1
|
Sharan K, Brandt C, Yusuf MA, Singh P, Halder N, Edwards ME, Mangu SVVSR, Das A, Mishra A, Kumar SS, Sharma A, Gupta A, Liu XS, Guo EX, Monani UR, Ponnalagu D, Ivanov II, Lal G, Clare S, Dougan G, Yadav VK. Rapid and relaying deleterious effects of a gastrointestinal pathogen, Citrobacter rodentium, on bone, an extra-intestinal organ. iScience 2025; 28:111802. [PMID: 39967874 PMCID: PMC11834125 DOI: 10.1016/j.isci.2025.111802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/04/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Enteropathogenic infections cause pathophysiological changes in the host but their effects beyond the gastrointestinal tract are undefined. Here, using Citrobacter rodentium infection in mouse, which mimics human diarrheal enteropathogenic Escherichia coli, we show that gastrointestinal infection negatively affects bone remodeling, leading to compromised bone architecture. Transmission of infection through fecal-oral route from Citrobacter rodentium-infected to non-infected mice caused bone loss in non-infected cage mates. Mice with B cell deficiency (Igh6-/- mice) failed to clear C. rodentium infection and exhibited more severe and long-term bone loss compared to WT mice. Unbiased cytokine profiling showed an increase in circulating tumor necrosis factor α (TNFα) levels following Citrobacter rodentium infection, and immunoneutralization of TNFα prevented infection-induced bone loss completely in WT and immunocompromised mice. These findings reveal rapid, relaying, and modifiable effects of enteropathogenic infections on an extraintestinal organ-bone, and provide insights into the mechanism(s) through which these infections affect extraintestinal organ homeostasis.
Collapse
Affiliation(s)
- Kunal Sharan
- Mouse Genetics Project, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
- Department of Molecular Nutrition, CSIR-CFTRI, Mysore, Karnataka, India
| | - Cordelia Brandt
- Host-Pathogen Interaction Group, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
| | - Mohd Aslam Yusuf
- Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, India
| | - Parminder Singh
- National Institute of Immunology, New Delhi, New Delhi, India
| | - Namrita Halder
- National Centre for Cell Science, Pune, Maharastra, India
| | - Madeline E. Edwards
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - SVVS Ravi Mangu
- Department of Molecular Nutrition, CSIR-CFTRI, Mysore, Karnataka, India
| | - Abhilipsa Das
- Department of Molecular Nutrition, CSIR-CFTRI, Mysore, Karnataka, India
| | - Amrita Mishra
- National Centre for Cell Science, Pune, Maharastra, India
| | - Shashi S. Kumar
- Center for Motor Neuron Biology & Disease, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Amita Sharma
- Pediatric Kidney Foundation, New Delhi, New Delhi, India
| | - Alka Gupta
- Reproductive Biology Laboratory, National Institute of Immunology, New Delhi, New Delhi, India
| | - Xiaowei S. Liu
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward X. Guo
- Bone Biomechanics Laboratory, Columbia University, New York, NY, USA
| | - Umrao R. Monani
- Center for Motor Neuron Biology & Disease, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | | | - Ivaylo I. Ivanov
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Girdhari Lal
- National Centre for Cell Science, Pune, Maharastra, India
| | - Simon Clare
- Host-Pathogen Interaction Group, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
| | - Gordon Dougan
- Host-Pathogen Interaction Group, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
- Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Centre for Translational Stem Cell Biology, Hong Kong, China
| | - Vijay K. Yadav
- Mouse Genetics Project, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
- National Institute of Immunology, New Delhi, New Delhi, India
- Department of Genetics and Development, Columbia University, New York, NY, USA
- Healthy Longevity Program, Department of Pathology, Immunology and Laboratory Medicine, Rutgers University, Newark, NJ, USA
- Center for Cell Signaling, Rutgers University, Newark, NJ, USA
