapse
Affiliation(s)
- Weihui Yan
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China
| | - Yongtao Xiao
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China
| | - Yunyi Zhang
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China
| | - Yijing Tao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Cao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Kunhui Liu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wei Cai
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China.
- Department of Pediatric Surgery, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China.
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China.
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China.
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
25
|
Intestinal immunoregulation: lessons from human mendelian diseases. Mucosal Immunol 2021; 14:1017-1037. [PMID: 33859369 DOI: 10.1038/s41385-021-00398-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023]
Abstract
The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.
Collapse
|
26
|
Hebbar S, Knust E. Reactive oxygen species (ROS) constitute an additional player in regulating epithelial development. Bioessays 2021; 43:e2100096. [PMID: 34260754 DOI: 10.1002/bies.202100096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules produced in cells. So far, they have mostly been connected to diseases and pathological conditions. More recent results revealed a somewhat unexpected role of ROS in control of developmental processes. In this review, we elaborate on ROS in development, focussing on their connection to epithelial tissue morphogenesis. After briefly summarising unique characteristics of epithelial cells, we present some characteristic features of ROS species, their production and targets, with a focus on proteins important for epithelial development and function. Finally, we provide examples of regulation of epithelial morphogenesis by ROS, and also of developmental genes that regulate the overall redox status. We conclude by discussing future avenues of research that will further elucidate ROS regulation in epithelial development.
Collapse
Affiliation(s)
- Sarita Hebbar
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
27
|
Bandyopadhyay S, Bonder E, Gao N. Tight Junction Proteins Join the Local Force for Bulk Endocytosis and Microvillus Inclusion. Cell Mol Gastroenterol Hepatol 2021; 12:348-349. [PMID: 33757764 PMCID: PMC8257456 DOI: 10.1016/j.jcmgh.2021.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 12/10/2022]
Affiliation(s)
| | | | - Nan Gao
- Correspondence Address correspondence to: Nan Gao, PhD, Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102.
| |
Collapse
|
28
|
Redhai S, Boutros M. The Role of Organelles in Intestinal Function, Physiology, and Disease. Trends Cell Biol 2021; 31:485-499. [PMID: 33551307 DOI: 10.1016/j.tcb.2021.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023]
Abstract
The intestine maintains homeostasis by coordinating internal biological processes to adjust to fluctuating external conditions. The intestinal epithelium is continuously renewed and comprises multiple cell types, including absorptive cells, secretory cells, and resident stem cells. An important feature of this organ is its ability to coordinate many processes including cell proliferation, differentiation, regeneration, damage/stress response, immune activity, feeding behavior, and age-related changes by using conserved signaling pathways. However, the subcellular spatial organization of these signaling events and the organelles involved has only recently been studied in detail. Here we discuss how organelles of intestinal cells serve to initiate, mediate, and terminate signals, that are vital for homeostasis.
Collapse
Affiliation(s)
- Siamak Redhai
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany.
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany.
| |
Collapse
|
29
|
Viral PDZ Binding Motifs Influence Cell Behavior Through the Interaction with Cellular Proteins Containing PDZ Domains. Methods Mol Biol 2021; 2256:217-236. [PMID: 34014525 DOI: 10.1007/978-1-0716-1166-1_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Viruses have evolved to interact with their hosts. Some viruses such as human papilloma virus, dengue virus, SARS-CoV, or influenza virus encode proteins including a PBM that interact with cellular proteins containing PDZ domains. There are more than 400 cellular protein isoforms with these domains in the human genome, indicating that viral PBMs have a high potential to influence the behavior of the cell. In this review we analyze the most relevant cellular processes known to be affected by viral PBM-cellular PDZ interactions including the establishment of cell-cell interactions and cell polarity, the regulation of cell survival and apoptosis and the activation of the immune system. Special attention has been provided to coronavirus PBM conservation throughout evolution and to the role of the PBMs of human coronaviruses SARS-CoV and MERS-CoV in pathogenesis.
Collapse
|
30
|
Levic DS, Ryan S, Marjoram L, Honeycutt J, Bagwell J, Bagnat M. Distinct roles for luminal acidification in apical protein sorting and trafficking in zebrafish. J Cell Biol 2020; 219:133852. [PMID: 32328632 PMCID: PMC7147097 DOI: 10.1083/jcb.201908225] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial cell physiology critically depends on the asymmetric distribution of channels and transporters. However, the mechanisms targeting membrane proteins to the apical surface are still poorly understood. Here, we performed a visual forward genetic screen in the zebrafish intestine and identified mutants with defective apical targeting of membrane proteins. One of these mutants, affecting the vacuolar H+-ATPase gene atp6ap1b, revealed specific requirements for luminal acidification in apical, but not basolateral, membrane protein sorting and transport. Using a low temperature block assay combined with genetic and pharmacologic perturbation of luminal pH, we monitored transport of newly synthesized membrane proteins from the TGN to apical membrane in live zebrafish. We show that vacuolar H+-ATPase activity regulates sorting of O-glycosylated proteins at the TGN, as well as Rab8-dependent post-Golgi trafficking of different classes of apical membrane proteins. Thus, luminal acidification plays distinct and specific roles in apical membrane biogenesis.
