1
|
Johansson Å, Venkita Subramani M, Yilmaz B, Nyström EE, Layunta E, Arike L, Sommer F, Rosenstiel P, Vereecke L, Mannerås-Holm L, Wullaert A, Pelaseyed T, Johansson ME, Birchenough GM. Neonatal microbiota colonization primes maturation of goblet cell-mediated protection in the pre-weaning colon. J Exp Med 2025; 222:e20241591. [PMID: 40323318 PMCID: PMC12051479 DOI: 10.1084/jem.20241591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/06/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Regulated host-microbe interactions are a critical aspect of lifelong health. Colonic goblet cells protect from microorganisms via the generation of a mucus barrier structure. Bacteria-sensing sentinel goblet cells provide secondary protection by orchestrating mucus secretion when microbes breach the mucus barrier. Mucus deficiencies in germ-free mice implicate a role for the microbiota in programming barrier generation, but its natural ontogeny remains undefined. We now investigate the mucus barrier and sentinel goblet cell development in relation to postnatal colonization. Combined in vivo and ex vivo analyses demonstrate rapid and sequential microbiota-dependent development of these primary and secondary goblet cell protective functions, with dynamic changes in mucus processing dependent on innate immune signaling via MyD88 and development of functional sentinel goblet cells dependent on the NADPH/dual oxidase family member Duox2. Our findings identify new mechanisms of microbiota-goblet cell regulatory interaction and highlight the critical importance of the pre-weaning period for the normal development of protective systems that are key legislators of host-microbiota interaction.
Collapse
Affiliation(s)
- Åsa Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mahadevan Venkita Subramani
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Bahtiyar Yilmaz
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Elisabeth E.L. Nyström
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Elena Layunta
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Liisa Arike
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Felix Sommer
- Institute of Clinical & Molecular Biology, University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical & Molecular Biology, University of Kiel, Kiel, Germany
| | - Lars Vereecke
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Louise Mannerås-Holm
- Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Andy Wullaert
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Biomedical Sciences, Cell Death Signalling Lab, University of Antwerp, Antwerp, Belgium
| | - Thaher Pelaseyed
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Malin E.V. Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - George M.H. Birchenough
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
2
|
Keller M, Gallagher C, Kreiselmaier S, Bickenbach K, Schmitt U, Marengo L, Taghikhah D, Abukhalaf M, Tholey A, Becker-Pauly C, Mittmann T, Pietrzik CU. Meprin β Modulates Brevican Proteolysis Impairing Neural Plasticity and Memory Formation. FASEB J 2025; 39:e70616. [PMID: 40396346 DOI: 10.1096/fj.202500017r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/11/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
The metalloprotease meprin β is known for its multifunctional involvement in various physiological processes throughout the body including the brain. However, its broader functions within the brain besides amyloid β generation remain largely unexplored. To investigate this, we utilized a mouse model overexpressing meprin β in neurons within the cortex and hippocampus, regions crucial for learning and memory. Behavioral assessments, employing the Morris' Water Maze paradigm test, revealed impaired cognitive functions in animals overexpressing meprin β. Furthermore, electrophysiological recordings in hippocampal slices using multielectrode arrays showed an impaired long-term potentiation (LTP) in meprin β-overexpressing mice compared to wild-type counterparts. Intriguingly, concomitant with the LTP impairment, we observed an increased neuronal excitability. These findings underline the complicated interplay between meprin β abundance and behavioral manifestations, suggesting a broader impact on neural circuit dynamics. To elucidate the molecular mechanisms underlying these observed deficits, western blotting analyses were conducted to address the expression of glutamatergic receptors. Neither the expression of the N-methyl-D-aspartate (NMDA) nor the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor showed variation relative to each other. The application of N-terminomics identified brevican as a proteolytic substrate of meprin β and thus a potential key mediator linking meprin β overexpression to the observed effects. Previous studies have reported that brevican knockout in animal models influences learning and memory. Our data demonstrate that meprin β modulates brevican expression, likely contributing to the effects we have observed in our mouse model. These results shed light on the broader functional significance of meprin β in neurological processes.