- Center for Immunity and Inflammation, Rutgers University, Newark, NJ, USA
| |
Collapse
|
2
|
Cremin M, Ramirez VT, Sanchez K, Tay E, Murray K, Brust-Mascher I, Reardon C. Substance P receptor signaling contributes to host maladaptive responses during enteric bacterial infection. Proc Natl Acad Sci U S A 2025; 122:e2415287122. [PMID: 39937862 PMCID: PMC11848390 DOI: 10.1073/pnas.2415287122] [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: 07/29/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025] Open
Abstract
Immune responses in the intestine are intricately balanced to prevent pathogen entry without inducing immunopathology. The nervous system is well established to interface with the immune system to fine-tune immunity in various organ systems including the gastrointestinal tract. Specialized sensory neurons can detect bacteria, bacterial products, and the resulting inflammation, to coordinate the immune response in the gastrointestinal tract. These sensory neurons release peptide neurotransmitters such as Substance P (SP), to induce both neuronal signaling and localized responses in nonneuronal cells. With this in mind, we assessed the immunoregulatory roles of SP receptor signaling during enteric bacterial infection with the noninvasive pathogen Citrobacter rodentium. Pharmacological antagonism of the SP receptor significantly reduced bacterial burden and prevented colonic crypt hyperplasia. Mice with SP receptor signaling blockade had significantly reduced inflammation and recruitment of T cells in the colon. Reduced colonic T cell recruitment is due to reduced expression of adhesion molecules on colonic endothelial cells in SP receptor antagonist-treated mice. Using SP receptor T cell conditional knockout mice, we further confirmed SP receptor signaling enhanced select aspects of T cell responses. Our data demonstrate that SP receptor signaling can significantly reduce inflammation and prevent host-maladaptive responses without impinging upon host protection.
Collapse
Affiliation(s)
- Michael Cremin
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA95616
| | - Valerie T. Ramirez
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA95616
| | - Kristina Sanchez
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA95616
| | - Emmy Tay
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA95616
| | - Kaitlin Murray
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA95616
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA95616
| | - Colin Reardon
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA95616
| |
Collapse
|
3
|
Liu Y, Xu D, Guo S, Wang S, Ding H, Siu C, Wan F. The gut microbiota-independent virulence of noninvasive bacterial pathogen Citrobacter rodentium. PLoS Pathog 2024; 20:e1012758. [PMID: 39630719 DOI: 10.1371/journal.ppat.1012758] [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: 07/08/2024] [Revised: 12/16/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
Attaching and effacing (A/E) bacterial pathogens consist of human pathogens enteropathogenic Escherichia coli, enterohemorrhagic E. coli and their murine equivalent Citrobacter rodentium (CR). Emerging evidence suggests that the complex pathogen-microbiota-host interactions are critical in conferring A/E pathogen infection-induced severe symptoms and lethality in immunocompromised hosts; however, the precise underlying mechanisms remain enigmatic. Here we report that CR infection causes severe colitis and mortality in interleukin 22 knockout (Il22-/-) and Rag1 knockout (Rag1-/-) mice under germ-free (GF) conditions. In a gut microbiota-independent manner, CR colonizes in GF Il22-/- and Rag1-/- animals, triggers colonic epithelial tissue damage and systemic dissemination of CR, and results in lethal infections. Pretreatment with cefoxitin, a broad-spectrum antibiotic, exacerbates CR-induced colitis and lethality in specific-pathogen-free (SPF) Il22-/- and Rag1-/- mice. Together our results reveal that CR possesses a gut microbiota-independent virulence, which is better illustrated during infections in immunocompromised hosts associated with severe outcomes.