Collapse
Affiliation(s)
| | - Sean Ryan
- Department of Cell Biology, Duke University, Durham, NC
| | | | | | | | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC
| |
Collapse
|
31
|
Bajinka O, Darboe A, Tan Y, Abdelhalim KA, Cham LB. Gut microbiota and the human gut physiological changes. ANN MICROBIOL 2020. [DOI: 10.1186/s13213-020-01608-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
The human gut can be colonized by number of microorganisms. The most studied are bacteria, which changes from birth to newborn born into adult-like gut microbiota. Much is known about the effects of dietary, medications, and lifestyles on the bacterial composition. However, the host physiological changes influencing the gut microbiota, the immediate consequences, and the possible gut microbiota therapy are not studied at length. This review is based profoundly on animal model studies through experimentation and some human clinical trials for the past 20 years.
Forward
The physiological factors studied to influences gut microbiota are bacterial mucosal receptors, mucin glycosylation, mucus, epithelial microvilli, and tight junction. Host secretions and immune response such as immunity, secretory A (sIgA), inflammasome, innate immunity, immune response, glycans, bile acids, peristalsis, microRNA, and adhesion to intestinal glycans are as well found to confer variety of alterations on gut microbial flora.
Conclusion
Despite the resilience of the gut microbiota in response to changes, chain of events causes the imbalance microbiota. Increased pro-inflammatory potential with the help of cell barriers, host secretions, and immune response mediate gut recovery.
Collapse
|
32
|
van Rijn JM, Werner L, Aydemir Y, Spronck JM, Pode-Shakked B, van Hoesel M, Shimshoni E, Polak-Charcon S, Talmi L, Eren M, Weiss B, H.J. Houwen R, Barshack I, Somech R, Nieuwenhuis EE, Sagi I, Raas-Rothschild A, Middendorp S, Shouval DS. Enhanced Collagen Deposition in the Duodenum of Patients with Hyaline Fibromatosis Syndrome and Protein Losing Enteropathy. Int J Mol Sci 2020; 21:E8200. [PMID: 33147779 PMCID: PMC7662532 DOI: 10.3390/ijms21218200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/25/2022] Open
Abstract
Hyaline fibromatosis syndrome (HFS), resulting from ANTXR2 mutations, is an ultra-rare disease that causes intestinal lymphangiectasia and protein-losing enteropathy (PLE). The mechanisms leading to the gastrointestinal phenotype in these patients are not well defined. We present two patients with congenital diarrhea, severe PLE and unique clinical features resulting from deleterious ANTXR2 mutations. Intestinal organoids were generated from one of the patients, along with CRISPR-Cas9 ANTXR2 knockout, and compared with organoids from two healthy controls. The ANTXR2-deficient organoids displayed normal growth and polarity, compared to controls. Using an anthrax-toxin assay we showed that the c.155C>T mutation causes loss-of-function of ANTXR2 protein. An intrinsic defect of monolayer formation in patient-derived or ANTXR2KO organoids was not apparent, suggesting normal epithelial function. However, electron microscopy and second harmonic generation imaging showed abnormal collagen deposition in duodenal samples of these patients. Specifically, collagen VI, which is known to bind ANTXR2, was highly expressed in the duodenum of these patients. In conclusion, despite resistance to anthrax-toxin, epithelial cell function, and specifically monolayer formation, is intact in patients with HFS. Nevertheless, loss of ANTXR2-mediated signaling leads to collagen VI accumulation in the duodenum and abnormal extracellular matrix composition, which likely plays a role in development of PLE.
Collapse
Affiliation(s)
- Jorik M. van Rijn
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), 3584 CT Utrecht, The Netherlands; (J.M.v.R.); (J.M.A.S.); (M.v.H.); (R.H.J.H.); (E.E.S.N.)
- Regenerative Medicine Center, UMCU, UU, 3584 CT Utrecht, The Netherlands
| | - Lael Werner
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel; (L.W.); (B.W.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
| | - Yusuf Aydemir
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir 26040, Turkey; (Y.A.); (M.E.)
| | - Joey M.A. Spronck
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), 3584 CT Utrecht, The Netherlands; (J.M.v.R.); (J.M.A.S.); (M.v.H.); (R.H.J.H.); (E.E.S.N.)
- Regenerative Medicine Center, UMCU, UU, 3584 CT Utrecht, The Netherlands
| | - Ben Pode-Shakked
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
- The Institute for Rare Diseases, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel
- Talpiot Medical Leadership Program, Sheba Medical Center, Ramat Gan 5262100, Israel
| | - Marliek van Hoesel
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), 3584 CT Utrecht, The Netherlands; (J.M.v.R.); (J.M.A.S.); (M.v.H.); (R.H.J.H.); (E.E.S.N.)
