1
|
Mayassi T, Li C, Segerstolpe Å, Brown EM, Weisberg R, Nakata T, Yano H, Herbst P, Artis D, Graham DB, Xavier RJ. Spatially restricted immune and microbiota-driven adaptation of the gut. Nature 2024; 636:447-456. [PMID: 39567686 DOI: 10.1038/s41586-024-08216-z] [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: 10/11/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
The intestine is characterized by an environment in which host requirements for nutrient and water absorption are consequently paired with the requirements to establish tolerance to the outside environment. To better understand how the intestine functions in health and disease, large efforts have been made to characterize the identity and composition of cells from different intestinal regions1-8. However, the robustness, nature of adaptability and extent of resilience of the transcriptional landscape and cellular underpinning of the intestine in space are still poorly understood. Here we generated an integrated resource of the spatial and cellular landscape of the murine intestine in the steady and perturbed states. Leveraging these data, we demonstrated that the spatial landscape of the intestine was robust to the influence of the microbiota and was adaptable in a spatially restricted manner. Deploying a model of spatiotemporal acute inflammation, we demonstrated that both robust and adaptable features of the landscape were resilient. Moreover, highlighting the physiological relevance and value of our dataset, we identified a region of the middle colon characterized by an immune-driven multicellular spatial adaptation of structural cells to the microbiota. Our results demonstrate that intestinal regionalization is characterized by robust and resilient structural cell states and that the intestine can adapt to environmental stress in a spatially controlled manner through the crosstalk between immunity and structural cell homeostasis.
Collapse
Affiliation(s)
- Toufic Mayassi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Chenhao Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Åsa Segerstolpe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rebecca Weisberg
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Toru Nakata
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hiroshi Yano
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Paula Herbst
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Réthi-Nagy Z, Juhász S. Microbiome's Universe: Impact on health, disease and cancer treatment. J Biotechnol 2024; 392:161-179. [PMID: 39009231 DOI: 10.1016/j.jbiotec.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/27/2024] [Accepted: 07/07/2024] [Indexed: 07/17/2024]
Abstract
The human microbiome is a diverse ecosystem of microorganisms that reside in the body and influence various aspects of health and well-being. Recent advances in sequencing technology have brought to light microbial communities in organs and tissues that were previously considered sterile. The gut microbiota plays an important role in host physiology, including metabolic functions and immune modulation. Disruptions in the balance of the microbiome, known as dysbiosis, have been linked to diseases such as cancer, inflammatory bowel disease and metabolic disorders. In addition, the administration of antibiotics can lead to dysbiosis by disrupting the structure and function of the gut microbial community. Targeting strategies are the key to rebalancing the microbiome and fighting disease, including cancer, through interventions such as probiotics, fecal microbiota transplantation (FMT), and bacteria-based therapies. Future research must focus on understanding the complex interactions between diet, the microbiome and cancer in order to optimize personalized interventions. Multidisciplinary collaborations are essential if we are going to translate microbiome research into clinical practice. This will revolutionize approaches to cancer prevention and treatment.
Collapse
Affiliation(s)
- Zsuzsánna Réthi-Nagy
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged H-6728, Hungary
| | - Szilvia Juhász
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged H-6728, Hungary.
| |
Collapse
|
3
|
Martinez-Blanco M, Mukhatayev Z, Chatila TA. Pathogenic mechanisms in the evolution of food allergy. Immunol Rev 2024; 326:219-226. [PMID: 39285835 PMCID: PMC11488529 DOI: 10.1111/imr.13398] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
The early development of the neonatal immune system is profoundly influenced by exposure to dietary and microbial antigens, which shapes mucosal tolerance. Successful oral tolerance induction is crucially dependent on microbially imprinted immune cells, most notably the RORγt+ regulatory T (Treg) and antigen presenting cells and is essential for preventing food allergy (FA). The development of FA can be envisioned to result from disruptions at key checkpoints (CKPTs) that govern oral tolerance induction. These include gut epithelial sensory and effector circuits that when dysregulated promote pro-allergic gut dysbiosis. They also include microbially imprinted immune regulatory circuits that are disrupted by dysbiosis and pro-allergic immune responses unleashed by the dysregulation of the aforementioned cascades. Understanding these checkpoints is essential for developing therapeutic strategies to restore immune homeostasis in FA.
Collapse
Affiliation(s)
- Monica Martinez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhussipbek Mukhatayev
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Ndjim M, Gasmi I, Herbert F, Joséphine C, Bas J, Lamrani A, Coutry N, Henry S, Zimmermann VS, Dardalhon V, Campillo Poveda M, Turtoi E, Thirard S, Forichon L, Giordano A, Ciancia C, Homayed Z, Pannequin J, Britton C, Devaney E, McNeilly TN, Berrard S, Turtoi A, Maizels RM, Gerbe F, Jay P. Tuft cell acetylcholine is released into the gut lumen to promote anti-helminth immunity. Immunity 2024; 57:1260-1273.e7. [PMID: 38744292 DOI: 10.1016/j.immuni.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 11/26/2023] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Upon parasitic helminth infection, activated intestinal tuft cells secrete interleukin-25 (IL-25), which initiates a type 2 immune response during which lamina propria type 2 innate lymphoid cells (ILC2s) produce IL-13. This causes epithelial remodeling, including tuft cell hyperplasia, the function of which is unknown. We identified a cholinergic effector function of tuft cells, which are the only epithelial cells that expressed choline acetyltransferase (ChAT). During parasite infection, mice with epithelial-specific deletion of ChAT had increased worm burden, fitness, and fecal egg counts, even though type 2 immune responses were comparable. Mechanistically, IL-13-amplified tuft cells release acetylcholine (ACh) into the gut lumen. Finally, we demonstrated a direct effect of ACh on worms, which reduced their fecundity via helminth-expressed muscarinic ACh receptors. Thus, tuft cells are sentinels in naive mice, and their amplification upon helminth infection provides an additional type 2 immune response effector function.
Collapse
Affiliation(s)
- Marième Ndjim
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Imène Gasmi
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Fabien Herbert
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Charlène Joséphine
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Julie Bas
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Ali Lamrani
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Nathalie Coutry
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Sylvain Henry
- Montpellier Alliance for Metabolomics and Metabolism Analysis, Platform for Translational Oncometabolomics (PLATON), Biocampus, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Marta Campillo Poveda
- Centre for Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| | - Evgenia Turtoi
- Montpellier Alliance for Metabolomics and Metabolism Analysis, Platform for Translational Oncometabolomics (PLATON), Biocampus, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Steeve Thirard
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Luc Forichon
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Alicia Giordano
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Claire Ciancia
- Centre for Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| | - Zeinab Homayed
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Julie Pannequin
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Collette Britton
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Eileen Devaney
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Tom N McNeilly
- Disease Control Department, Moredun Research Institute, Penicuik, UK
| | - Sylvie Berrard
- University Paris Cité, Inserm, NeuroDiderot, Paris, France
| | - Andrei Turtoi
- Montpellier Alliance for Metabolomics and Metabolism Analysis, Platform for Translational Oncometabolomics (PLATON), Biocampus, CNRS, INSERM, Université de Montpellier, Montpellier, France; Cancer Research Institute of Montpellier (IRCM), University of Montpellier, Inserm, Montpellier, France
| | - Rick M Maizels
- Centre for Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| | - François Gerbe
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France.
| | - Philippe Jay
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France.
| |
Collapse
|
5
|
Didriksen BJ, Eshleman EM, Alenghat T. Epithelial regulation of microbiota-immune cell dynamics. Mucosal Immunol 2024; 17:303-313. [PMID: 38428738 PMCID: PMC11412483 DOI: 10.1016/j.mucimm.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
The mammalian gastrointestinal tract hosts a diverse community of trillions of microorganisms, collectively termed the microbiota, which play a fundamental role in regulating tissue physiology and immunity. Recent studies have sought to dissect the cellular and molecular mechanisms mediating communication between the microbiota and host immune system. Epithelial cells line the intestine and form an initial barrier separating the microbiota from underlying immune cells, and disruption of epithelial function has been associated with various conditions ranging from infection to inflammatory bowel diseases and cancer. From several studies, it is now clear that epithelial cells integrate signals from commensal microbes. Importantly, these non-hematopoietic cells also direct regulatory mechanisms that instruct the recruitment and function of microbiota-sensitive immune cells. In this review, we discuss the central role that has emerged for epithelial cells in orchestrating intestinal immunity and highlight epithelial pathways through which the microbiota can calibrate tissue-intrinsic immune responses.
Collapse
Affiliation(s)
- Bailey J Didriksen
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily M Eshleman
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Theresa Alenghat
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
6
|
Forsyth CB, Shaikh M, Engen PA, Preuss F, Naqib A, Palmen BA, Green SJ, Zhang L, Bogin ZR, Lawrence K, Sharma D, Swanson GR, Bishehsari F, Voigt RM, Keshavarzian A. Evidence that the loss of colonic anti-microbial peptides may promote dysbiotic Gram-negative inflammaging-associated bacteria in aging mice. FRONTIERS IN AGING 2024; 5:1352299. [PMID: 38501032 PMCID: PMC10945560 DOI: 10.3389/fragi.2024.1352299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/02/2024] [Indexed: 03/20/2024]
Abstract
Introduction: Aging studies in humans and mice have played a key role in understanding the intestinal microbiome and an increased abundance of "inflammaging" Gram-negative (Gn) bacteria. The mechanisms underlying this inflammatory profile in the aging microbiome are unknown. We tested the hypothesis that an aging-related decrease in colonic crypt epithelial cell anti-microbial peptide (AMP) gene expression could promote colonic microbiome inflammatory Gn dysbiosis and inflammaging. Methods: As a model of aging, C57BL/6J mice fecal (colonic) microbiota (16S) and isolated colonic crypt epithelial cell gene expression (RNA-seq) were assessed at 2 months (mth) (human: 18 years old; yo), 15 mth (human: 50 yo), and 25 mth (human: 84 yo). Informatics examined aging-related microbial compositions, differential colonic crypt epithelial cell gene expressions, and correlations between colonic bacteria and colonic crypt epithelial cell gene expressions. Results: Fecal microbiota exhibited significantly increased relative abundances of pro-inflammatory Gn bacteria with aging. Colonic crypt epithelial cell gene expression analysis showed significant age-related downregulation of key AMP genes that repress the growth of Gn bacteria. The aging-related decrease in AMP gene expressions is significantly correlated with an increased abundance in Gn bacteria (dysbiosis), loss of colonic barrier gene expression, and senescence- and inflammation-related gene expression. Conclusion: This study supports the proposed model that aging-related loss of colonic crypt epithelial cell AMP gene expression promotes increased relative abundances of Gn inflammaging-associated bacteria and gene expression markers of colonic inflammaging. These data may support new targets for aging-related therapies based on intestinal genes and microbiomes.