Collapse
Affiliation(s)
- Yue Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Dongqing Xu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Songwei Guo
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shuyu Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Hua Ding
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Catherine Siu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
4
|
Solaymani-Mohammadi S. The IL-21/IL-21R signaling axis regulates CD4+ T-cell responsiveness to IL-12 to promote bacterial-induced colitis. J Leukoc Biol 2024; 116:726-737. [PMID: 38498592 PMCID: PMC11408709 DOI: 10.1093/jleuko/qiae069] [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: 11/21/2023] [Revised: 02/21/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
IL-21/IL-21R signaling dysregulation is linked to multiple chronic intestinal inflammatory disorders in humans and animal models of human diseases. In addition to its critical requirement for the generation and development of germinal center B cells, IL-21/IL-21R signaling can also regulate the effector functions of a variety of T-cell subsets. The antibody-mediated abrogation of IL-21/IL-21R signaling led to the impaired expression of IFN-γ by mucosal CD4+ T cells from human subjects with colitis, suggesting an IL-21/IL-21R-triggered positive feedback loop of the TH1 immune response in the colon. Despite recent advances in our understanding of the mechanisms underpinning the regulation of proinflammatory immune responses by the IL-21/IL-21R signaling axis, it remains unclear how this pathway or its downstream molecules contribute to inflammation during bacterial-induced colitis. This study found that IL-21 enhances the surface expression of IL-12Rβ2, but not IL-12Rβ1, in CD4+ T cells, leading to TH1 differentiation and stability. Consistently, these findings also point to an indispensable role of the IL-12Rβ2 signaling axis in promoting proinflammatory immune responses during Citrobacter rodentium-induced colitis. Genetic deletion of the IL-12Rβ2 signaling pathway led to the attenuation of C. rodentium-induced colitis in vivo. The genetic deletion of the IL-12Rβ2 signaling pathway did not alter the host's ability to respond adequately to C. rodentium infection or the ability of Il12rb2-/- mice to express antigen-specific cytokines (IFN-γ, IL-17A). IL-21 is a pleiotropic cytokine exerting a wide range of immunomodulatory functions in multiple tissues, and its direct targeting may result in undesirable off-target consequences. These findings highlight the possibility for targeted manipulations of signaling cascades downstream of main regulators of proinflammatory responses to control invading pathogens while preserving the integrity of host immune responses. A better understanding of the novel mechanisms by which IL-21/IL-21R signaling regulates bacterial-induced colitis will provide insights into the development of new therapeutic and preventive strategies to harness IL-21/IL-21R signaling or its downstream molecules to treat infectious colitis.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, 1301 North Columbia Road, Suite W315, Stop 9037, Grand Forks, ND, United States
| |
Collapse
|
5
|
Bonetti L, Horkova V, Grusdat M, Longworth J, Guerra L, Kurniawan H, Franchina DG, Soriano-Baguet L, Binsfeld C, Verschueren C, Spath S, Ewen A, Koncina E, Gérardy JJ, Kobayashi T, Dostert C, Farinelle S, Härm J, Fan YT, Chen Y, Harris IS, Lang PA, Vasiliou V, Waisman A, Letellier E, Becher B, Mittelbronn M, Brenner D. A Th17 cell-intrinsic glutathione/mitochondrial-IL-22 axis protects against intestinal inflammation. Cell Metab 2024; 36:1726-1744.e10. [PMID: 38986617 DOI: 10.1016/j.cmet.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/06/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
The intestinal tract generates significant reactive oxygen species (ROS), but the role of T cell antioxidant mechanisms in maintaining intestinal homeostasis is poorly understood. We used T cell-specific ablation of the catalytic subunit of glutamate cysteine ligase (Gclc), which impaired glutathione (GSH) production, crucially reducing IL-22 production by Th17 cells in the lamina propria, which is critical for gut protection. Under steady-state conditions, Gclc deficiency did not alter cytokine secretion; however, C. rodentium infection induced increased ROS and disrupted mitochondrial function and TFAM-driven mitochondrial gene expression, resulting in decreased cellular ATP. These changes impaired the PI3K/AKT/mTOR pathway, reducing phosphorylation of 4E-BP1 and consequently limiting IL-22 translation. The resultant low IL-22 levels led to poor bacterial clearance, severe intestinal damage, and high mortality. Our findings highlight a previously unrecognized, essential role of Th17 cell-intrinsic GSH in promoting mitochondrial function and cellular signaling for IL-22 protein synthesis, which is critical for intestinal integrity and defense against gastrointestinal infections.