- Regenerative Medicine Center, UMCU, UU, 3584 CT Utrecht, The Netherlands
| | - Elee Shimshoni
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel; (E.S.); (I.S.)
| | - Sylvie Polak-Charcon
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
- Institute of Pathology, Sheba Medical Center, Ramat Gan 5262100, Israel
| | - Liron Talmi
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
- Pediatric Department A, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel
| | - Makbule Eren
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir 26040, Turkey; (Y.A.); (M.E.)
| | - Batia Weiss
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel; (L.W.); (B.W.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
| | - Roderick H.J. Houwen
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), 3584 CT Utrecht, The Netherlands; (J.M.v.R.); (J.M.A.S.); (M.v.H.); (R.H.J.H.); (E.E.S.N.)
| | - Iris Barshack
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
- Institute of Pathology, Sheba Medical Center, Ramat Gan 5262100, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
- Pediatric Department A, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel
- Immunology Service, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel
- Jeffrey Modell Foundation Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel
| | - Edward E.S. Nieuwenhuis
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), 3584 CT Utrecht, The Netherlands; (J.M.v.R.); (J.M.A.S.); (M.v.H.); (R.H.J.H.); (E.E.S.N.)
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel; (E.S.); (I.S.)
| | - Annick Raas-Rothschild
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
- The Institute for Rare Diseases, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel
| | - Sabine Middendorp
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), 3584 CT Utrecht, The Netherlands; (J.M.v.R.); (J.M.A.S.); (M.v.H.); (R.H.J.H.); (E.E.S.N.)
- Regenerative Medicine Center, UMCU, UU, 3584 CT Utrecht, The Netherlands
| | - Dror S. Shouval
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan 5262100, Israel; (L.W.); (B.W.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel; (B.P.-S.); (S.P.-C.); (L.T.); (I.B.); (R.S.); (A.R.-R.)
| |
Collapse
|
33
|
Alix M, Gasset E, Bardon-Albaret A, Noel J, Pirot N, Perez V, Coves D, Saulnier D, Lignot JH, Cucchi PN. Description of the unusual digestive tract of Platax orbicularis and the potential impact of Tenacibaculum maritimum infection. PeerJ 2020; 8:e9966. [PMID: 33024633 PMCID: PMC7520087 DOI: 10.7717/peerj.9966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Background Ephippidae fish are characterized by a discoid shape with a very small visceral cavity. Among them Platax orbicularis has a high economic potential due to its flesh quality and flesh to carcass ratio. Nonetheless, the development of its aquaculture is limited by high mortality rates, especially due to Tenacibaculum maritimum infection, occurring one to three weeks after the transfer of fishes from bio-secure land-based aquaculture system to the lagoon cages for growth. Among the lines of defense against this microbial infection, the gastrointestinal tract (GIT) is less studied. The knowledge about the morphofunctional anatomy of this organ in P. orbicularis is still scarce. Therefore, the aims of this study are to characterize the GIT in non-infected P. orbicularis juveniles to then investigate the impact of T. maritimum on this multifunctional organ. Methods In the first place, the morpho-anatomy of the GIT in non-infected individuals was characterized using various histological techniques. Then, infected individuals, experimentally challenged by T. maritimum were analysed and compared to the previously established GIT reference. Results The overlapped shape of the GIT of P. orbicularis is probably due to its constrained compaction in a narrow visceral cavity. Firstly, the GIT was divided into 10 sections, from the esophagus to the rectum. For each section, the structure of the walls was characterized, with a focus on mucus secretions and the presence of the Na+/K+ ATPase pump. An identification key allowing the characterization of the GIT sections using in toto histology is given. Secondly, individuals challenged with T. maritimum exhibited differences in mucus type and proportion and, modifications in the mucosal and muscle layers. These changes could induce an imbalance in the trade-off between the GIT functions which may be in favour of protection and immunity to the disadvantage of nutrition capacities.