Collapse
Affiliation(s)
- Maximilian Keller
- Institute for Pathobiochemistry, University Medical Center, Mainz, Germany
| | - Celine Gallagher
- Institute for Physiology, University Medical Center, Mainz, Germany
| | - Simon Kreiselmaier
- Institute for Pathobiochemistry, University Medical Center, Mainz, Germany
| | - Kira Bickenbach
- Unit for Degradomics of the Protease Web, Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Ulrich Schmitt
- Leibniz-Institute for Resilience Research, Mainz, Germany
| | - Liana Marengo
- Institute for Pathobiochemistry, University Medical Center, Mainz, Germany
| | - Dayan Taghikhah
- Institute for Pathobiochemistry, University Medical Center, Mainz, Germany
| | - Mohammad Abukhalaf
- Systematic Proteome Research & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany
| | - Andreas Tholey
- Systematic Proteome Research & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany
| | - Christoph Becker-Pauly
- Unit for Degradomics of the Protease Web, Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Thomas Mittmann
- Institute for Physiology, University Medical Center, Mainz, Germany
| | - Claus U Pietrzik
- Institute for Pathobiochemistry, University Medical Center, Mainz, Germany
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
3
|
Fang X, Liu H, Liu J, Du Y, Chi Z, Bian Y, Zhao X, Teng T, Shi B. Isobutyrate Confers Resistance to Inflammatory Bowel Disease through Host-Microbiota Interactions in Pigs. RESEARCH (WASHINGTON, D.C.) 2025; 8:0673. [PMID: 40342298 PMCID: PMC12059313 DOI: 10.34133/research.0673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 05/11/2025]
Abstract
Supplementation with short-chain fatty acids (SCFAs) is a potential therapeutic approach for inflammatory bowel disease (IBD). However, the therapeutic effects and mechanisms of action of isobutyrate in IBD remain unclear. Clinical data indicate that the fecal levels of isobutyrate are markedly lower in patients with Crohn's disease than in healthy controls. Compared with healthy mice and healthy pigs, mice and pigs with colitis presented significantly lower isobutyrate levels. Furthermore, the level of isobutyrate in pigs was significantly negatively correlated with the disease activity index. We speculate that isobutyrate may play a crucial role in regulating host gut homeostasis. We established a model of dextran sulfate sodium-induced colitis in pigs, which have gastrointestinal structure and function similar to those of humans; we performed multiomic analysis to investigate the therapeutic effects and potential mechanisms of isobutyrate on IBD at both the animal and cellular levels and validated the results. Phenotypically, isobutyrate can significantly alleviate diarrhea, bloody stools, weight loss, and colon shortening caused by colitis in pigs. Mechanistically, isobutyrate can increase the relative abundance of Lactobacillus reuteri, thereby increasing the production of indole-3-lactic acid, regulating aryl hydrocarbon receptor expression and downstream signaling pathways, and regulating Foxp3+ CD4+ T cell recruitment to alleviate colitis. Isobutyrate can directly activate G protein-coupled receptor 109A, promote the expression of Claudin-1, and improve intestinal barrier function. In addition, isobutyrate can increase the production of intestinal SCFAs and 3-hydroxybutyric acid and inhibit the TLR4/MyD88/NF-κB signaling pathway to suppress intestinal inflammation. In conclusion, our findings demonstrate that isobutyrate confers resistance to IBD through host-microbiota interactions, providing a theoretical basis for the use of isobutyrate in alleviating colitis.
Collapse
Affiliation(s)
| | | | - Junling Liu
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Yongqing Du
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Zihan Chi
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Yiqi Bian
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Xuan Zhao
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Teng Teng
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| | - Baoming Shi
- College of Animal Science and Technology,
Northeast Agricultural University, Harbin 150030, People’s Republic of China
| |
Collapse
|
4
|
Bickenbach K, David N, Koudelka T, Joos C, Scharfenberg F, Rüffer M, Armbrust F, Georgiadis D, Beau F, Stahmer L, Rahn S, Tholey A, Pietrzik C, Becker-Pauly C. Targeted approach to determine the impact of cancer-associated protease variants. SCIENCE ADVANCES 2025; 11:eadp5958. [PMID: 39937919 PMCID: PMC11818018 DOI: 10.1126/sciadv.adp5958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 01/09/2025] [Indexed: 02/14/2025]
Abstract
Several steps of cancer progression, from tumor onset to metastasis, critically involve proteolytic activity. To elucidate the role of proteases in cancer, it is particularly important to consider single-nucleotide variants (SNVs) that affect the active site of proteases, thereby influencing cleavage specificity, substrate processing, and thus cancer cell behavior. To facilitate systematic studies, we here present a targeted approach to determine the impact of cancer-associated protease variants (TACAP). Starting with the semiautomated identification of potential specificity-modulating SNVs, our workflow comprises mass spectrometry-based cleavage specificity profiling and substrate identification, localization, and inhibitor studies, followed by functional analyses investigating cancer cell properties. To demonstrate the feasibility of TACAP, we analyzed the meprin β R238Q variant. This amino acid exchange R238Q leads to a loss of meprin β's characteristic cleavage preference for acidic amino acids at P1' position, accompanied with changes in substrate pool and inhibitor affinity compared to meprin β wild type.