Collapse
Affiliation(s)
- Lynn Bonetti
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Veronika Horkova
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Melanie Grusdat
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Luana Guerra
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Henry Kurniawan
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Davide G Franchina
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Leticia Soriano-Baguet
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Carole Binsfeld
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Charlène Verschueren
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sabine Spath
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland; Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Anouk Ewen
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Eric Koncina
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Jean-Jacques Gérardy
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg
| | - Takumi Kobayashi
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Catherine Dostert
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sophie Farinelle
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Janika Härm
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Yu-Tong Fan
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Isaac S Harris
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland
| | - Michel Mittelbronn
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), 1526 Luxembourg, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
6
|
Cremin M, Ramirez VT, Sanchez K, Tay E, Murray K, Brust-Mascher I, Reardon C. Substance P receptor signaling contributes to host maladaptive responses during enteric bacterial infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.599421. [PMID: 38979288 PMCID: PMC11230291 DOI: 10.1101/2024.06.24.599421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Immune responses in the intestine are intricately balanced to prevent pathogen entry without inducing immunopathology. The nervous system is well-established to interface with the immune system to fine-tune immunity in various organ systems including the gastrointestinal tract. Specialized sensory neurons can detect bacteria, bacterial products, and the resulting inflammation, to coordinate the immune response in the gastrointestinal tract. These sensory neurons release peptide neurotransmitters such as Substance P (SP), to induce both neuronal signaling and localized responses in non-neuronal cells. With this in mind, we assessed the immunoregulatory roles of SP receptor signaling during enteric bacterial infection with the non-invasive pathogen Citrobacter rodentium. Pharmacological antagonism of the SP receptor significantly reduced bacterial burden and prevented colonic crypt hyperplasia. Mice with SP receptor signaling blockade had significantly reduced inflammation and recruitment of T-cells in the colon. Reduced colonic T-cell recruitment is due to reduced expression of adhesion molecules on colonic endothelial cells in SP receptor antagonist-treated mice. Using SP receptor T-cell conditional knockout mice, we further confirmed SP receptor signaling enhanced select aspects of T-cell responses. Our data demonstrates that SP receptor signaling can significantly reduce inflammation and prevent host-maladaptive responses without impinging upon host protection.
Collapse
Affiliation(s)
- Michael Cremin
- UC Davis, School of Veterinary Medicine, Anatomy, Physiology & Cell Biology
| | - Valerie T. Ramirez
- UC Davis, School of Veterinary Medicine, Anatomy, Physiology & Cell Biology
| | - Kristina Sanchez
- University of California, School of Veterinary Medicine, Anatomy, Physiology & Cell Biology
| | - Emmy Tay
- University of California, School of Veterinary Medicine, Anatomy, Physiology & Cell Biology
| | - Kaitlin Murray
- University of California, School of Veterinary Medicine, Anatomy, Physiology & Cell Biology
| | - Ingrid Brust-Mascher
- University of California, School of Veterinary Medicine, Anatomy, Physiology & Cell Biology
| | - Colin Reardon
- UC Davis, School of Veterinary Medicine, Anatomy, Physiology & Cell Biology
| |
Collapse
|
7
|
Hartley VL, Qaqish AM, Wood MJ, Studnicka BT, Iwai K, Liu TC, MacDuff DA. HOIL1 Regulates Group 3 Innate Lymphoid Cells in the Colon and Protects against Systemic Dissemination, Colonic Ulceration, and Lethality from Citrobacter rodentium Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1823-1834. [PMID: 37902285 PMCID: PMC10841105 DOI: 10.4049/jimmunol.2300351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023]
Abstract
Heme-oxidized IRP2 ubiquitin ligase-1 (HOIL1)-deficient patients experience chronic intestinal inflammation and diarrhea as well as increased susceptibility to bacterial infections. HOIL1 is a component of the linear ubiquitin chain assembly complex that regulates immune signaling pathways, including NF-κB-activating pathways. We have shown previously that HOIL1 is essential for survival following Citrobacter rodentium gastrointestinal infection of mice, but the mechanism of protection by HOIL1 was not examined. C. rodentium is an important murine model for human attaching and effacing pathogens, enteropathogenic and enterohemorrhagic Escherichia coli that cause diarrhea and foodborne illnesses and lead to severe disease in children and immunocompromised individuals. In this study, we found that C. rodentium infection resulted in severe colitis and dissemination of C. rodentium to systemic organs in HOIL1-deficient mice. HOIL1 was important in the innate immune response to limit early replication and dissemination of C. rodentium. Using bone marrow chimeras and cell type-specific knockout mice, we found that HOIL1 functioned in radiation-resistant cells and partly in radiation-sensitive cells and in myeloid cells to limit disease, but it was dispensable in intestinal epithelial cells. HOIL1 deficiency significantly impaired the expansion of group 3 innate lymphoid cells and their production of IL-22 during C. rodentium infection. Understanding the role HOIL1 plays in type 3 inflammation and in limiting the pathogenesis of attaching and effacing lesion-forming bacteria will provide further insight into the innate immune response to gastrointestinal pathogens and inflammatory disorders.