Collapse
Affiliation(s)
- Christopher B. Forsyth
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Phillip A. Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Fabian Preuss
- Department of Biological Sciences, University of Wisconsin Parkside, Kenosha, WI, United States
| | - Ankur Naqib
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Breanna A. Palmen
- Department of Biological Sciences, University of Wisconsin Parkside, Kenosha, WI, United States
| | - Stefan J. Green
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Lijuan Zhang
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Zlata R. Bogin
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Kristi Lawrence
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Deepak Sharma
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Garth R. Swanson
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Faraz Bishehsari
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
7
|
DeSana AJ, Estus S, Barrett TA, Saatman KE. Acute gastrointestinal permeability after traumatic brain injury in mice precedes a bloom in Akkermansia muciniphila supported by intestinal hypoxia. Sci Rep 2024; 14:2990. [PMID: 38316862 PMCID: PMC10844296 DOI: 10.1038/s41598-024-53430-4] [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: 11/08/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024] Open
Abstract
Traumatic brain injury (TBI) increases gastrointestinal morbidity and associated mortality. Clinical and preclinical studies implicate gut dysbiosis as a consequence of TBI and an amplifier of brain damage. However, little is known about the association of gut dysbiosis with structural and functional changes of the gastrointestinal tract after an isolated TBI. To assess gastrointestinal dysfunction, mice received a controlled cortical impact or sham brain injury and intestinal permeability was assessed at 4 h, 8 h, 1 d, and 3 d after injury by oral administration of 4 kDa FITC Dextran prior to euthanasia. Quantification of serum fluorescence revealed an acute, short-lived increase in permeability 4 h after TBI. Despite transient intestinal dysfunction, no overt morphological changes were evident in the ileum or colon across timepoints from 4 h to 4 wks post-injury. To elucidate the timeline of microbiome changes after TBI, 16 s gene sequencing was performed on DNA extracted from fecal samples collected prior to and over the first month after TBI. Differential abundance analysis revealed that the phylum Verrucomicrobiota was increased at 1, 2, and 3 d after TBI. The Verrucomicrobiota species was identified by qPCR as Akkermansia muciniphila, an obligate anaerobe that resides in the intestinal mucus bilayer and produces short chain fatty acids (e.g. butyrate) utilized by intestinal epithelial cells. We postulated that TBI promotes intestinal changes favorable for the bloom of A. muciniphila. Consistent with this premise, the relative area of mucus-producing goblet cells in the medial colon was significantly increased at 1 d after injury, while colon hypoxia was significantly increased at 3 d. Our findings reveal acute gastrointestinal functional changes coupled with an increase of beneficial bacteria suggesting a potential compensatory response to systemic stress after TBI.
Collapse
Affiliation(s)
- Anthony J DeSana
- Department of Physiology, University of Kentucky, Biomedical and Biological Sciences Research Building (BBSRB), B473, 741 South Limestone St., Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Biomedical and Biological Sciences Research Building (BBSRB), B473, 741 South Limestone St., Lexington, KY, 40536, USA
| | - Steven Estus
- Department of Physiology, University of Kentucky, Biomedical and Biological Sciences Research Building (BBSRB), B473, 741 South Limestone St., Lexington, KY, 40536, USA
- Sanders Brown Center on Aging, University of Kentucky, Lee T. Todd, Jr. Building, Rm: 537, 789 South Limestone St., Lexington, KY, 40536, USA
| | - Terrence A Barrett
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine - Digestive Health, University of Kentucky, Lexington, KY, 40536, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Medical Science Building, MN649, 780 Rose St., Lexington, KY, 40536, USA
| | - Kathryn E Saatman
- Department of Physiology, University of Kentucky, Biomedical and Biological Sciences Research Building (BBSRB), B473, 741 South Limestone St., Lexington, KY, 40536, USA.
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Biomedical and Biological Sciences Research Building (BBSRB), B473, 741 South Limestone St., Lexington, KY, 40536, USA.
| |
Collapse
|
8
|
Schumacher MA. The emerging roles of deep crypt secretory cells in colonic physiology. Am J Physiol Gastrointest Liver Physiol 2023; 325:G493-G500. [PMID: 37697924 PMCID: PMC10887841 DOI: 10.1152/ajpgi.00093.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/18/2023] [Accepted: 09/03/2023] [Indexed: 09/13/2023]
Abstract
Deep crypt secretory (DCS) cells are a population of epithelial cells located at the colonic crypt base that share some similarities to Paneth and goblet cells. They were initially defined as c-Kit expressing cells, though subsequent work showed that they are more specifically marked by Reg4 in the murine colon. The best-understood function of DCS cells at present is supporting the stem cell niche by generating Notch and EGF ligands. However, as these cells also express immunoregulatory (e.g., Ccl6) and host defense (e.g., Retnlb) genes, it is likely they have additional functions in maintaining colonic health outside of maintenance of the stem niche. Recent advances in single-cell transcriptomic profiling hint at additional epithelial and immune roles that may exist for these cells and have aided in elucidating their developmental lineage. This review highlights the emerging evidence supporting a crucial role for DCS cells in intestinal physiology, the current understanding of how these cells are regulated, and their potential role(s) in colonic disease.
Collapse
Affiliation(s)
- Michael A Schumacher
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California, United States
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States
| |
Collapse
|
9
|
Mules TC, Inns S, Le Gros G. Helminths' therapeutic potential to treat intestinal barrier dysfunction. Allergy 2023; 78:2892-2905. [PMID: 37449458 DOI: 10.1111/all.15812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
The intestinal barrier is a dynamic multi-layered structure which can adapt to environmental changes within the intestinal lumen. It has the complex task of allowing nutrient absorption while limiting entry of harmful microbes and microbial antigens present in the intestinal lumen. Excessive entry of microbial antigens via microbial translocation due to 'intestinal barrier dysfunction' is hypothesised to contribute to the increasing incidence of allergic, autoimmune and metabolic diseases, a concept referred to as the 'epithelial barrier theory'. Helminths reside in the intestinal tract are in intimate contact with the mucosal surfaces and induce a range of local immunological changes which affect the layers of the intestinal barrier. Helminths are proposed to prevent, or even treat, many of the diseases implicated in the epithelial barrier theory. This review will focus on the effect of helminths on intestinal barrier function and explore whether this could explain the proposed health benefits delivered by helminths.
Collapse
Affiliation(s)
- Thomas C Mules
- Malaghan Institute of Medical Research, Wellington, New Zealand
- University of Otago, Wellington, New Zealand
| | | | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
10
|
Vornewald PM, Forman R, Yao R, Parmar N, Lindholm HT, Lee LSK, Martín-Alonso M, Else KJ, Oudhoff MJ. Mmp17-deficient mice exhibit heightened goblet cell effector expression in the colon and increased resistance to chronic Trichuris muris infection. Front Immunol 2023; 14:1243528. [PMID: 37869014 PMCID: PMC10587605 DOI: 10.3389/fimmu.2023.1243528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Intestinal epithelial homeostasis is maintained by intrinsic and extrinsic signals. The extrinsic signals include those provided by mesenchymal cell populations that surround intestinal crypts and is further facilitated by the extracellular matrix (ECM), which is modulated by proteases such as matrix metalloproteinases (MMPs). Extrinsic signals ensure an appropriate balance between intestinal epithelial proliferation and differentiation. This study explores the role of MMP17, which is preferentially expressed by smooth muscle cells in the intestine, in intestinal homeostasis and during immunity to infection. Mice lacking MMP17 expressed high levels of goblet-cell associated genes and proteins, such as CLCA1 and RELM-β, which are normally associated with immune responses to infection. Nevertheless, Mmp17 KO mice did not have altered resistance during a bacterial Citrobacter rodentium infection. However, when challenged with a low dose of the helminth Trichuris muris, Mmp17 KO mice had increased resistance, without a clear role for an altered immune response during infection. Mechanistically, we did not find changes in traditional modulators of goblet cell effectors such as the NOTCH pathway or specific cytokines. We found MMP17 expression in smooth muscle cells as well as lamina propria cells such as macrophages. Together, our data suggest that MMP17 extrinsically alters goblet cell maturation which is sufficient to alter clearance in a helminth infection model.
Collapse
Affiliation(s)
- Pia M. Vornewald
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Ruth Forman
- Lydia Becker Institute of Immunology & Inflammation, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Rouan Yao
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Naveen Parmar
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard T. Lindholm
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Lilith S. K. Lee
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Mara Martín-Alonso
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Kathryn J. Else
- Lydia Becker Institute of Immunology & Inflammation, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Menno J. Oudhoff
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
11
|
Maizels RM, Gause WC. Targeting helminths: The expanding world of type 2 immune effector mechanisms. J Exp Med 2023; 220:e20221381. [PMID: 37638887 PMCID: PMC10460967 DOI: 10.1084/jem.20221381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/24/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
In this new review, Rick Maizels and Bill Gause summarize how type 2 immune responses combat helminth parasites through novel mechanisms, coordinating multiple innate and adaptive cell and molecular players that can eliminate infection and repair-resultant tissue damage.
Collapse
Affiliation(s)
- Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - William C. Gause
- Center for Immunity and Inflammation, Rutgers Biomedical Health Sciences Institute for Infectious and Inflammatory Diseases, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, USA
| |
Collapse
|
12
|
Zaini A, Dalit L, Sheikh AA, Zhang Y, Thiele D, Runting J, Rodrigues G, Ng J, Bramhall M, Scheer S, Hailes L, Groom JR, Good-Jacobson KL, Zaph C. Heterogeneous Tfh cell populations that develop during enteric helminth infection predict the quality of type 2 protective response. Mucosal Immunol 2023; 16:642-657. [PMID: 37392971 DOI: 10.1016/j.mucimm.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/03/2023]
Abstract
T follicular helper (Tfh) cells are an important component of germinal center (GC)-mediated humoral immunity. Yet, how a chronic type 1 versus protective type 2 helminth infection modulates Tfh-GC responses remains poorly understood. Here, we employ the helminth Trichuris muris model and demonstrate that Tfh cell phenotypes and GC are differentially regulated in acute versus chronic infection. The latter failed to induce Tfh-GC B cell responses, with Tfh cells expressing Τ-bet and interferon-γ. In contrast, interleukin-4-producing Tfh cells dominate responses to an acute, resolving infection. Heightened expression and increased chromatin accessibility of T helper (Th)1- and Th2 cell-associated genes are observed in chronic and acute induced Tfh cells, respectively. Blockade of the Th1 cell response by T-cell-intrinsic T-bet deletion promoted Tfh cell expansion during chronic infection, pointing to a correlation between a robust Tfh cell response and protective immunity to parasites. Finally, blockade of Tfh-GC interactions impaired type 2 immunity, revealing the critical protective role of GC-dependent Th2-like Tfh cell responses during acute infection. Collectively, these results provide new insights into the protective roles of Tfh-GC responses and identify distinct transcriptional and epigenetic features of Tfh cells that emerge during resolving or chronic T. muris infection.