Collapse
Affiliation(s)
- Kira Bickenbach
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Nele David
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Tomas Koudelka
- Systematic Proteomics and Bioanalytics, Institute for Experimental Medicine, University of Kiel, Kiel, Germany
| | - Corentin Joos
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Franka Scharfenberg
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Malina Rüffer
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Fred Armbrust
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Dimitris Georgiadis
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Fabrice Beau
- CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Lea Stahmer
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Sascha Rahn
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Andreas Tholey
- Systematic Proteomics and Bioanalytics, Institute for Experimental Medicine, University of Kiel, Kiel, Germany
| | - Claus Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christoph Becker-Pauly
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| |
Collapse
|
5
|
Qiao Y, He C, Xia Y, Ocansey DKW, Mao F. Intestinal mucus barrier: A potential therapeutic target for IBD. Autoimmun Rev 2025; 24:103717. [PMID: 39662652 DOI: 10.1016/j.autrev.2024.103717] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Intestinal mucus, a viscoelastic medium with mucin2 (MUC2) as its main component, covers the surface of intestinal epithelial cells and protects the intestine from invasion, forming the first barrier of the intestinal tract. Unlike the small intestine, where the mucus layer is a single layer, the colonic mucus layer can be divided into a sterile inner layer and an outer layer with bacterial colonization. Many of the substances in the mucus layer have beneficial effects on the intestinal epithelium, but the mucus layer is often affected by a variety of factors, mainly microbiological, dietary, and immunological. Inflammatory bowel disease (IBD) is a disease of increasing morbidity worldwide, with a complex etiology and a high relapse rate. In recent years, the mucus barrier in IBD has received increasing attention and is considered a key factor in the pathogenesis of IBD. Loss of goblet cells (GCs) and changes in the composition and properties of the mucus layer material are commonly found in the colon of IBD patients. Damage to the mucus layer may make it easier for microorganisms to access the intestinal epithelium and cause inflammation. There are currently a number of herbs and other therapies that can be used to treat IBD and repair the damaged mucus barrier. This review highlights the important role of the mucus layer in IBD and the therapies that target the mucus layer in IBD.
Collapse
Affiliation(s)
- Yaru Qiao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Changer He
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang 212399, Jiangsu, PR China
| | - Yuxuan Xia
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast CC0959347, Ghana
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
6
|
Hada A, Xiao Z. Ligands for Intestinal Intraepithelial T Lymphocytes in Health and Disease. Pathogens 2025; 14:109. [PMID: 40005486 PMCID: PMC11858322 DOI: 10.3390/pathogens14020109] [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: 12/26/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
The intestinal tract is constantly exposed to a diverse mixture of luminal antigens, such as those derived from commensals, dietary substances, and potential pathogens. It also serves as a primary route of entry for pathogens. At the forefront of this intestinal defense is a single layer of epithelial cells that forms a critical barrier between the gastrointestinal (GI) lumen and the underlying host tissue. The intestinal intraepithelial T lymphocytes (T-IELs), one of the most abundant lymphocyte populations in the body, play a crucial role in actively surveilling and maintaining the integrity of this barrier by tolerating non-harmful factors such as commensal microbiota and dietary components, promoting epithelial turnover and renewal while also defending against pathogens. This immune balance is maintained through interactions between ligands in the GI microenvironment and receptors on T-IELs. This review provides a detailed examination of the ligands present in the intestinal epithelia and the corresponding receptors expressed on T-IELs, including T cell receptors (TCRs) and non-TCRs, as well as how these ligand-receptor interactions influence T-IEL functions under both steady-state and pathological conditions. By understanding these engagements, we aim to shed light on the mechanisms that govern T-IEL activities within the GI microenvironment. This knowledge may help in developing strategies to target GI ligands and modulate T-IEL receptor expression, offering precise approaches for treating intestinal disorders.