Collapse
Affiliation(s)
- Victoria L. Hartley
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Arwa M. Qaqish
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Matthew J. Wood
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Brian T. Studnicka
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Donna A. MacDuff
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
8
|
Latour YL, McNamara KM, Allaman MM, Barry DP, Smith TM, Asim M, Williams KJ, Hawkins CV, Jacobse J, Goettel JA, Delgado AG, Piazuelo MB, Washington MK, Gobert AP, Wilson KT. Myeloid deletion of talin-1 reduces mucosal macrophages and protects mice from colonic inflammation. Sci Rep 2023; 13:22368. [PMID: 38102166 PMCID: PMC10724268 DOI: 10.1038/s41598-023-49614-z] [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: 04/07/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023] Open
Abstract
The intestinal immune response is crucial in maintaining a healthy gut, but the enhanced migration of macrophages in response to pathogens is a major contributor to disease pathogenesis. Integrins are ubiquitously expressed cellular receptors that are highly involved in immune cell adhesion to endothelial cells while in the circulation and help facilitate extravasation into tissues. Here we show that specific deletion of the Tln1 gene encoding the protein talin-1, an integrin-activating scaffold protein, from cells of the myeloid lineage using the Lyz2-cre driver mouse reduces epithelial damage, attenuates colitis, downregulates the expression of macrophage markers, decreases the number of differentiated colonic mucosal macrophages, and diminishes the presence of CD68-positive cells in the colonic mucosa of mice infected with the enteric pathogen Citrobacter rodentium. Bone marrow-derived macrophages lacking expression of Tln1 did not exhibit a cell-autonomous phenotype; there was no impaired proinflammatory gene expression, nitric oxide production, phagocytic ability, or surface expression of CD11b, CD86, or major histocompatibility complex II in response to C. rodentium. Thus, we demonstrate that talin-1 plays a role in the manifestation of infectious colitis by increasing mucosal macrophages, with an effect that is independent of macrophage activation.
Collapse
Affiliation(s)
- Yvonne L Latour
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 2215B Garland Ave., 1030C MRB IV, Nashville, TN, 37232-0252, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kara M McNamara
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Margaret M Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel P Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thaddeus M Smith
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kamery J Williams
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caroline V Hawkins
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin Jacobse
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 2215B Garland Ave., 1030C MRB IV, Nashville, TN, 37232-0252, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeremy A Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 2215B Garland Ave., 1030C MRB IV, Nashville, TN, 37232-0252, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 2215B Garland Ave., 1030C MRB IV, Nashville, TN, 37232-0252, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alain P Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 2215B Garland Ave., 1030C MRB IV, Nashville, TN, 37232-0252, USA.
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA.