Collapse
Affiliation(s)
- Aidil Zaini
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia; Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, Australia
| | - Lennard Dalit
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Amania A Sheikh
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Yan Zhang
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Daniel Thiele
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Microbiology, Monash University, Clayton, Australia
| | - Jessica Runting
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Grace Rodrigues
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Judy Ng
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Michael Bramhall
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Sebastian Scheer
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Lauren Hailes
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia
| | - Joanna R Groom
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Kim L Good-Jacobson
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia.
| | - Colby Zaph
- Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia; Department of Biochemistry and Molecular Biology, Clayton, Australia.
| |
Collapse
|
13
|
Fung C, Fraser L, Barrón G, Gologorsky M, Atkinson S, Gerrick E, Hayward M, Ziegelbauer J, Li J, Nico K, Tyner M, DeSchepper L, Pan A, Salzman N, Howitt M. Tuft cells mediate commensal remodeling of the small intestinal antimicrobial landscape. Proc Natl Acad Sci U S A 2023; 120:e2216908120. [PMID: 37253002 PMCID: PMC10266004 DOI: 10.1073/pnas.2216908120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/19/2023] [Indexed: 06/01/2023] Open
Abstract
Succinate produced by the commensal protist Tritrichomonas musculis (T. mu) stimulates chemosensory tuft cells, resulting in intestinal type 2 immunity. Tuft cells express the succinate receptor SUCNR1, yet this receptor does not mediate antihelminth immunity nor alter protist colonization. Here, we report that microbial-derived succinate increases Paneth cell numbers and profoundly alters the antimicrobial peptide (AMP) landscape in the small intestine. Succinate was sufficient to drive this epithelial remodeling, but not in mice lacking tuft cell chemosensory components required to detect this metabolite. Tuft cells respond to succinate by stimulating type 2 immunity, leading to interleukin-13-mediated epithelial and AMP expression changes. Moreover, type 2 immunity decreases the total number of mucosa-associated bacteria and alters the small intestinal microbiota composition. Finally, tuft cells can detect short-term bacterial dysbiosis that leads to a spike in luminal succinate levels and modulate AMP production in response. These findings demonstrate that a single metabolite produced by commensals can markedly shift the intestinal AMP profile and suggest that tuft cells utilize SUCNR1 and succinate sensing to modulate bacterial homeostasis.
Collapse
Affiliation(s)
- Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Lisa M. Fraser
- Division of Gastroenterology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI53226
| | - Gabriel M. Barrón
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | | | - Samantha N. Atkinson
- Department of Microbiology and Immunology, Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI53226
| | - Elias R. Gerrick
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Michael Hayward
- Division of Gastroenterology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI53226
- Department of Microbiology and Immunology, Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI53226
| | - Jennifer Ziegelbauer
- Department of Microbiology and Immunology, Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI53226
| | - Jessica A. Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Katherine F. Nico
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Miles D. W. Tyner
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Leila B. DeSchepper
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Amy Pan
- Department of Microbiology and Immunology, Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI53226
- Division of Quantitative Health Services, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI53226
| | - Nita H. Salzman
- Division of Gastroenterology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI53226
- Department of Microbiology and Immunology, Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI53226
| | - Michael R. Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
14
|
Saad AE, Ashour DS, Rashad E. Immunomodulatory effects of chronic trichinellosis on Toxoplasma gondii RH virulent strain in experimental rats. Pathog Glob Health 2023; 117:417-434. [PMID: 36922743 PMCID: PMC10177679 DOI: 10.1080/20477724.2023.2191233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Mixed parasitic infections could affect the host immunological responses and re-design the pathogenesis of each other. The impact of Toxoplasma gondii (T. gondii) and Trichinella spiralis (T. spiralis) co-infection on the immune response remains unclear. The objective of the present study was to investigate the possible effect of chronic trichinellosis on the immune response of rats infected with T. gondii virulent RH strain. Animals were divided into four groups: group I: non-infected negative control; group II: infected with T. spiralis; group III: infected with T. gondii and group IV: infected with T. spiralis then infected with T. gondii 35 days post T. spiralis infection (co-infected group). The interaction between T. spiralis and T. gondii was evaluated by histopathological examination of liver and brain tissues, immunohistochemical expression of inducible nitric oxide synthase (iNOS), and β-catenin in the brain tissues, and CD4+ and CD8+ T cells percentages, and tumor necrosis factor (TNF)-alpha expression in the spleen tissues. Along with, splenic interleukin (IL)-4 and IL-10 mRNA expression levels were measured 15 days post-Toxoplasma infection. Our study revealed that prior infection with T. spiralis leads to attenuation of Th1 response against T. gondii, including iNOS, TNF-α, and CD8+ T-cell response with improvement of the histopathological changes in the tissues. In conclusion, in the co-infected rats, a balanced immune response has been developed with the end result, improvement of the histopathological changes in the liver and brain.
Collapse
Affiliation(s)
- Abeer E. Saad
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
- Medical Parasitology sub-unit, Pathology Department, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Dalia S. Ashour
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman Rashad
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
15
|
Abstract
Just as mammals have coevolved with the intestinal bacterial communities that are part of the microbiota, intestinal helminths represent an important selective force on their mammalian host. The complex interaction between helminths, microbes, and their mammalian host is likely an important determinant of mutual fitness. The host immune system in particular is a critical interface with both helminths and the microbiota, and this crosstalk often determines the balance between tolerance and resistance against these widespread parasites. Hence, there are many examples of how both helminths and the microbiota can influence tissue homeostasis and homeostatic immunity. Understanding these processes at a cellular and molecular level is an exciting area of research that we seek to highlight in this review and that will potentially guide future treatment approaches.
Collapse
Affiliation(s)
- P'ng Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia.
| |
Collapse
|
16
|
Jian H, Liu Y, Wang X, Dong X, Zou X. Akkermansia muciniphila as a Next-Generation Probiotic in Modulating Human Metabolic Homeostasis and Disease Progression: A Role Mediated by Gut-Liver-Brain Axes? Int J Mol Sci 2023; 24:ijms24043900. [PMID: 36835309 PMCID: PMC9959343 DOI: 10.3390/ijms24043900] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Appreciation of the importance of Akkermansia muciniphila is growing, and it is becoming increasingly relevant to identify preventive and/or therapeutic solutions targeting gut-liver-brain axes for multiple diseases via Akkermansia muciniphila. In recent years, Akkermansia muciniphila and its components such as outer membrane proteins and extracellular vesicles have been known to ameliorate host metabolic health and intestinal homeostasis. However, the impacts of Akkermansia muciniphila on host health and disease are complex, as both potentially beneficial and adverse effects are mediated by Akkermansia muciniphila and its derivatives, and in some cases, these effects are dependent upon the host physiology microenvironment and the forms, genotypes, and strain sources of Akkermansia muciniphila. Therefore, this review aims to summarize the current knowledge of how Akkermansia muciniphila interacts with the host and influences host metabolic homeostasis and disease progression. Details of Akkermansia muciniphila will be discussed including its biological and genetic characteristics; biological functions including anti-obesity, anti-diabetes, anti-metabolic-syndrome, anti-inflammation, anti-aging, anti-neurodegenerative disease, and anti-cancer therapy functions; and strategies to elevate its abundance. Key events will be referred to in some specific disease states, and this knowledge should facilitate the identification of Akkermansia muciniphila-based probiotic therapy targeting multiple diseases via gut-liver-brain axes.
Collapse
|
17
|
Fernández-Lainez C, Aan de Stegge M, Silva-Lagos LA, López-Velázquez G, de Vos P. β(2 → 1)-β(2 → 6) branched graminan-type fructans and β(2 → 1) linear fructans impact mucus-related and endoplasmic reticulum stress-related genes in goblet cells and attenuate inflammatory responses in a fructan dependent fashion. Food Funct 2023; 14:1338-1348. [PMID: 36656019 DOI: 10.1039/d2fo02710k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Dietary fibers such as fructans have beneficial effects on intestinal health but it is unknown whether they impact goblet cells (GCs). Here we studied the effects of inulin-type fructans (ITFs) and graminan-type fructans (GTFs) with different molecular weights on mucus- and endoplasmic reticulum (ER) stress-related genes in intestinal GCs. To that end, GCs were incubated in the presence of ITFs or GTFs, or ITFs and GTFs + TNFα or the N-glycosylation inhibitor tunicamycin (Tm). IL-8 production by GCs was studied as a marker of inflammation. Effects between ITFs and GTFs were compared. We found a beneficial impact of GTFs especially on the expression of RETNLB. GTF II protects from the TNFα-induced gene expression dysregulation of MUC2, TFF3, GAL3ST2, and CHST5. Also, all the studied fructans prevented Tm-induced dysregulation of GAL3ST2. Interestingly, only the short chain fructans ITF I and GTF I have anti-inflammatory properties on GCs. All the studied fructans except ITF I decreased the expression of the ER stress-related HSPA5 and XBP1. All these benefits were fructan-structure and chain length dependent. Our study contributes to a better understanding of chemical structure-dependent beneficial effects of ITFs and GTFs on gut barrier function, which could contribute to prevention of gut inflammatory disorders.
Collapse
Affiliation(s)
- Cynthia Fernández-Lainez
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands. .,Laboratorio de Errores innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Av. Iman 1, 04530, Ciudad de México, Mexico.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México. Edificio D, 1° Piso. Circuito de Posgrados, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Myrthe Aan de Stegge
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Luis Alfredo Silva-Lagos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Gabriel López-Velázquez
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Av. Iman 1, 04530, Cuidad de México, Mexico.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
18
|
Resistin-like beta reduction is associated to low survival rate and is downregulated by adjuvant therapy in colorectal cancer patients. Sci Rep 2023; 13:1490. [PMID: 36707698 PMCID: PMC9883247 DOI: 10.1038/s41598-023-28450-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Colorectal Cancer (CRC) is one of the most common cancers accounting for 1.8 million new cases worldwide every year. Therefore, the identification of new potential therapeutic targets represents a continuous challenge to improve survival and quality of CRC patient's life. We performed a microarray analysis dataset consisting of colon biopsies of healthy subjects (HS) and CRC patients. These results were further confirmed in a clinical setting evaluating a series of CRC patients to assess the expression of Resistin-Like Beta (RETNLB) and to correlate it with their clinical data. Our results showed a significant reduction of RETNLB expression in CRC biopsies compared to the HS mucosa. Furthermore, such reduction was significantly associated with the TNM grade and patients' age. Furthermore, a significantly positive correlation was found within mutated subjects for KRAS, TP53, and BRAF. In particular, patients with poor prognosis at 5 years exhibited RETNLB lower levels. In-silico analysis data were confirmed by histochemical analysis in a series of CRC patients recruited by our group. The results obtained provided that RETNLB low levels are associated with an unfavorable prognosis in CRC patients and its expression is also dependent on adjuvant therapy. Further studies are warranted in order to evaluate the molecular mechanisms underlying the role of RETNLB in CRC progression.