Collapse
Affiliation(s)
- Maud Alix
- MARBEC, Univ Montpellier, CNRS, Ifremer, IRD, Montpellier, France.,Institute of Marine Research, Bergen, Norway
| | - Eric Gasset
- MARBEC, Univ Montpellier, CNRS, Ifremer, IRD, Montpellier, France
| | - Agnes Bardon-Albaret
- Ifremer, UMR Ecosystèmes Insulaires Océaniens, UPF, ILM, IRD, Tahiti, French Polynesia
| | - Jean Noel
- BCM, Université de Montpellier, CNRS, INSERM, Montpellier, France.,IRCM, Université de Montpellier, ICM, INSERM, Montpellier, France
| | - Nelly Pirot
- BCM, Université de Montpellier, CNRS, INSERM, Montpellier, France.,IRCM, Université de Montpellier, ICM, INSERM, Montpellier, France
| | - Valérie Perez
- MARBEC, Univ Montpellier, CNRS, Ifremer, IRD, Montpellier, France
| | - Denis Coves
- MARBEC, Univ Montpellier, CNRS, Ifremer, IRD, Montpellier, France
| | - Denis Saulnier
- Ifremer, UMR Ecosystèmes Insulaires Océaniens, UPF, ILM, IRD, Tahiti, French Polynesia
| | | | | |
Collapse
|
34
|
Badawy A, Elfadul M, Aziabi M, Ageel HI, Aqeel A. Challenges of Microvillus Inclusion Disease in the NICU. Neoreviews 2020; 21:e600-e604. [PMID: 32873653 DOI: 10.1542/neo.21-9-e600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mutations in the myosin 5β, syntaxin-binding protein 2, and syntaxin 3 genes lead to microvillus inclusion disease (MVID), an autosomal recessive congenital enteropathy. This rare disease is characterized by lack of microvilli on the surface of enterocytes in the small intestine, the presence of pathognomonic intracellular microvillus inclusions, and vesicular bodies within these enterocytes. This pathology leads to the characteristic intractable, life-threatening, watery diarrhea. In the more common early-onset form, affected patients present in the first few days after birth, whereas in the late-onset form, clinical manifestations appear at approximately 2 to 3 months of age. Genetic testing can confirm the diagnosis, but the infant's medical history, clinical presentation, and small intestinal biopsy results are strongly suggestive of the diagnosis. The prevalence of MVID is thought to be higher in countries with a high degree of consanguinity. Patients with MVID cannot tolerate feedings and require continuous total parenteral nutrition. Mortality is extremely high in the early-onset type with reports of survival in patients treated with small intestinal transplantation. Medical counseling for parents of infants with MVID needs to reflect our current understanding of the various genetic forms of this disease, the feasible management, and anticipated outcomes.
Collapse
Affiliation(s)
| | | | | | - Hossain Ibrahim Ageel
- Gastroenterology Unit, Pediatrics Department, King Fahd Central Hospital, Gazan, Saudi Arabia
| | | |
Collapse
|
35
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
36
|
Kakni P, Hueber R, Knoops K, López‐Iglesias C, Truckenmüller R, Habibovic P, Giselbrecht S. Intestinal Organoid Culture in Polymer Film‐Based Microwell Arrays. ACTA ACUST UNITED AC 2020; 4:e2000126. [DOI: 10.1002/adbi.202000126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/12/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Panagiota Kakni
- Department of Instructive Biomaterials Engineering MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University Universiteitssingel 40 Maastricht ER 6229 The Netherlands
| | - Rui Hueber
- Department of Instructive Biomaterials Engineering MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University Universiteitssingel 40 Maastricht ER 6229 The Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab Maastricht Multimodal Molecular Imaging Institute (M4I) Maastricht University Universiteitssingel 50 Maastricht ER 6229 The Netherlands
| | - Carmen López‐Iglesias
- Microscopy CORE Lab Maastricht Multimodal Molecular Imaging Institute (M4I) Maastricht University Universiteitssingel 50 Maastricht ER 6229 The Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University Universiteitssingel 40 Maastricht ER 6229 The Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University Universiteitssingel 40 Maastricht ER 6229 The Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University Universiteitssingel 40 Maastricht ER 6229 The Netherlands
| |
Collapse
|
37
|
Zhang F, Qiao S, Li C, Wu B, Reischl S, Neumann PA. The immunologic changes during different phases of intestinal anastomotic healing. J Clin Lab Anal 2020; 34:e23493. [PMID: 32692419 PMCID: PMC7676198 DOI: 10.1002/jcla.23493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 01/06/2023] Open
Abstract
Intestinal anatomosis is a complex and multicellular process that involving three overlapped phases: exudative phase, proliferative phase, and reparative phase. Undisturbed anastomotic healings are crucial for the recovery of patients after operations but unsuccessful healings are linked with a considerable mortality. This time, we concentrate on the immunologic changes during different phases of intestinal anastomotic healing and select several major immune cells and cytokines of each phase to get a better understanding of these immunologic changes in different phases, which will be significant for more precise therapy strategies in anastomoses.