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
9
|
Cremin M, Tay EXY, Ramirez VT, Murray K, Nichols RK, Brust-Mascher I, Reardon C. TRPV1 controls innate immunity during Citrobacter rodentium enteric infection. PLoS Pathog 2023; 19:e1011576. [PMID: 38109366 PMCID: PMC10758261 DOI: 10.1371/journal.ppat.1011576] [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: 07/26/2023] [Revised: 01/01/2024] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
Mucosal immunity is critical to host protection from enteric pathogens and must be carefully controlled to prevent immunopathology. Regulation of immune responses can occur through a diverse range of mechanisms including bi-directional communication with neurons. Among which include specialized sensory neurons that detect noxious stimuli due to the expression of transient receptor potential vanilloid receptor 1 (TRPV1) ion channel and have a significant role in the coordination of host-protective responses to enteric bacterial pathogens. Here we have used the mouse-adapted attaching and effacing pathogen Citrobacter rodentium to assess the specific role of TRPV1 in coordinating the host response. TRPV1 knockout (TRPV1-/-) mice had a significantly higher C. rodentium burden in the distal colon and fecal pellets compared to wild-type (WT) mice. Increased bacterial burden was correlated with significantly increased colonic crypt hyperplasia and proliferating intestinal epithelial cells in TRPV1-/- mice compared to WT. Despite the increased C. rodentium burden and histopathology, the recruitment of colonic T cells producing IFNγ, IL-17, or IL-22 was similar between TRPV1-/- and WT mice. In evaluating the innate immune response, we identified that colonic neutrophil recruitment in C. rodentium infected TRPV1-/- mice was significantly reduced compared to WT mice; however, this was independent of neutrophil development and maturation within the bone marrow compartment. TRPV1-/- mice were found to have significantly decreased expression of the neutrophil-specific chemokine Cxcl6 and the adhesion molecules Icam1 in the distal colon compared to WT mice. Corroborating these findings, a significant reduction in ICAM-1 and VCAM-1, but not MAdCAM-1 protein on the surface of colonic blood endothelial cells from C. rodentium infected TRPV1-/- mice compared to WT was observed. These findings demonstrate the critical role of TRPV1 in regulating the host protective responses to enteric bacterial pathogens, and mucosal immune responses.
Collapse
Affiliation(s)
- Michael Cremin
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Emmy Xue Yun Tay
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Valerie T. Ramirez
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Kaitlin Murray
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Rene K. Nichols
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Colin Reardon
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| |
Collapse
|
10
|
Cremin M, Tay E, Ramirez VT, Murray K, Nichols RK, Brust-Mascher I, Reardon C. TRPV1 controls innate immunity during Citrobacter rodentium enteric infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550772. [PMID: 37546968 PMCID: PMC10402119 DOI: 10.1101/2023.07.26.550772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Mucosal immunity is critical to host protection from enteric pathogens and must be carefully controlled to prevent immunopathology. Regulation of immune responses can occur through a diverse range of mechanisms including bi-directional communication with the neurons. Among which include specialized sensory neurons that detect noxious stimuli due to the expression of transient receptor potential vanilloid receptor 1 (TRPV1) ion channel and have a significant role in the coordination of host-protective responses to enteric bacterial pathogens. Here we have used the mouse-adapted attaching and effacing pathogen Citrobacter rodentium to assess the specific role of the TRPV1 channel in coordinating the host response. TRPV1 knockout (TRPV1-/-) mice had a significantly higher C. rodentium burden in the distal colon and fecal pellets compared to wild-type (WT) mice. Increased bacterial burden was correlated with significantly increased colonic crypt hyperplasia and proliferating intestinal epithelial cells in TRPV1-/- mice compared to WT. Despite the increased C. rodentium burden and histopathology, the recruitment of colonic T cells producing IFNγ, IL-17, or IL-22 was similar between TRPV1-/- and WT mice. In evaluating the innate immune response, we identified that colonic neutrophil recruitment in C. rodentium infected TRPV1-/- mice was significantly reduced compared to WT mice; however, this was independent of neutrophil development and maturation within the bone marrow compartment. TRPV1-/- mice were found to have significantly decreased expression of the neutrophil-specific chemokine Cxcl6 and the adhesion molecules Icam1 in the distal colon compared to WT mice. Corroborating these findings, a significant reduction in ICAM-1 and VCAM-1, but not MAdCAM-1 protein on the surface of colonic blood endothelial cells from C. rodentium infected TRPV1-/- mice compared to WT was observed. These findings demonstrate the critical role of TRPV1 in regulating the host protective responses to enteric bacterial pathogens, and mucosal immune responses.