Collapse
|
19
|
Shi Y, Zhu N, Qiu Y, Tan J, Wang F, Qin L, Dai A. Resistin-like molecules: a marker, mediator and therapeutic target for multiple diseases. Cell Commun Signal 2023; 21:18. [PMID: 36691020 PMCID: PMC9869618 DOI: 10.1186/s12964-022-01032-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/25/2023] Open
Abstract
Resistin-like molecules (RELMs) are highly cysteine-rich proteins, including RELMα, RELMβ, Resistin, and RELMγ. However, RELMs exhibit significant differences in structure, distribution, and function. The expression of RELMs is regulated by various signaling molecules, such as IL-4, IL-13, and their receptors. In addition, RELMs can mediate numerous signaling pathways, including HMGB1/RAGE, IL-4/IL-4Rα, PI3K/Akt/mTOR signaling pathways, and so on. RELMs proteins are involved in wide range of physiological and pathological processes, including inflammatory response, cell proliferation, glucose metabolism, barrier defense, etc., and participate in the progression of numerous diseases such as lung diseases, intestinal diseases, cardiovascular diseases, and cancers. Meanwhile, RELMs can serve as biomarkers, risk predictors, and therapeutic targets for these diseases. An in-depth understanding of the role of RELMs may provide novel targets or strategies for the treatment and prevention of related diseases. Video abstract.
Collapse
Affiliation(s)
- Yaning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China
| | - Yun Qiu
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Junlan Tan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China
| | - Feiying Wang
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China.
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Medicine, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China.
| |
Collapse
|
20
|
Schumacher MA, Liu CY, Katada K, Thai MH, Hsieh JJ, Hansten BJ, Waddell A, Rosen MJ, Frey MR. Deep Crypt Secretory Cell Differentiation in the Colonic Epithelium Is Regulated by Sprouty2 and Interleukin 13. Cell Mol Gastroenterol Hepatol 2022; 15:971-984. [PMID: 36414210 PMCID: PMC9982040 DOI: 10.1016/j.jcmgh.2022.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND & AIMS Deep crypt secretory (DCS) cells are a critical component of the colonic stem cell niche. However, the regulatory mechanisms controlling DCS cell numbers and function are not well understood. Sprouty2 is an inflammation-responsive regulator of intracellular signaling that influences colonic secretory cell numbers in colitis via an epithelial-stromal interleukin (IL)33/IL13 signaling loop. Here, we tested the hypothesis that IL13, induced by epithelial Sprouty2 down-regulation, promotes DCS cell differentiation and function. METHODS Distal colons from mice with an intestinal epithelial-specific Sprouty2 deletion (Spry2ΔIE) and littermate controls were analyzed by in situ hybridization for Reg4+ DCS cells. Single-cell RNA sequencing and immunostaining were used to identify DCS cell-derived host defense peptides (HDPs) and localization of IL13 and IL13 receptor; bulk RNA sequencing and quantitative polymerase chain reaction were used to quantify changes in expression of identified HDPs. Cytokine-treated colonoids were assessed for DCS cells. A requirement for an IL33/IL13 signaling loop in the regulation of DCS cells was assessed in vivo using IL13 null mice. RESULTS Reg4+ DCS cell numbers were increased 2-fold in distal colons of Spry2ΔIE mice with a concomitant overall increase in DCS cell marker expression (Reg4, Spink4, and Agr2). Single-cell transcriptomics showed the HDP Retnlb/Resistin Like Beta (RELMβ) is highly enriched in DCS cells. Retnlb/RELMβ expression was increased in Spry2ΔIE colons. IL13, but not IL33, induced Reg4 and Retnlb expression in colonic epithelial organoids, and IL33-mediated expansion of the DCS cell population in vivo was dependent on IL13, which was expressed predominantly by type II innate lymphoid cells in the colonic mucosa. CONCLUSIONS Sprouty2 limits colonic DCS cell differentiation through suppression of IL13 signaling. At homeostasis, DCS cells are marked by high levels of the HDP RELMβ. Loss of epithelial Sprouty2 activates type II innate lymphoid cells to release IL13, promoting expansion of the DCS cell population and increased colonic RELMβ levels.
Collapse
Affiliation(s)
- Michael A Schumacher
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.
| | - Cambrian Y Liu
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Kay Katada
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Megan H Thai
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Jonathan J Hsieh
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Britany J Hansten
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Amanda Waddell
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael J Rosen
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Mark R Frey
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; Department of Biochemistry and Molecular Medicine, University of Southern California Keck School of Medicine, Los Angeles, California.
| |
Collapse
|
21
|
Colombo SAP, Thompson S, Bancroft AJ, Grencis RK. Anti-Trichuris mucosal responses are maintained during H. bakeri co-infection despite impaired parasite expulsion. Parasite Immunol 2022; 44:e12936. [PMID: 35586956 PMCID: PMC9542692 DOI: 10.1111/pim.12936] [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: 02/25/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022]
Abstract
In endemic regions concurrent infection with multiple gastrointestinal (GI) helminth species is more common than single species infection. However, the majority of model helminth infections focus on single species infections leading to a lack of understanding of how co-infection influences anti-parasite immune responses. Here, we use a model co-infection of Trichuris muris (Tm) and Heligmosomoides bakeri (Hb) to investigate the effect of Hb on anti-Tm immune responses. We observed a complete impairment of Tm expulsion in immune competent C57BL/6 mice when co-infected with Hb. This was coupled with reduced cellularity in the colonic mesenteric lymph node (cMLN) proximal to the caecum, however, cMLN cytokine responses and caecal mucosal immune responses in co-infected mice were not significantly different from mice infected with Tm alone. Interestingly, in immune-compromised mice, we found co-infection resulted in enhanced growth and fecundity of female Tm parasites. These data suggest that during helminth-helminth co-infection, immune-independent signals between species may promote survival and growth.
Collapse
Affiliation(s)
- Stefano A. P. Colombo
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, University of ManchesterManchesterUK
| | - Seona Thompson
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, University of ManchesterManchesterUK
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, University of ManchesterManchesterUK
| | - Allison J. Bancroft
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, University of ManchesterManchesterUK
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, University of ManchesterManchesterUK
| | - Richard K. Grencis
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, University of ManchesterManchesterUK
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, University of ManchesterManchesterUK
| |
Collapse
|
22
|
Rahman MM, Islam F, -Or-Rashid MH, Mamun AA, Rahaman MS, Islam MM, Meem AFK, Sutradhar PR, Mitra S, Mimi AA, Emran TB, Fatimawali, Idroes R, Tallei TE, Ahmed M, Cavalu S. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front Cell Infect Microbiol 2022; 12:903570. [PMID: 35795187 PMCID: PMC9251340 DOI: 10.3389/fcimb.2022.903570] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 12/11/2022] Open
Abstract
In the last two decades, considerable interest has been shown in understanding the development of the gut microbiota and its internal and external effects on the intestine, as well as the risk factors for cardiovascular diseases (CVDs) such as metabolic syndrome. The intestinal microbiota plays a pivotal role in human health and disease. Recent studies revealed that the gut microbiota can affect the host body. CVDs are a leading cause of morbidity and mortality, and patients favor death over chronic kidney disease. For the function of gut microbiota in the host, molecules have to penetrate the intestinal epithelium or the surface cells of the host. Gut microbiota can utilize trimethylamine, N-oxide, short-chain fatty acids, and primary and secondary bile acid pathways. By affecting these living cells, the gut microbiota can cause heart failure, atherosclerosis, hypertension, myocardial fibrosis, myocardial infarction, and coronary artery disease. Previous studies of the gut microbiota and its relation to stroke pathogenesis and its consequences can provide new therapeutic prospects. This review highlights the interplay between the microbiota and its metabolites and addresses related interventions for the treatment of CVDs.
Collapse
|
23
|
Inclan-Rico JM, Rossi HL, Herbert DR. "Every cell is an immune cell; contributions of non-hematopoietic cells to anti-helminth immunity". Mucosal Immunol 2022; 15:1199-1211. [PMID: 35538230 PMCID: PMC9646929 DOI: 10.1038/s41385-022-00518-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
Helminths are remarkably successful parasites that can invade various mammalian hosts and establish chronic infections that can go unnoticed for years despite causing severe tissue damage. To complete their life cycles, helminths migrate through multiple barrier sites that are densely populated by a complex array of hematopoietic and non-hematopoietic cells. While it is clear that type 2 cytokine responses elicited by immune cells promote worm clearance and tissue healing, the actions of non-hematopoietic cells are increasingly recognized as initiators, effectors and regulators of anti-helminth immunity. This review will highlight the collective actions of specialized epithelial cells, stromal niches, stem, muscle and neuroendocrine cells as well as peripheral neurons in the detection and elimination of helminths at mucosal sites. Studies dissecting the interactions between immune and non-hematopoietic cells will truly provide a better understanding of the mechanisms that ensure homeostasis in the context of helminth infections.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather L Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Wegierska AE, Charitos IA, Topi S, Potenza MA, Montagnani M, Santacroce L. The Connection Between Physical Exercise and Gut Microbiota: Implications for Competitive Sports Athletes. Sports Med 2022; 52:2355-2369. [PMID: 35596883 PMCID: PMC9474385 DOI: 10.1007/s40279-022-01696-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2022] [Indexed: 12/16/2022]
Abstract
Gut microbiota refers to those microorganisms in the human digestive tract that display activities fundamental in human life. With at least 4 million different bacterial types, the gut microbiota is composed of bacteria that are present at levels sixfold greater than the total number of cells in the entire human body. Among its multiple functions, the microbiota helps promote the bioavailability of some nutrients and the metabolization of food, and protects the intestinal mucosa from the aggression of pathogenic microorganisms. Moreover, by stimulating the production of intestinal mediators able to reach the central nervous system (gut/brain axis), the gut microbiota participates in the modulation of human moods and behaviors. Several endogenous and exogenous factors can cause dysbiosis with important consequences on the composition and functions of the microbiota. Recent research underlines the importance of appropriate physical activity (such as sports), nutrition, and a healthy lifestyle to ensure the presence of a functional physiological microbiota working to maintain the health of the whole human organism. Indeed, in addition to bowel disturbances, variations in the qualitative and quantitative microbial composition of the gastrointestinal tract might have systemic negative effects. Here, we review recent studies on the effects of physical activity on gut microbiota with the aim of identifying potential mechanisms by which exercise could affect gut microbiota composition and function. Whether physical exercise of variable work intensity might reflect changes in intestinal health is analyzed.