Collapse
Affiliation(s)
- Feng Zhang
- Department of General Surgery, Tongren Municipal People's Hospital of Guizhou Medical University(GMU), Guizhou, 554300, China.,Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| | - Song Qiao
- Department of General Surgery, Tongren Municipal People's Hospital of Guizhou Medical University(GMU), Guizhou, 554300, China
| | - Chunqiao Li
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| | - Bo Wu
- Department of General Surgery, Tongren Municipal People's Hospital of Guizhou Medical University(GMU), Guizhou, 554300, China
| | - Stefan Reischl
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| | - Philipp-Alexander Neumann
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich(TUM), Munich, 81675, Germany
| |
Collapse
|
38
|
Xu R, Karrow NA, Shandilya UK, Sun LH, Kitazawa H. In-Vitro Cell Culture for Efficient Assessment of Mycotoxin Exposure, Toxicity and Risk Mitigation. Toxins (Basel) 2020; 12:E146. [PMID: 32120954 PMCID: PMC7150844 DOI: 10.3390/toxins12030146] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
Mycotoxins are toxic secondary fungal metabolites that commonly contaminate crops and food by-products and thus, animal feed. Ingestion of mycotoxins can lead to mycotoxicosis in both animals and humans, and at subclinical concentrations may affect animal production and adulterate feed and animal by-products. Mycotoxicity mechanisms of action (MOA) are largely unknown, and co-contamination, which is often the case, raises the likelihood of mycotoxin interactions. Mitigation strategies for reducing the risk of mycotoxicity are diverse and may not necessarily provide protection against all mycotoxins. These factors, as well as the species-specific risk of toxicity, collectively make an assessment of exposure, toxicity, and risk mitigation very challenging and costly; thus, in-vitro cell culture models provide a useful tool for their initial assessment. Since ingestion is the most common route of mycotoxin exposure, the intestinal epithelial barrier comprised of epithelial cells (IECs) and immune cells such as macrophages, represents ground zero where mycotoxins are absorbed, biotransformed, and elicit toxicity. This article aims to review different in-vitro IEC or co-culture models that can be used for assessing mycotoxin exposure, toxicity, and risk mitigation, and their suitability and limitations for the safety assessment of animal foods and food by-products.
Collapse
Affiliation(s)
- Ran Xu
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.X.); (U.K.S.)
| | - Niel A. Karrow
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.X.); (U.K.S.)
| | - Umesh K. Shandilya
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.X.); (U.K.S.)
| | - Lv-hui Sun
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| |
Collapse
|
39
|
Snyder J, Wang CM, Zhang AQ, Li Y, Luchan J, Hosic S, Koppes R, Carrier RL, Koppes A. Materials and Microenvironments for Engineering the Intestinal Epithelium. Ann Biomed Eng 2020; 48:1916-1940. [DOI: 10.1007/s10439-020-02470-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022]
|
40
|
Tyrosine-Based Signals Regulate the Assembly of Daple⋅PARD3 Complex at Cell-Cell Junctions. iScience 2020; 23:100859. [PMID: 32058970 PMCID: PMC7005484 DOI: 10.1016/j.isci.2020.100859] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/27/2019] [Accepted: 01/16/2020] [Indexed: 01/06/2023] Open
Abstract
Polarized distribution of organelles and molecules inside a cell is vital for a range of cellular processes and its loss is frequently encountered in disease. Polarization during planar cell migration is a special condition in which cellular orientation is triggered by cell-cell contact. We demonstrate that the protein Daple (CCDC88C) is a component of cell junctions in epithelial cells which serves like a cellular “compass” for establishing and maintaining contact-triggered planar polarity. Furthermore, these processes may be mediated through interaction with the polarity regulator PARD3. This interaction, mediated by Daple's PDZ-binding motif (PBM) and the third PDZ domain of PARD3, is fine-tuned by tyrosine phosphorylation on Daple's PBM by receptor and non-receptor tyrosine kinases, such as Src. Hypophosphorylation strengthens the interaction, whereas hyperphosphorylation disrupts it, thereby revealing an unexpected role of Daple as a platform for signal integration and gradient sensing for tyrosine-based signals within the planar cell polarity pathway. Daple localizes to cell junction, regulates planar cell migration Localization requires Daple's C-terminal PDZ-binding motif (PBM) The PBM binds a PDZ module of the polarity determinant PARD3 The Daple⋅PARD3 interaction is regulated by tyrosine-based signals
Collapse
|
41
|
Taverner A, MacKay J, Laurent F, Hunter T, Liu K, Mangat K, Song L, Seto E, Postlethwaite S, Alam A, Chandalia A, Seung M, Saberi M, Feng W, Mrsny RJ. Cholix protein domain I functions as a carrier element for efficient apical to basal epithelial transcytosis. Tissue Barriers 2020; 8:1710429. [PMID: 31928299 PMCID: PMC7063863 DOI: 10.1080/21688370.2019.1710429] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cholix (Chx) is expressed by the intestinal pathogen Vibrio cholerae as a single chain of 634 amino acids (~70.7 kDa protein) that folds into three distinct domains, with elements of the second and third domains being involved in accessing the cytoplasm of nonpolarized cells and inciting cell death via ADP-ribosylation of elongation factor 2, respectively. In order to reach nonpolarized cells within the intestinal lamina propria, however, Chx must cross the polarized epithelial barrier in an intact form. Here, we provide in vitro and in vivo demonstrations that a nontoxic Chx transports across intestinal epithelium via a vesicular trafficking pathway that rapidly achieves vesicular apical to basal (A→B) transcytosis and avoids routing to lysosomes. Specifically, Chx traffics in apical endocytic Rab7+ vesicles and in basal exocytic Rab11+ vesicles with a transition between these domains occurring in the ER-Golgi intermediate compartment (ERGIC) through interactions with the lectin mannose-binding protein 1 (LMAN1) protein that undergoes an intracellular re-distribution that coincides with the re-organization of COPI+ and COPII+ vesicular structures. Truncation studies demonstrated that domain I of Chx alone was sufficient to efficiently complete A→B transcytosis and capable of ferrying genetically conjoined human growth hormone (hGH). These studies provide evidence for a pathophysiological strategy where native Chx exotoxin secreted in the intestinal lumen by nonpandemic V. cholerae can reach nonpolarized cells within the lamina propria in an intact form by using a nondestructive pathway to cross in the intestinal epithelial that appears useful for oral delivery of biopharmaceuticals.One-Sentence Summary: Elements within the first domain of the Cholix exotoxin protein are essential and sufficient for the apical to basal transcytosis of this Vibrio cholerae-derived virulence factor across polarized intestinal epithelial cells.