Collapse
Affiliation(s)
| | - Zhengguo Xiao
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
7
|
Hashemi Shahraki F, Evazzadeh N, Aminzadeh S. Heterologous expression, purification, and biochemical characterization of protease 3075 from Cohnella sp. A01. PLoS One 2024; 19:e0310910. [PMID: 39680596 DOI: 10.1371/journal.pone.0310910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/09/2024] [Indexed: 12/18/2024] Open
Abstract
Proteases as one of the most significant categories of commercial enzymes, serve nowadays as the key ingredients in detergent formulations. Therefore, identifying detergent-compatible proteases with better properties is a continuous exercise. Accordingly, we were interested in the recombinant production and characterization of protease 3075 as a novel enzyme from thermophilic indigenous Cohnella sp. A01. The biochemical and structural features of the protease were probed by employing bioinformatic methods and in vitro studies. The bioinformatics analysis discovered that protease 3075 belongs to the C56-PfpI superfamily. The protease 3075 gene was cloned and heterologous expressed in Escherichia coli (E. coli) BL21. It was found that the enzyme contains 175 amino acids and 525 bp with a molecular weight of 19 kDa. Protease 3075 revealed acceptable activity in the range of 40-80°C and pH 5.5-8. The optimum activity of the enzyme was observed at 70°C and pH 6. The activity of protease 3075 increased about 4-fold in the presence of Tween 80 and acetone, while its activity attenuated in the presence of iodoacetic acid and iodoacetamide. Docking analyses revealed the dominant interaction between Tween 80 and protease 3075, mediated by hydrogen bonds and Van der Waals forces. Furthermore, molecular dynamics simulations (MDS) showed that Tween 80 increased the stability of the protease 3075 structure. Altogether, our data provided a novel enzyme by genetic manipulation process that could have significant industrial applications.
Collapse
Affiliation(s)
- Fatemeh Hashemi Shahraki
- Bioprocess Engineering Group, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Narges Evazzadeh
- Bioprocess Engineering Group, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Saeed Aminzadeh
- Bioprocess Engineering Group, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
8
|
Lozinski BM, Ta K, Dong Y. Emerging role of galectin 3 in neuroinflammation and neurodegeneration. Neural Regen Res 2024; 19:2004-2009. [PMID: 38227529 DOI: 10.4103/1673-5374.391181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
Neuroinflammation and neurodegeneration are key processes that mediate the development and progression of neurological diseases. However, the mechanisms modulating these processes in different diseases remain incompletely understood. Advances in single cell based multi-omic analyses have helped to identify distinct molecular signatures such as Lgals3 that is associated with neuroinflammation and neurodegeneration in the central nervous system (CNS). Lgals3 encodes galectin-3 (Gal3), a β-galactoside and glycan binding glycoprotein that is frequently upregulated by reactive microglia/macrophages in the CNS during various neurological diseases. While Gal3 has previously been associated with non-CNS inflammatory and fibrotic diseases, recent studies highlight Gal3 as a prominent regulator of inflammation and neuroaxonal damage in the CNS during diseases such as multiple sclerosis, Alzheimer's disease, and Parkinson's disease. In this review, we summarize the pleiotropic functions of Gal3 and discuss evidence that demonstrates its detrimental role in neuroinflammation and neurodegeneration during different neurological diseases. We also consider the challenges of translating preclinical observations into targeting Gal3 in the human CNS.
Collapse
Affiliation(s)
- Brian M Lozinski
- Department of Clinical Neuroscience, University of Calgary, Calgary, AB, Canada
| | - Khanh Ta
- Deparment of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yifei Dong
- Deparment of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
9
|
Xie Y, Zhao F, Wang Y, Borowski S, Freitag N, Tirado-Gonzalez I, Hofsink N, Matschl U, Plösch T, Garcia MG, Blois SM. Fetal growth restriction induced by maternal gal-3 deficiency is associated with altered gut-placenta axis. Cell Death Dis 2024; 15:575. [PMID: 39117607 PMCID: PMC11310209 DOI: 10.1038/s41419-024-06962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Adverse intrauterine conditions may cause fetal growth restriction (FGR), a pregnancy complication frequently linked to perinatal morbidity and mortality. Although many studies have focused on FGR, the pathophysiological processes underlying this disorder are complex and incompletely understood. We have recently determined that galectin-3 (gal-3), a β-galactoside-binding protein, regulates pregnancy-associated processes, including uterine receptibility, maternal vascular adaptation and placentation. Because gal-3 is expressed at both sides of the maternal-fetal interface, we unraveled the contribution of maternal- and paternal-derived gal-3 on fetal-placental development in the prenatal window and its effects on the post-natal period. Deficiency of maternal gal-3 induced maternal gut microbiome dysbiosis, resulting in a sex-specific fetal growth restriction mainly observed in female fetuses and offspring. In addition, poor placental metabolic adaptions (characterized by decreased trophoblast glycogen content and insulin-like growth factor 2 (Igf2) gene hypomethylation) were only associated with a lack of maternal-derived gal-3. Paternal gal-3 deficiency caused compromised vascularization in the placental labyrinth without affecting fetal growth trajectory. Thus, maternal-derived gal-3 may play a key role in fetal-placental development through the gut-placenta axis.