Collapse
Affiliation(s)
- Angelika Elzbieta Wegierska
- Interdisciplinary Department of Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy.,Italian Athletics Federation (FIDAL), Rome, Italy
| | - Ioannis Alexandros Charitos
- Emergency/Urgent Department, National Poisoning Center, Riuniti University Hospital of Foggia, Foggia, Italy
| | - Skender Topi
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan "A. Xhuvani", Elbasan, Albania
| | - Maria Assunta Potenza
- Department of Biomedical Sciences and Human Oncology-Section of Pharmacology, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology-Section of Pharmacology, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, p.zza G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
25
|
Wu X, Gu B, Yang H. The role of γδ T cells in the interaction between commensal and pathogenic bacteria in the intestinal mucosa. Int Rev Immunol 2022; 42:379-392. [PMID: 35583374 DOI: 10.1080/08830185.2022.2076846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 12/22/2022]
Abstract
The intestinal mucosa is an important structure involved in resistance to pathogen infection. It is mainly composed of four barriers, which have different but interrelated functions. Pathogenic bacteria can damage these intestinal mucosal barriers. Here, we mainly review the mechanisms of pathogen damage to biological barriers. Most γδ T cells are located on the surface of the intestinal mucosa, with the ability to migrate and engage in crosstalk with microorganisms. Commensal bacteria are involved in the activation and migration of γδ T cells to monitor the invasion of pathogens. Pathogen invasion alters the migration pattern of γδ T cells. γδ T cells accelerate pathogen clearance and limit opportunistic invasion of commensal bacteria. By discussing these interactions among γδ T cells, commensal bacteria and pathogenic bacteria, we suggest that γδ T cells may link the interactions between commensal bacteria and pathogenic bacteria.
Collapse
Affiliation(s)
- Xiaoxiao Wu
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bing Gu
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
26
|
Lindholm HT, Parmar N, Drurey C, Poveda MC, Vornewald P, Ostrop J, Díez-Sanchez A, Maizels RM, Oudhoff MJ. BMP signaling in the intestinal epithelium drives a critical feedback loop to restrain IL-13-driven tuft cell hyperplasia. Sci Immunol 2022; 7:eabl6543. [PMID: 35559665 PMCID: PMC7614132 DOI: 10.1126/sciimmunol.abl6543] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The intestinal tract is a common site for various types of infections including viruses, bacteria, and helminths, each requiring specific modes of immune defense. The intestinal epithelium has a pivotal role in both immune initiation and effector stages, which are coordinated by lymphocyte cytokines such as IFNγ, IL-13, and IL-22. Here, we studied intestinal epithelial immune responses using organoid image analysis based on a convolutional neural network, transcriptomic analysis, and in vivo infection models. We found that IL-13 and IL-22 both induce genes associated with goblet cells, but the resulting goblet cell phenotypes are dichotomous. Moreover, only IL-13-driven goblet cells are associated with classical NOTCH signaling. We further showed that IL-13 induces the bone morphogenetic protein (BMP) pathway, which acts in a negative feedback loop on immune type 2-driven tuft cell hyperplasia. This is associated with inhibiting Sox4 expression to putatively limit the tuft cell progenitor population. Blocking ALK2, a BMP receptor, with the inhibitor dorsomorphin homolog 1 (DMH1) interrupted the feedback loop, resulting in greater tuft cell numbers both in vitro and in vivo after infection with Nippostrongylus brasiliensis. Together, this investigation of cytokine effector responses revealed an unexpected and critical role for the BMP pathway in regulating type 2 immunity, which can be exploited to tailor epithelial immune responses.
Collapse
Affiliation(s)
- Håvard T. Lindholm
- CEMIR - Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Naveen Parmar
- CEMIR - Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunology and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Marta Campillo Poveda
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunology and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Pia Vornewald
- CEMIR - Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Jenny Ostrop
- CEMIR - Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Alberto Díez-Sanchez
- CEMIR - Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunology and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Menno J. Oudhoff
- CEMIR - Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
27
|
Liu XC, Guo L, Ling KR, Hu XY, Shen YJ, Li LQ. Serum Relmβ combined with abdominal signs may predict surgical timing in neonates with NEC: A cohort study. Front Pediatr 2022; 10:943320. [PMID: 36147817 PMCID: PMC9485553 DOI: 10.3389/fped.2022.943320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS To examine the predictive value of serum biomarkers combined with other indicators for necrotizing enterocolitis (NEC) surgery decision-making. METHODS Clinical data, including baseline information, clinical features, imaging presentation and serum assessment, of the infants enrolled were collected, and the serum concentrations of HBD2, HMGB-1, Claudin-3 and Relmβ were determined. Student's t test, the Mann-Whitney U test, the chi-square test and logistic regression analysis were used. Receiver operating characteristic (ROC) curves were also generated. RESULTS Forty-nine infants were enrolled, with 23 in the surgical NEC group and 26 in the medical NEC group. There were no differences in the baseline clinical information, including birth weight, gestational age, admission age and risk factors, during pregnancy and before enrollment (P > 0.05). Peritonitis, intestinal adhesion and sepsis were more common in the surgical group (P < 0.05). The incidences of abdominal distention, abdominal wall tenseness, abdominal tenderness and absent bowel sounds in the surgical group were significantly higher when NEC occurred (P < 0.05). There were no differences between the two groups in the imaging presentation (P > 0.05). The concentration of Relmβ {[8.66 (4.29, 19.28) vs. 20.65 (9.51, 44.65)]} in the surgical group was significantly higher (P < 0.05). Abdominal wall tenseness, abdominal tenderness and a Relmβ concentration > 19.7 μmol/L were included in the predictive model, and the AUC of the predictive score was 0.943 (95% CI: 0.891-1.000) (P < 0.05). CONCLUSION Serum Relmβ concentration combined with abdominal wall tenseness and abdominal tenderness may be useful in determining surgical timing in neonates with NEC.
Collapse
Affiliation(s)
- Xiao-Chen Liu
- Neonatal Diagnosis and Treatment Centre of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu Guo
- Neonatal Diagnosis and Treatment Centre of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ke-Ran Ling
- Neonatal Diagnosis and Treatment Centre of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiao-Yu Hu
- Neonatal Diagnosis and Treatment Centre of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu-Jie Shen
- Neonatal Diagnosis and Treatment Centre of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu-Quan Li
- Neonatal Diagnosis and Treatment Centre of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
28
|
El-Sahhar S, Varga-Weisz P. The gut microbiome in health and disease: Inflammatory bowel diseases. ADV ECOL RES 2022. [DOI: 10.1016/bs.aecr.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
29
|
Hu Z, Zhang C, Sifuentes-Dominguez L, Zarek CM, Propheter DC, Kuang Z, Wang Y, Pendse M, Ruhn KA, Hassell B, Behrendt CL, Zhang B, Raj P, Harris-Tryon TA, Reese TA, Hooper LV. Small proline-rich protein 2A is a gut bactericidal protein deployed during helminth infection. Science 2021; 374:eabe6723. [PMID: 34735226 DOI: 10.1126/science.abe6723] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A diverse group of antimicrobial proteins (AMPs) helps protect the mammalian intestine from varied microbial challenges. We show that small proline-rich protein 2A (SPRR2A) is an intestinal antibacterial protein that is phylogenetically unrelated to previously discovered mammalian AMPs. In this study, SPRR2A was expressed in Paneth cells and goblet cells and selectively killed Gram-positive bacteria by disrupting their membranes. SPRR2A shaped intestinal microbiota composition, restricted bacterial association with the intestinal surface, and protected against Listeria monocytogenes infection. SPRR2A differed from other intestinal AMPs in that it was induced by type 2 cytokines produced during helminth infection. Moreover, SPRR2A protected against helminth-induced bacterial invasion of intestinal tissue. Thus, SPRR2A is a distinctive AMP triggered by type 2 immunity that protects the intestinal barrier during helminth infection.
Collapse
Affiliation(s)
- Zehan Hu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chenlu Zhang
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Luis Sifuentes-Dominguez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christina M Zarek
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel C Propheter
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zheng Kuang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuhao Wang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mihir Pendse
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kelly A Ruhn
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Brian Hassell
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cassie L Behrendt
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bo Zhang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tamia A Harris-Tryon
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tiffany A Reese
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lora V Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
30
|
Hasani A, Ebrahimzadeh S, Hemmati F, Khabbaz A, Hasani A, Gholizadeh P. The role of Akkermansia muciniphila in obesity, diabetes and atherosclerosis. J Med Microbiol 2021; 70. [PMID: 34623232 DOI: 10.1099/jmm.0.001435] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alteration in the composition of the gut microbiota can lead to a number of chronic clinical diseases. Akkermansia muciniphila is an anaerobic bacteria constituting 3-5% of the gut microbial community in healthy adults. This bacterium is responsible for degenerating mucin in the gut; its scarcity leads to diverse clinical disorders. In this review, we focus on the role of A. muciniphila in diabetes, obesity and atherosclerosis, as well as the use of this bacterium as a next-generation probiotic. In regard to obesity and diabetes, human and animal trials have shown that A. muciniphila controls the essential regulatory system of glucose and energy metabolism. However, the underlying mechanisms by which A. muciniphila alleviates the complications of obesity, diabetes and atherosclerosis are unclear. At the same time, its abundance suggests improved metabolic disorders, such as metabolic endotoxemia, adiposity insulin resistance and glucose tolerance. The role of A. muciniphila is implicated in declining aortic lesions and atherosclerosis. Well-characterized virulence factors, antigens and cell wall extracts of A. muciniphila may act as effector molecules in these diseases. These molecules may provide novel mechanisms and strategies by which this bacterium could be used as a probiotic for the treatment of obesity, diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Alka Hasani
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Ebrahimzadeh
- Department of Food Science and Technology, Faculty of Agriculture, Urmia University, Urmia, Iran
| | - Fatemeh Hemmati
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aytak Khabbaz
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Hasani
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pourya Gholizadeh
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Yousefi Y, Haq S, Banskota S, Kwon YH, Khan WI. Trichuris muris Model: Role in Understanding Intestinal Immune Response, Inflammation and Host Defense. Pathogens 2021; 10:pathogens10080925. [PMID: 34451389 PMCID: PMC8399713 DOI: 10.3390/pathogens10080925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
Several parasites have evolved to survive in the human intestinal tract and over 1 billion people around the world, specifically in developing countries, are infected with enteric helminths. Trichuris trichiura is one of the world’s most common intestinal parasites that causes human parasitic infections. Trichuris muris, as an immunologically well-defined mouse model of T. trichiura, is extensively used to study different aspects of the innate and adaptive components of the immune system. Studies on T. muris model offer insights into understanding host immunity, since this parasite generates two distinct immune responses in resistant and susceptible strains of mouse. Apart from the immune cells, T. muris infection also influences various components of the intestinal tract, especially the gut microbiota, mucus layer, epithelial cells and smooth muscle cells. Here, we reviewed the different immune responses generated by innate and adaptive immune components during acute and chronic T. muris infections. Furthermore, we discussed the importance of studying T. muris model in understanding host–parasite interaction in the context of alteration in the host’s microbiota, intestinal barrier, inflammation, and host defense, and in parasite infection-mediated modulation of other immune and inflammatory diseases.