Collapse
Affiliation(s)
- Alistair Taverner
- Department of Pharmacy and Pharmacology, University of Bath, Bath, England
| | - Julia MacKay
- Department of Pharmacy and Pharmacology, University of Bath, Bath, England
| | - Floriane Laurent
- Department of Pharmacy and Pharmacology, University of Bath, Bath, England
| | - Tom Hunter
- Applied Molecular Transport, South San Francisco, CA, USA
| | - Keyi Liu
- Applied Molecular Transport, South San Francisco, CA, USA
| | | | - Lisa Song
- Applied Molecular Transport, South San Francisco, CA, USA
| | - Elbert Seto
- Applied Molecular Transport, South San Francisco, CA, USA
| | | | - Aatif Alam
- Applied Molecular Transport, South San Francisco, CA, USA
| | | | - Minji Seung
- Applied Molecular Transport, South San Francisco, CA, USA
| | - Mazi Saberi
- Applied Molecular Transport, South San Francisco, CA, USA
| | - Weijun Feng
- Applied Molecular Transport, South San Francisco, CA, USA
| | - Randall J Mrsny
- Department of Pharmacy and Pharmacology, University of Bath, Bath, England.,Applied Molecular Transport, South San Francisco, CA, USA
| |
Collapse
|
42
|
Nakajima T, Sasaki K, Yamamori A, Sakurai K, Miyata K, Watanabe T, Matsunaga YT. A simple three-dimensional gut model constructed in a restricted ductal microspace induces intestinal epithelial cell integrity and facilitates absorption assays. Biomater Sci 2020; 8:5615-5627. [DOI: 10.1039/d0bm00763c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A new 3D gut-on-a-chip on a ductal scaffold induced a differentiated epithelial layer and allowed permeability and absorption assay.
Collapse
Affiliation(s)
- Tadaaki Nakajima
- Institute of Industrial Science
- The University of Tokyo
- Tokyo 153-8505
- Japan
| | | | | | | | | | | | | |
Collapse
|
43
|
Zhang Y, Zheng L, Le M, Nakano Y, Chan B, Huang Y, Torbaty PM, Kohwi Y, Marcucio R, Habelitz S, Den Besten PK, Kohwi-Shigematsu T. SATB1 establishes ameloblast cell polarity and regulates directional amelogenin secretion for enamel formation. BMC Biol 2019; 17:104. [PMID: 31830989 PMCID: PMC6909472 DOI: 10.1186/s12915-019-0722-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 11/13/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Polarity is necessary for epithelial cells to perform distinct functions at their apical and basal surfaces. Oral epithelial cell-derived ameloblasts at secretory stage (SABs) synthesize large amounts of enamel matrix proteins (EMPs), largely amelogenins. EMPs are unidirectionally secreted into the enamel space through their apical cytoplasmic protrusions, or Tomes' processes (TPs), to guide the enamel formation. Little is known about the transcriptional regulation underlying the establishment of cell polarity and unidirectional secretion of SABs. RESULTS The higher-order chromatin architecture of eukaryotic genome plays important roles in cell- and stage-specific transcriptional programming. A genome organizer, special AT-rich sequence-binding protein 1 (SATB1), was discovered to be significantly upregulated in ameloblasts compared to oral epithelial cells using a whole-transcript microarray analysis. The Satb1-/- mice possessed deformed ameloblasts and a thin layer of hypomineralized and non-prismatic enamel. Remarkably, Satb1-/- ameloblasts at the secretory stage lost many morphological characteristics found at the apical surface of wild-type (wt) SABs, including the loss of Tomes' processes, defective inter-ameloblastic adhesion, and filamentous actin architecture. As expected, the secretory function of Satb1-/- SABs was compromised as amelogenins were largely retained in cells. We found the expression of epidermal growth factor receptor pathway substrate 8 (Eps8), a known regulator for actin filament assembly and small intestinal epithelial cytoplasmic protrusion formation, to be SATB1 dependent. In contrast to wt SABs, EPS8 could not be detected at the apical surface of Satb1-/- SABs. Eps8 expression was greatly reduced in small intestinal epithelial cells in Satb1-/- mice as well, displaying defective intestinal microvilli. CONCLUSIONS Our data show that SATB1 is essential for establishing secretory ameloblast cell polarity and for EMP secretion. In line with the deformed apical architecture, amelogenin transport to the apical secretory front and secretion into enamel space were impeded in Satb1-/- SABs resulting in a massive cytoplasmic accumulation of amelogenins and a thin layer of hypomineralized enamel. Our studies strongly suggest that SATB1-dependent Eps8 expression plays a critical role in cytoplasmic protrusion formation in both SABs and in small intestines. This study demonstrates the role of SATB1 in the regulation of amelogenesis and the potential application of SATB1 in ameloblast/enamel regeneration.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Orofacial Sciences, University of California, San Francisco, USA.