Collapse
Affiliation(s)
- Yiran Xie
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fangqi Zhao
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yiru Wang
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sophia Borowski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and Institute of Biochemistry, Berlin, Germany and Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Nancy Freitag
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and Institute of Biochemistry, Berlin, Germany and Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Irene Tirado-Gonzalez
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Naomi Hofsink
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Urte Matschl
- Department Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Perinatal Neurobiology, Department of Human Medicine, School of Medicine and Health Sciences, Carlvon Ossietzky University Oldenburg, Oldenburg, Germany
| | - Mariana G Garcia
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
10
|
Johansson Å, Subramani MV, Yilmaz B, Nyström E, Layunta E, Arike L, Sommer F, Rosenstiel P, Vereecke L, Holm LM, Wullaert A, Pelaseyed T, Johansson MEV, Birchenough GMH. Neonatal microbiota colonization drives maturation of primary and secondary goblet cell mediated protection in the pre-weaning colon. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601781. [PMID: 39005291 PMCID: PMC11245021 DOI: 10.1101/2024.07.03.601781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In the distal colon, mucus secreting goblet cells primarily confer protection from luminal microorganisms via generation of a sterile inner mucus layer barrier structure. Bacteria-sensing sentinel goblet cells provide a secondary defensive mechanism that orchestrates mucus secretion in response to microbes that breach the mucus barrier. Previous reports have identified mucus barrier deficiencies in adult germ-free mice, thus implicating a fundamental role for the microbiota in programming mucus barrier generation. In this study, we have investigated the natural neonatal development of the mucus barrier and sentinel goblet cell-dependent secretory responses upon postnatal colonization. Combined in vivo and ex vivo analyses of pre- and post-weaning colonic mucus barrier and sentinel goblet cell maturation demonstrated a sequential microbiota-dependent development of these primary and secondary goblet cell-intrinsic protective functions, with dynamic changes in mucus processing dependent on innate immune signalling via MyD88, and development of functional sentinel goblet cells dependent on the NADPH/Dual oxidase family member Duox2. Our findings therefore identify new mechanisms of microbiota-goblet cell regulatory interaction and highlight the critical importance of the pre-weaning period for the normal development of colonic barrier function.
Collapse
|
11
|
Paganelli A, Diomede F, Marconi GD, Pizzicannella J, Rajan TS, Trubiani O, Paganelli R. Inhibition of LPS-Induced Inflammatory Response of Oral Mesenchymal Stem Cells in the Presence of Galectin-3. Biomedicines 2023; 11:1519. [PMID: 37371614 DOI: 10.3390/biomedicines11061519] [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: 04/17/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Galectin-3 (GAL-3) is a beta-galactoside binding lectin produced by mesenchymal stem cells (MSCs) and other cell sources under inflammatory conditions. Several studies have reported that GAL-3 exerts an anti-inflammatory action, regulated by its natural ligand GAL-3 BP. In the present study, we aimed to assess the GAL-3 mediated regulation of the MSC function in an LPS-induced inflammation setting. Human gingival mesenchymal stem cells (hGMSCs) were stimulated in vitro with LPSs; the expression of TLR4, NFκB p65, MyD88 and NALP3 were assessed in the hGMSCs via immunofluorescence imaging using confocal microscopy, Western blot assay, and RT-PCR before and after the addition of GAL-3, both alone and with the addition of its inhibitors. LPSs stimulated the expression of TLR4, NFκB p65, MyD88 and NALP3 in hGMSCs, which was inhibited by GAL-3. The addition of either GAL3-BP or the antibody to GAL-3 were able to revert the GAL-3-mediated effects, restoring the expression of TLR4, NFκB p65, MyD88 and NALP3. GAL-3 induces the downregulation of the LPS-induced inflammatory program in MSCs.
Collapse
Affiliation(s)
- Alessia Paganelli
- PhD Course in Clinical and Experimental Medicine, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine and Dentistry, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Guya Diletta Marconi
- Department of Innovative Technologies in Medicine and Dentistry, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Jacopo Pizzicannella
- Department of Engineering and Geology, University "G. d'Annunzio" Chieti-Pescara, Viale Pindaro, 42, 65127 Pescara, Italy
| | - Thangavelu Soundara Rajan
- Research and Development Unit, Theertha Biopharma Private Limited, KIADB, Industrial Area, Bommasandra, Jigani Link Road, Bangalore 560105, India
| | - Oriana Trubiani
- Department of Innovative Technologies in Medicine and Dentistry, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Roberto Paganelli
- Saint Camillus International University of Health and Medical Sciences (UniCamillus), 00131 Rome, Italy
| |
Collapse
|