Collapse
Affiliation(s)
- Yeganeh Yousefi
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Sabah Haq
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Suhrid Banskota
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Yun Han Kwon
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Waliul I. Khan
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
- Correspondence: ; Tel.: +1-905-521-2100 (ext. 22846)
| |
Collapse
|
32
|
Jin H, Miao H, Nie YW, Lin YY. Investigating resistin like beta (RETNLB) as a tumor promoter for oral squamous cell carcinoma. Head Face Med 2021; 17:20. [PMID: 34158059 PMCID: PMC8218422 DOI: 10.1186/s13005-021-00272-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Oral cavity cancer ranks the sixth most common malignancy worldwide, of which oral squamous cell carcinoma is the predominant type. This study aimed to investigate the function and the underlying mechanism of resistin like beta (RETNLB) in oral squamous cell carcinoma. METHODS The data of oral squamous cell carcinoma samples from The Cancer Genome Atlas database was used to examine RETNLB expression and assess its correlation with the clinical outcomes. Biological functions of RETNLB on the growth, invasion and migration of cells were determined by cell counting kit 8, clonogenic growth, and Transwell assays. Gene set enrichment analysis was utilized to identify the important gene sets associated with RETNLB expression, which was further confirmed by western blot. RESULTS We found that RETNLB was upregulated in oral squamous cell carcinoma tissues and cells. High expression of RETNLB was closely linked to age and pathological tumor, and significantly related to poor survival of oral squamous cell carcinoma patients. Further functional experiments showed that knockdown of RETNLB significantly reduced the viability, mobility and invasiveness of cells. Moreover, gene set enrichment analysis suggested that Toll-like receptor signaling pathway was significantly correlated with high RETNLB expression. Further western blot analysis verified that silencing RETNLB could notably suppress the protein levels of Toll-like receptor 2, Toll-like receptor 4 and phosphor- extracellular signal-regulated kinase. CONCLUSIONS These results suggested that downregulation of RETNLB may restrain the progression of oral squamous cell carcinoma by inactivating TLR/2/4/ERK pathway.
Collapse
Affiliation(s)
- Hong Jin
- College of Stomatology, Mudanjiang Medical University, Mudanjiang, 157000, Heilongjiang, China
| | - Hui Miao
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, 157000, Heilongjiang, China
| | - Yuan-Wen Nie
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, 157000, Heilongjiang, China
| | - Yang-Yang Lin
- Department of Stomatology, Hongqi Hospital Affiliated to Mudanjiang Medical University, No.708 of Guanghua Street, Mudanjiang, 157000, Heilongjiang, China.
| |
Collapse
|
33
|
The role of mucosal barriers in human gut health. Arch Pharm Res 2021; 44:325-341. [PMID: 33890250 DOI: 10.1007/s12272-021-01327-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022]
Abstract
The intestinal mucosa is continuously exposed to a large number of commensal or pathogenic microbiota and foreign food antigens. The intestinal epithelium forms a dynamic physicochemical barrier to maintain immune homeostasis. To efficiently absorb nutrients from food, the epithelium in the small intestine has thin, permeable layers spread over a vast surface area. Epithelial cells are renewed from the crypt toward the villi, accompanying epithelial cell death and shedding, to control bacterial colonization. Tight junction and adherens junction proteins provide epithelial cell-cell integrity. Microbial signals are recognized by epithelial cells via toll-like receptors. Environmental signals from short-chain fatty acids derived from commensal microbiota metabolites, aryl hydrocarbon receptors, and hypoxia-induced factors fortify gut barrier function. Here we summarize recent findings regarding various environmental factors for gut barrier function. Further, we discuss the role of gut barriers in the pathogenesis of human intestinal disease and the challenges of therapeutic strategies targeting gut barrier restoration.
Collapse
|
34
|
Initiation and Pathogenesis of Severe Asthma with Fungal Sensitization. Cells 2021; 10:cells10040913. [PMID: 33921169 PMCID: PMC8071493 DOI: 10.3390/cells10040913] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 12/26/2022] Open
Abstract
Fungi represent one of the most diverse and abundant eukaryotes on earth, and their ubiquity and small proteolytically active products make them pervasive allergens that affect humans and other mammals. The immunologic parameters surrounding fungal allergies are still not fully elucidated despite their importance given that a large proportion of severe asthmatics are sensitized to fungal allergens. Herein, we explore fungal allergic asthma with emphasis on mouse models that recapitulate the characteristics of human disease, and the main leukocyte players in the pathogenesis of fungal allergies. The endogenous mycobiome may also contribute to fungal asthma, a phenomenon that we discuss only superficially, as much remains to be discovered.
Collapse
|
35
|
Lee CB, Chae SU, Jo SJ, Jerng UM, Bae SK. The Relationship between the Gut Microbiome and Metformin as a Key for Treating Type 2 Diabetes Mellitus. Int J Mol Sci 2021; 22:ijms22073566. [PMID: 33808194 PMCID: PMC8037857 DOI: 10.3390/ijms22073566] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 02/08/2023] Open
Abstract
Metformin is the first-line pharmacotherapy for treating type 2 diabetes mellitus (T2DM); however, its mechanism of modulating glucose metabolism is elusive. Recent advances have identified the gut as a potential target of metformin. As patients with metabolic disorders exhibit dysbiosis, the gut microbiome has garnered interest as a potential target for metabolic disease. Henceforth, studies have focused on unraveling the relationship of metabolic disorders with the human gut microbiome. According to various metagenome studies, gut dysbiosis is evident in T2DM patients. Besides this, alterations in the gut microbiome were also observed in the metformin-treated T2DM patients compared to the non-treated T2DM patients. Thus, several studies on rodents have suggested potential mechanisms interacting with the gut microbiome, including regulation of glucose metabolism, an increase in short-chain fatty acids, strengthening intestinal permeability against lipopolysaccharides, modulating the immune response, and interaction with bile acids. Furthermore, human studies have demonstrated evidence substantiating the hypotheses based on rodent studies. This review discusses the current knowledge of how metformin modulates T2DM with respect to the gut microbiome and discusses the prospect of harnessing this mechanism in treating T2DM.
Collapse
Affiliation(s)
- Chae Bin Lee
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
| | - Soon Uk Chae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
| | - Seong Jun Jo
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
| | - Ui Min Jerng
- Department of Internal Medicine, College of Korean Medicine, Sangji University, Wonju 26339, Korea;
| | - Soo Kyung Bae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
- Correspondence: ; Tel.: +82-2-2164-4054
| |
Collapse
|
36
|
Iftekhar A, Sigal M. Defence and adaptation mechanisms of the intestinal epithelium upon infection. Int J Med Microbiol 2021; 311:151486. [PMID: 33684844 DOI: 10.1016/j.ijmm.2021.151486] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/15/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium is a monolayer of polarized columnar cells that act as a border between the host and its environment and are the first line of defence against the luminal microbes. In addition to providing a physical barrier, the epithelium possesses a multitude of active mechanisms to fight invading pathogens and regulate the composition and spatial distribution of commensals. The different epithelial cell types have unique functions in this context, and crosstalk with the immune system further modulates their intricate antimicrobial responses. The epithelium is organized into clonal crypt units with a high cellular turnover that is driven by stem cells located at the base. There is increasing evidence that this anatomical organization, the stem cell turnover, and the lineage determination processes are essential for barrier maintenance. These processes can be modulated by microbes directly or by the immune responses to enteric pathogens, resulting in a rapid and efficient adaptation of the epithelium to environmental perturbations, injuries, and infections. Here we discuss the complex host-microbial interactions that shape the mucosa and how the epithelium maintains and re-establishes homeostasis after infection.
Collapse
Affiliation(s)
- Amina Iftekhar
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Michael Sigal
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Department of Internal Medicine, Gastroenterology and Hepatology, Charité University Medicine, Berlin, Germany; Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
37
|
Li J, Zhang L, Wu T, Li Y, Zhou X, Ruan Z. Indole-3-propionic Acid Improved the Intestinal Barrier by Enhancing Epithelial Barrier and Mucus Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:1487-1495. [PMID: 33356219 DOI: 10.1021/acs.jafc.0c05205] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Destruction in intestinal barrier is concomitant with the intestinal diseases. There is growing evidence that tryptophan-derived intestinal bacterial metabolites play a critical role in maintaining the balance of intestinal mucosa. In this study, the Caco-2/HT29 coculture model was used to evaluate the effect of indole-3-propionic acid (IPA) on the intestinal barrier and explore its underlying mechanism. We found that IPA increased transepithelial electrical resistance and decreased paracellular permeability which was consistent with the increase in tight junction proteins (claudin-1, occludin, and ZO-1). Furthermore, IPA strengthened the mucus barrier by increasing mucins (MUC2 and MUC4) and goblet cell secretion products (TFF3 and RELMβ). Additionally, IPA weakened the expression of LPS-induced inflammatory factors. These discoveries provide new views for understanding the improvement of intestinal barrier by gut microbial metabolites of aromatic amino acids.