| | - Liwei Zheng
- Department of Orofacial Sciences, University of California, San Francisco, USA
| | - Michael Le
- Department of Orofacial Sciences, University of California, San Francisco, USA
| | - Yukiko Nakano
- Department of Orofacial Sciences, University of California, San Francisco, USA
| | - Barry Chan
- Department of Orofacial Sciences, University of California, San Francisco, USA
| | - Yulei Huang
- Department of Orofacial Sciences, University of California, San Francisco, USA
| | | | - Yoshinori Kohwi
- Department of Orofacial Sciences, University of California, San Francisco, USA
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, University of California, San Francisco, USA
| | - Stefan Habelitz
- Preventive and Restorative Dental Sciences, University of California, San Francisco, USA
| | - Pamela K Den Besten
- Department of Orofacial Sciences, University of California, San Francisco, USA
| | | |
Collapse
|
44
|
Jayawardena D, Alrefai WA, Dudeja PK, Gill RK. Recent advances in understanding and managing malabsorption: focus on microvillus inclusion disease. F1000Res 2019; 8. [PMID: 31824659 PMCID: PMC6896243 DOI: 10.12688/f1000research.20762.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2019] [Indexed: 12/11/2022] Open
Abstract
Microvillus inclusion disease (MVID) is a rare congenital severe malabsorptive and secretory diarrheal disease characterized by blunted or absent microvilli with accumulation of secretory granules and inclusion bodies in enterocytes. The typical clinical presentation of the disease is severe chronic diarrhea that rapidly leads to dehydration and metabolic acidosis. Despite significant advances in our understanding of the causative factors, to date, no curative therapy for MVID and associated diarrhea exists. Prognosis mainly relies on life-long total parenteral nutrition (TPN) and eventual small bowel and/or liver transplantation. Both TPN and intestinal transplantation are challenging and present with many side effects. A breakthrough in the understanding of MVID emanated from seminal findings revealing mutations in
MYO5B as a cause for MVID. During the last decade, many studies have thus utilized cell lines and animal models with knockdown of
MYO5B to closely recapitulate the human disease and investigate potential therapeutic options in disease management. We will review the most recent advances made in the research pertaining to MVID. We will also highlight the tools and models developed that can be utilized for basic and applied research to increase our understanding of MVID and develop novel and effective targeted therapies.
Collapse
Affiliation(s)
- Dulari Jayawardena
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Waddah A Alrefai
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep K Dudeja
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
45
|
Kim R, Attayek PJ, Wang Y, Furtado KL, Tamayo R, Sims CE, Allbritton NL. An in vitro intestinal platform with a self-sustaining oxygen gradient to study the human gut/microbiome interface. Biofabrication 2019; 12:015006. [PMID: 31519008 PMCID: PMC6933551 DOI: 10.1088/1758-5090/ab446e] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An oxygen gradient formed along the length of colonic crypts supports stem-cell proliferation at the normoxic crypt base while supporting obligate anaerobe growth in the anoxic colonic lumen. Primary human colonic epithelial cells derived from human gastrointestinal stem cells were cultured within a device possessing materials of tailored oxygen permeability to produce an oxygen-depleted luminal (0.8% ± 0.1% O2) and oxygen-rich basal (11.1% ± 0.5% O2) compartment. This oxygen difference created a stable oxygen gradient across the colonic epithelial cells which remained viable and properly polarized. Facultative and obligate anaerobes Lactobacillus rhamnosus, Bifidobacterium adolescentis, and Clostridium difficile grew readily within the luminal compartment. When formed along the length of an in vitro crypt, the oxygen gradient facilitated cell compartmentalization within the crypt by enhancing confinement of the proliferative cells to the crypt base. This platform provides a simple system to create a physiological oxygen gradient across an intestinal mimic while simultaneously supporting anaerobe co-culture.