Collapse
Affiliation(s)
- Jiaojiao Li
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang 330047, China
| | - Li Zhang
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang 330047, China
| | - Tao Wu
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang 330047, China
| | - Yafei Li
- University Campus Hospital and Jiangxi Academy of Medical Science, Nanchang University, Nanchang 330006, China
| | - Xiaojun Zhou
- School of Public Health, Jiangxi Province Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, China
| | - Zheng Ruan
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang 330047, China
| |
Collapse
|
38
|
Sprouty2 limits intestinal tuft and goblet cell numbers through GSK3β-mediated restriction of epithelial IL-33. Nat Commun 2021; 12:836. [PMID: 33547321 PMCID: PMC7864916 DOI: 10.1038/s41467-021-21113-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Dynamic regulation of intestinal cell differentiation is crucial for both homeostasis and the response to injury or inflammation. Sprouty2, an intracellular signaling regulator, controls pathways including PI3K and MAPKs that are implicated in differentiation and are dysregulated in inflammatory bowel disease. Here, we ask whether Sprouty2 controls secretory cell differentiation and the response to colitis. We report that colonic epithelial Sprouty2 deletion leads to expanded tuft and goblet cell populations. Sprouty2 loss induces PI3K/Akt signaling, leading to GSK3β inhibition and epithelial interleukin (IL)-33 expression. In vivo, this results in increased stromal IL-13+ cells. IL-13 in turn induces tuft and goblet cell expansion in vitro and in vivo. Sprouty2 is downregulated by acute inflammation; this appears to be a protective response, as VillinCre;Sprouty2F/F mice are resistant to DSS colitis. In contrast, Sprouty2 is elevated in chronic colitis and in colons of inflammatory bowel disease patients, suggesting that this protective epithelial-stromal signaling mechanism is lost in disease. Dynamic regulation of colonic secretory cell numbers is a critical component of the response to intestinal injury and inflammation. Here, the authors show that loss of the intracellular signalling regulator Sprouty2 in the intestinal epithelial cells is a protective response to injury that leads to increased secretory cell numbers, thus limiting colitis severity.
Collapse
|
39
|
Zawawi A, Else KJ. Soil-Transmitted Helminth Vaccines: Are We Getting Closer? Front Immunol 2020; 11:576748. [PMID: 33133094 PMCID: PMC7565266 DOI: 10.3389/fimmu.2020.576748] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/02/2020] [Indexed: 01/07/2023] Open
Abstract
Parasitic helminths infect over one-fourth of the human population resulting in significant morbidity, and in some cases, death in endemic countries. Despite mass drug administration (MDA) to school-aged children and other control measures, helminth infections are spreading into new areas. Thus, there is a strong rationale for developing anthelminthic vaccines as cost-effective, long-term immunological control strategies, which, unlike MDA, are not haunted by the threat of emerging drug-resistant helminths nor limited by reinfection risk. Advances in vaccinology, immunology, and immunomics include the development of new tools that improve the safety, immunogenicity, and efficacy of vaccines; and some of these tools have been used in the development of helminth vaccines. The development of anthelminthic vaccines is fraught with difficulty. Multiple lifecycle stages exist each presenting stage-specific antigens. Further, helminth parasites are notorious for their ability to dampen down and regulate host immunity. One of the first significant challenges in developing any vaccine is identifying suitable candidate protective antigens. This review explores our current knowledge in lead antigen identification and reports on recent pre-clinical and clinical trials in the context of the soil-transmitted helminths Trichuris, the hookworms and Ascaris. Ultimately, a multivalent anthelminthic vaccine could become an essential tool for achieving the medium-to long-term goal of controlling, or even eliminating helminth infections.
Collapse
Affiliation(s)
- Ayat Zawawi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Ayat Zawawi
| | - Kathryn J. Else
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine, and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom,Kathryn J. Else
| |
Collapse
|
40
|
Koepsell H. Glucose transporters in the small intestine in health and disease. Pflugers Arch 2020; 472:1207-1248. [PMID: 32829466 PMCID: PMC7462918 DOI: 10.1007/s00424-020-02439-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/11/2020] [Accepted: 07/17/2020] [Indexed: 12/23/2022]
Abstract
Absorption of monosaccharides is mainly mediated by Na+-D-glucose cotransporter SGLT1 and the facititative transporters GLUT2 and GLUT5. SGLT1 and GLUT2 are relevant for absorption of D-glucose and D-galactose while GLUT5 is relevant for D-fructose absorption. SGLT1 and GLUT5 are constantly localized in the brush border membrane (BBM) of enterocytes, whereas GLUT2 is localized in the basolateral membrane (BLM) or the BBM plus BLM at low and high luminal D-glucose concentrations, respectively. At high luminal D-glucose, the abundance SGLT1 in the BBM is increased. Hence, D-glucose absorption at low luminal glucose is mediated via SGLT1 in the BBM and GLUT2 in the BLM whereas high-capacity D-glucose absorption at high luminal glucose is mediated by SGLT1 plus GLUT2 in the BBM and GLUT2 in the BLM. The review describes functions and regulations of SGLT1, GLUT2, and GLUT5 in the small intestine including diurnal variations and carbohydrate-dependent regulations. Also, the roles of SGLT1 and GLUT2 for secretion of enterohormones are discussed. Furthermore, diseases are described that are caused by malfunctions of small intestinal monosaccharide transporters, such as glucose-galactose malabsorption, Fanconi syndrome, and fructose intolerance. Moreover, it is reported how diabetes, small intestinal inflammation, parental nutrition, bariatric surgery, and metformin treatment affect expression of monosaccharide transporters in the small intestine. Finally, food components that decrease D-glucose absorption and drugs in development that inhibit or downregulate SGLT1 in the small intestine are compiled. Models for regulations and combined functions of glucose transporters, and for interplay between D-fructose transport and metabolism, are discussed.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstr 6, 97070, Würzburg, Germany.
| |
Collapse
|
41
|
Miller MM, Reinhardt RL. The Heterogeneity, Origins, and Impact of Migratory iILC2 Cells in Anti-helminth Immunity. Front Immunol 2020; 11:1594. [PMID: 32793230 PMCID: PMC7390839 DOI: 10.3389/fimmu.2020.01594] [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] [Received: 04/22/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Soil-transmitted helminths represent a major global health burden with infections and infection-related comorbidities causing significant reductions in the quality of life for individuals living in endemic areas. Repeated infections and chronic colonization by these large extracellular worms in mammals led to the evolution of type-2 immunity characterized by the production of the type-2 cytokines interleukin (IL)-4, IL-5, and IL-13. Although a number of adaptive and innate immune cells produce type-2 cytokines, a key cellular source in the context of helminth infection is group 2 innate lymphoid cells (ILC2s). ILC2s promote mucosal barrier homeostasis, integrity, and repair by rapidly responding to epithelial cues in mucosal tissues. Though tissue-resident ILC2s (nILC2s) have been studied in detail over the last decade, considerably less is known with regard to a subset of inflammatory ILC2s (iILC2s) that migrate to the lungs of mice early after Nippostrongylus brasiliensis infection and are potent early producers of type-2 cytokines. This review will discuss the relationship and differences between nILC2s and iILC2s that establish their unique roles in anti-helminth immunity. We have placed particular emphasis on studies investigating iILC2 origin, function, and their potential long-term contribution to tissue-resident ILC2 reservoirs in settings of helminth infection.
Collapse
Affiliation(s)
- Mindy M Miller
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| | - R Lee Reinhardt
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States.,Department of Immunology and Microbiology, University of Colorado-Anschutz Medical, Aurora, CO, United States
| |
Collapse
|
42
|
Puértolas-Balint F, Schroeder BO. Does an Apple a Day Also Keep the Microbes Away? The Interplay Between Diet, Microbiota, and Host Defense Peptides at the Intestinal Mucosal Barrier. Front Immunol 2020; 11:1164. [PMID: 32655555 PMCID: PMC7325984 DOI: 10.3389/fimmu.2020.01164] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
A crucial mechanism of intestinal defense includes the production and secretion of host defense peptides (HDPs). HDPs control pathogens and commensals at the intestinal interface by direct killing, by sequestering vital ions, or by causing bacterial cells to aggregate in the mucus layer. Accordingly, the combined activity of various HDPs neutralizes gut bacteria before reaching the mucosa and thus helps to maintain the homeostatic balance between the host and its microbes at the mucosal barrier. Defects in the mucosal barrier have been associated with various diseases that are on the rise in the Western world. These include metabolic diseases, such as obesity and type 2 diabetes, and inflammatory intestinal disorders, including ulcerative colitis and Crohn's disease, the two major entities of inflammatory bowel disease. While the etiology of these diseases is multifactorial, highly processed Western-style diet (WSD) that is rich in carbohydrates and fat and low in dietary fiber content, is considered to be a contributing lifestyle factor. As such, WSD does not only profoundly affect the resident microbes in the intestine, but can also directly alter HDP function, thereby potentially contributing to intestinal mucosal barrier dysfunction. In this review we aim to decipher the complex interaction between diet, microbiota, and HDPs. We discuss how HDP expression can be modulated by specific microbes and their metabolites as well as by dietary factors, including fibers, lipids, polyphenols and vitamins. We identify several dietary compounds that lead to reduced HDP function, but also factors that stimulate HDP production in the intestine. Furthermore, we argue that the effect of HDPs against commensal bacteria has been understudied when compared to pathogens, and that local environmental conditions also need to be considered. In addition, we discuss the known molecular mechanisms behind HDP modulation. We believe that a better understanding of the diet-microbiota-HDP interdependence will provide insights into factors underlying modern diseases and will help to identify potential dietary interventions or probiotic supplementation that can promote HDP-mediated intestinal barrier function in the Western gut.
Collapse
Affiliation(s)
- Fabiola Puértolas-Balint
- Laboratory for Molecular Infection Medicine Sweden (MIMS) -The Nordic EMBL Partnership for Molecular Medicine, Umeå University, Umeå, Sweden.,Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Bjoern O Schroeder
- Laboratory for Molecular Infection Medicine Sweden (MIMS) -The Nordic EMBL Partnership for Molecular Medicine, Umeå University, Umeå, Sweden.,Department of Molecular Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
43
|
Colombo SAP, Grencis RK. Immunity to Soil-Transmitted Helminths: Evidence From the Field and Laboratory Models. Front Immunol 2020; 11:1286. [PMID: 32655568 PMCID: PMC7324686 DOI: 10.3389/fimmu.2020.01286] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
Infection with soil-transmitted helminths (STH) remains a major burden on global health and agriculture. Our understanding of the immunological mechanisms that govern whether an individual is resistant or susceptible to infection is derived primarily from model infections in rodents. Typically, experimental infections employ an artificially high, single bolus of parasites that leads to rapid expulsion of the primary infection and robust immunity to subsequent challenges. However, immunity in natura is generated slowly, and is only partially effective, with individuals in endemic areas retaining low-level infections throughout their lives. Therefore, there is a gap between traditional model STH systems and observations in the field. Here, we review the immune response to traditional model STH infections in the laboratory. We compare these data to studies of natural infection in humans and rodents in endemic areas, highlighting crucial differences between experimental and natural infection. We then detail the literature to date on the use of "trickle" infections to experimentally model the kinetics of natural infection.
Collapse
Affiliation(s)
- Stefano A. P. Colombo
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Richard K. Grencis
- Division of Infection, Immunity and Respiratory Medicine, Wellcome Trust Centre for Cell Matrix Research, Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
44
|
Lin Q, Johns RA. Resistin family proteins in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2020; 319:L422-L434. [PMID: 32692581 DOI: 10.1152/ajplung.00040.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The family of resistin-like molecules (RELMs) consists of four members in rodents (RELMα/FIZZ1/HIMF, RELMβ/FIZZ2, Resistin/FIZZ3, and RELMγ/FIZZ4) and two members in humans (Resistin and RELMβ), all of which exhibit inflammation-regulating, chemokine, and growth factor properties. The importance of these cytokines in many aspects of physiology and pathophysiology, especially in cardiothoracic diseases, is rapidly evolving in the literature. In this review article, we attempt to summarize the contribution of RELM signaling to the initiation and progression of lung diseases, such as pulmonary hypertension, asthma/allergic airway inflammation, chronic obstructive pulmonary disease, fibrosis, cancers, infection, and other acute lung injuries. The potential of RELMs to be used as biomarkers or risk predictors of these diseases also will be discussed. Better understanding of RELM signaling in the pathogenesis of pulmonary diseases may offer novel targets or approaches for the development of therapeutics to treat or prevent a variety of inflammation, tissue remodeling, and fibrosis-related disorders in respiratory, cardiovascular, and other systems.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
45
|
Blyth GAD, Connors L, Fodor C, Cobo ER. The Network of Colonic Host Defense Peptides as an Innate Immune Defense Against Enteropathogenic Bacteria. Front Immunol 2020; 11:965. [PMID: 32508838 PMCID: PMC7251035 DOI: 10.3389/fimmu.2020.00965] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Host defense peptides, abundantly secreted by colonic epithelial cells and leukocytes, are proposed to be critical components of an innate immune response in the colon against enteropathogenic bacteria, including Shigella spp., Salmonella spp., Clostridium difficile, and attaching and effacing Escherichia coli and Citrobacter rodentium. These short cationic peptides are bactericidal against both Gram-positive and -negative enteric pathogens, but may also exert killing effects on intestinal luminal microbiota. Simultaneously, these peptides modulate numerous cellular responses crucial for gut defenses, including leukocyte chemotaxis and migration, wound healing, cytokine production, cell proliferation, and pathogen sensing. This review discusses recent advances in our understanding of expression, mechanisms of action and microbicidal and immunomodulatory functions of major colonic host defense peptides, namely cathelicidins, β-defensins, and members of the Regenerating islet-derived protein III (RegIII) and Resistin-like molecule (RELM) families. In a theoretical framework where these peptides work synergistically, aspects of pathogenesis of infectious colitis reviewed herein uncover roles of host defense peptides aimed to promote epithelial defenses and prevent pathogen colonization, mediated through a combination of direct antimicrobial function and fine-tuning of host immune response and inflammation. This interactive host defense peptide network may decode how the intestinal immune system functions to quickly clear infections, restore homeostasis and avoid damaging inflammation associated with pathogen persistence during infectious colitis. This information is of interest in development of host defense peptides (either alone or in combination with reduced doses of antibiotics) as antimicrobial and immunomodulatory therapeutics for controlling infectious colitis.
Collapse
Affiliation(s)
- Graham A D Blyth
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Liam Connors
- Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Cristina Fodor
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Eduardo R Cobo
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
46
|
Helicobacter pylori Infection Facilitates the Expression of Resistin-like Molecule Beta in Gastric Carcinoma and Precursor Lesions. Curr Med Sci 2020; 40:95-103. [PMID: 32166670 DOI: 10.1007/s11596-020-2151-y] [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: 10/24/2018] [Revised: 04/08/2019] [Indexed: 10/24/2022]
Abstract
Helicobacter pylori (H. pylori) was reported to be associated with gastric carcinogenesis. Resistin-like molecule beta (RELMβ), a recently described goblet cell-specific protein, was demonstrated to aberrantly express in gastric cancer and correlated with its clinicopathological features. This study aimed to examine the association between H. pylori and RELMβ expression in gastric carcinoma and precursor lesions. H. pylori infection and RELMβ expression were immunohistochemically evaluated in gastric biopsies from 230 patients. The biopsies consisted of normal gastric mucosa (n=20), mucosa with chronic gastritis (n=41), intestinal metaplasia (n=42), dysplasia (n=31), intestinal-type adenocarcinoma (n=56), and diffuse-type adenocarcinoma (n=40). RELMβ expression was measured in gastric biopsies after H. pylori eradication therapy in a subgroup of 32 patients. Cultured gastric cancer cell line SGC-7901 was infected with H. pylori strains, and RELMβ expression was detected by reverse transcription PCR, real-time PCR and Western blotting. Higher RELMβ immunoreactivity was observed in H. pylori-positive intestinal metaplasia (P=0.003), dysplasia (P=0.032), intestinal-type (P=0.037) and diffuse-type adenocarcinomas (P=0.001) than in H. pylori-negative specimens. Expression rates of RELMβ in dysplasia (P=0.005), intestinal-type adenocarcinoma (P<0.001), and diffuse-type adenocarcinoma (P=0.001) were significantly correlated with the grade of H. pylori density. In addition, H. pylori eradication reduced the RELMβ intensity in intestinal metaplasia (P=0.001). Infection of gastric cancer SGC-7901 cells with cag pathogenicity island (PAI)-positive H. pylori TN2, but not with its PAI totally deleted mutant (TN2-ΔPAI) for 4-8 h, resulted in enhanced protein and transcript levels of RELMβ (P<0.05). In summary, our study suggested that H. pylori infection facilitated the expression of RELMβ in gastric garcinoma and precursor lesions.
Collapse
|
47
|
Watanabe K, Itoh K, Park SH, Kaku M, Ishii K, Sasano H, Naitoh T, Unno M, Fukushima K. Resistin-like molecule beta, a colonic epithelial protein, exhibits antimicrobial activity against Staphylococcus aureus including methicillin-resistant strains. Surg Today 2020; 50:920-930. [PMID: 32062787 DOI: 10.1007/s00595-020-01974-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/10/2020] [Indexed: 01/20/2023]
Abstract
PURPOSE Resistin-like molecule beta (RELMβ) is a small cysteine-rich protein secreted by colonic epithelial cells. RELMβ mRNA and protein expressions are dramatically induced by bacterial exposure in germ-free mice. We hypothesized that RELMβ has antimicrobial activity. METHODS The antimicrobial activity of RELMβ was screened by an agar spot test and confirmed by a liquid broth test. The amount of RELMβ in human stools was semi-quantified by Western blot analysis. The induction of RELMβ mRNA and protein expression by bacteria was measured by quantitative RT-PCR using LS174T cells. Electron microscopic immunohistochemistry was performed using polyclonal anti-RELMβ antibody. RESULTS RELMβ showed antimicrobial activity against S. aureus and all MRSAs examined in a dose- and pH-dependent fashion. Western blot study showed that the amount of RELMβ in healthy human stools was comparable to that exhibiting antimicrobial activity in vitro. Both RELMβ mRNA and protein expression were induced by heat-inactivated S. aureus, but not by E. coli in LS174T cells. Electron microscopic immunohistochemistry showed that RELMβ bound to the cell surface of S. aureus, followed by destruction of the bacterial cytoplasm. CONCLUSIONS RELMβ is a colonic antimicrobial protein and its antibacterial activity is species selective. Because RELMβ is abundant in healthy human stool, RELMβ may modulate gut flora.
Collapse
Affiliation(s)
- Kazuhiro Watanabe
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan.
| | - Kikuji Itoh
- Laboratory of Veterinary Public Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Sang-Hee Park
- Laboratory of Veterinary Public Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.,Clinical Research Center, Masan National Tuberculosis Hospital, Changwon, South Korea
| | - Mitsuo Kaku
- Department of Infection Control and Laboratory Diagnostics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keiko Ishii
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Hospital, Sendai, Japan
| | - Takeshi Naitoh
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan
| | - Kouhei Fukushima
- Laboratory of GI Tract Reconstruction, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan.,Department of Molecular and Surgical Pathophysiology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
48
|
Norkin M, Capdevila C, Calderon RI, Su T, Trifas M, Ordóñez-Morán P, Yan KS. Single-Cell Studies of Intestinal Stem Cell Heterogeneity During Homeostasis and Regeneration. Methods Mol Biol 2020; 2171:155-167. [PMID: 32705640 DOI: 10.1007/978-1-0716-0747-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Single-cell RNA-sequencing (scRNA-seq) provides a unique opportunity to study heterogeneous cell populations within tissues, including the intestinal epithelium, to gain detailed molecular insights into their biology. Many new putative markers of intestinal stem cells and their progeny have been described using single-cell transcriptomics, which has contributed to the identification of novel subpopulations of mature cell types and insight into their developmental trajectories. This approach has revealed tremendous cellular heterogeneity within the intestinal epithelium that is concordant with its diverse and multifaceted functions. We discuss the function of these subpopulations during tissue homeostasis, as well as putative subpopulations with inducible regenerative potential following tissue injury.
Collapse
Affiliation(s)
- Maxim Norkin
- Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Claudia Capdevila
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia Stem Cell Initiative, New York, NY, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Ruben I Calderon
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia Stem Cell Initiative, New York, NY, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Tianhong Su
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia Stem Cell Initiative, New York, NY, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Maria Trifas
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia Stem Cell Initiative, New York, NY, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Paloma Ordóñez-Morán
- Department of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| | - Kelley S Yan
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia Stem Cell Initiative, New York, NY, USA. .,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
49
|
Yasuda K, Kuroda E. Role of eosinophils in protective immunity against secondary nematode infections. Immunol Med 2019; 42:148-155. [DOI: 10.1080/25785826.2019.1697135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Koubun Yasuda
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Etsushi Kuroda
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
50
|
Soderholm AT, Pedicord VA. Intestinal epithelial cells: at the interface of the microbiota and mucosal immunity. Immunology 2019; 158:267-280. [PMID: 31509239 PMCID: PMC6856932 DOI: 10.1111/imm.13117] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/27/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
The intestinal epithelium forms a barrier between the microbiota and the rest of the body. In addition, beyond acting as a physical barrier, the function of intestinal epithelial cells (IECs) in sensing and responding to microbial signals is increasingly appreciated and likely has numerous implications for the vast network of immune cells within and below the intestinal epithelium. IECs also respond to factors produced by immune cells, and these can regulate IEC barrier function, proliferation and differentiation, as well as influence the composition of the microbiota. The mechanisms involved in IEC-microbe-immune interactions, however, are not fully characterized. In this review, we explore the ability of IECs to direct intestinal homeostasis by orchestrating communication between intestinal microbes and mucosal innate and adaptive immune cells during physiological and inflammatory conditions. We focus primarily on the most recent findings and call attention to the numerous remaining unknowns regarding the complex crosstalk between IECs, the microbiota and intestinal immune cells.
Collapse
Affiliation(s)
- Amelia T. Soderholm
- Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Virginia A. Pedicord
- Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| |
Collapse
|