Collapse
Affiliation(s)
- Raehyun Kim
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Peter J. Attayek
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen L. Furtado
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Rita Tamayo
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Nancy L. Allbritton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
46
|
Current understanding of the gut microbiota shaping mechanisms. J Biomed Sci 2019; 26:59. [PMID: 31434568 PMCID: PMC6702754 DOI: 10.1186/s12929-019-0554-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Increasing evidences have shown strong associations between gut microbiota and many human diseases, and understanding the dynamic crosstalks of host-microbe interaction in the gut has become necessary for the detection, prevention, or therapy of diseases. Many reports have showed that diet, nutrient, pharmacologic factors and many other stimuli play dominant roles in the modulation of gut microbial compositions. However, it is inappropriate to neglect the impact of host factors on shaping the gut microbiota. In this review, we highlighted the current findings of the host factors that could modulate the gut microbiota. Particularly the epithelium-associated factors, including the innate immune sensors, anti-microbial peptides, mucus barrier, secretory IgAs, epithelial microvilli, epithelial tight junctions, epithelium metabolism, oxygen barrier, and even the microRNAs are discussed in the context of the microbiota shaping. With these shaping factors, the gut epithelial cells could select the residing microbes and affect the microbial composition. This knowledge not only could provide the opportunities to better control many diseases, but may also be used for predicting the success of fecal microbiota transplantation clinically.
Collapse
|
47
|
Meenderink LM, Gaeta IM, Postema MM, Cencer CS, Chinowsky CR, Krystofiak ES, Millis BA, Tyska MJ. Actin Dynamics Drive Microvillar Motility and Clustering during Brush Border Assembly. Dev Cell 2019; 50:545-556.e4. [PMID: 31378589 DOI: 10.1016/j.devcel.2019.07.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 05/03/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Transporting epithelial cells generate arrays of microvilli, known as a brush border, to enhance functional capacity. To understand brush border formation, we used live cell imaging to visualize apical remodeling early in this process. Strikingly, we found that individual microvilli exhibit persistent active motility, translocating across the cell surface at ∼0.2 μm/min. Perturbation with inhibitors and photokinetic experiments revealed that microvillar motility is driven by actin assembly at the barbed ends of core bundles, which in turn is linked to robust treadmilling of these structures. Actin regulatory factors IRTKS and EPS8 localize to the barbed ends of motile microvilli, where they control the kinetics and nature of movement. As the apical surface of differentiating epithelial cells is crowded with nascent microvilli, persistent motility promotes collisions between protrusions and ultimately clustering and consolidation into higher-order arrays. Thus, microvillar motility represents a previously unrecognized driving force for apical surface remodeling and maturation during epithelial differentiation.
Collapse
Affiliation(s)
- Leslie M Meenderink
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Isabella M Gaeta
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Meagan M Postema
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Caroline S Cencer
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Colbie R Chinowsky
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Evan S Krystofiak
- Cell Imaging Shared Resource, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA; Cell Imaging Shared Resource, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
48
|
Bidaud-Meynard A, Nicolle O, Heck M, Le Cunff Y, Michaux G. A V0-ATPase-dependent apical trafficking pathway maintains the polarity of the intestinal absorptive membrane. Development 2019; 146:dev174508. [PMID: 31110027 PMCID: PMC7376742 DOI: 10.1242/dev.174508] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/07/2019] [Indexed: 12/18/2022]
Abstract
Intestine function relies on the strong polarity of intestinal epithelial cells and the array of microvilli forming a brush border at their luminal pole. Combining a genetic RNA interference (RNAi) screen with in vivo super-resolution imaging in the Caenorhabditiselegans intestine, we found that the V0 sector of the vacuolar ATPase (V0-ATPase) controls a late apical trafficking step, involving Ras-related protein 11 (RAB-11)+ endosomes and the N-ethylmaleimide-sensitive factor-attachment protein receptor (SNARE) synaptosome-associated protein 29 (SNAP-29), and is necessary to maintain the polarized localization of both apical polarity modules and brush border proteins. We show that the V0-ATPase pathway also genetically interacts with glycosphingolipids and clathrin in enterocyte polarity maintenance. Finally, we demonstrate that silencing of the V0-ATPase fully recapitulates the severe structural, polarity and trafficking defects observed in enterocytes from individuals with microvillus inclusion disease (MVID) and use this new in vivo MVID model to follow the dynamics of microvillus inclusions. Thus, we describe a new function for V0-ATPase in apical trafficking and epithelial polarity maintenance and the promising use of the C. elegans intestine as an in vivo model to better understand the molecular mechanisms of rare genetic enteropathies.
Collapse
Affiliation(s)
- Aurélien Bidaud-Meynard
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Ophélie Nicolle
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Markus Heck
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Yann Le Cunff
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Grégoire Michaux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| |
Collapse
|
49
|
Modeling Host-Pathogen Interactions in the Context of the Microenvironment: Three-Dimensional Cell Culture Comes of Age. Infect Immun 2018; 86:IAI.00282-18. [PMID: 30181350 DOI: 10.1128/iai.00282-18] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.
Collapse
|