Collapse
Affiliation(s)
- Michael Cremin
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Emmy Tay
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Valerie T. Ramirez
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Kaitlin Murray
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Rene K. Nichols
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Colin Reardon
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| |
Collapse
|
11
|
Liang Q, Ma C, Crowley SM, Allaire JM, Han X, Chong RWW, Packer NH, Yu HB, Vallance BA. Sialic acid plays a pivotal role in licensing Citrobacter rodentium's transition from the intestinal lumen to a mucosal adherent niche. Proc Natl Acad Sci U S A 2023; 120:e2301115120. [PMID: 37399418 PMCID: PMC10334811 DOI: 10.1073/pnas.2301115120] [Citation(s) in RCA: 12] [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: 01/24/2023] [Accepted: 05/30/2023] [Indexed: 07/05/2023] Open
Abstract
Enteric bacterial pathogens pose significant threats to human health; however, the mechanisms by which they infect the mammalian gut in the face of daunting host defenses and an established microbiota remain poorly defined. For the attaching and effacing (A/E) bacterial family member and murine pathogen Citrobacter rodentium, its virulence strategy likely involves metabolic adaptation to the host's intestinal luminal environment, as a necessary precursor to reach and infect the mucosal surface. Suspecting this adaptation involved the intestinal mucus layer, we found that C. rodentium was able to catabolize sialic acid, a monosaccharide derived from mucins, and utilize it as its sole carbon source for growth. Moreover, C. rodentium also sensed and displayed chemotactic activity toward sialic acid. These activities were abolished when the nanT gene, encoding a sialic acid transporter, was deleted (ΔnanT). Correspondingly, the ΔnanT C. rodentium strain was significantly impaired in its ability to colonize the murine intestine. Intriguingly, sialic acid was also found to induce the secretion of two autotransporter proteins, Pic and EspC, which possess mucinolytic and host-adherent properties. As a result, sialic acid enhanced the ability of C. rodentium to degrade intestinal mucus (through Pic), as well as to adhere to intestinal epithelial cells (through EspC). We thus demonstrate that sialic acid, a monosaccharide constituent of the intestinal mucus layer, functions as an important nutrient and a key signal for an A/E bacterial pathogen to escape the colonic lumen and directly infect its host's intestinal mucosa.
Collapse
Affiliation(s)
- Qiaochu Liang
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Caixia Ma
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Shauna M. Crowley
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Joannie M. Allaire
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Xiao Han
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Raymond W. W. Chong
- ARC Centre of Excellence for Synthetic Biology, School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, North Ryde, Sydney, NSW2109, Australia
| | - Nicolle H. Packer
- ARC Centre of Excellence for Synthetic Biology, School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, North Ryde, Sydney, NSW2109, Australia
| | - Hong Bing Yu
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Bruce A. Vallance
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| |
Collapse
|
12
|
Bonetti L, Horkova V, Longworth J, Guerra L, Kurniawan H, Franchina DG, Soriano-Baguet L, Grusdat M, Spath S, Koncina E, Ewen A, Binsfeld C, Verschueren C, Gérardy JJ, Kobayashi T, Dostert C, Farinelle S, Härm J, Chen Y, Harris IS, Lang PA, Vasiliou V, Waisman A, Letellier E, Becher B, Mittelbronn M, Brenner D. A Th17 cell-intrinsic glutathione/mitochondrial-IL-22 axis protects against intestinal inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.06.547932. [PMID: 37489135 PMCID: PMC10363291 DOI: 10.1101/2023.07.06.547932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Although the intestinal tract is a major site of reactive oxygen species (ROS) generation, the mechanisms by which antioxidant defense in gut T cells contribute to intestinal homeostasis are currently unknown. Here we show, using T cell-specific ablation of the catalytic subunit of glutamate cysteine ligase (Gclc), that the ensuing loss of glutathione (GSH) impairs the production of gut-protective IL-22 by Th17 cells within the lamina propria. Although Gclc ablation does not affect T cell cytokine secretion in the gut of mice at steady-state, infection with C. rodentium increases ROS, inhibits mitochondrial gene expression and mitochondrial function in Gclc-deficient Th17 cells. These mitochondrial deficits affect the PI3K/AKT/mTOR pathway, leading to reduced phosphorylation of the translation repressor 4E-BP1. As a consequence, the initiation of translation is restricted, resulting in decreased protein synthesis of IL-22. Loss of IL-22 results in poor bacterial clearance, enhanced intestinal damage, and high mortality. ROS-scavenging, reconstitution of IL-22 expression or IL-22 supplementation in vivo prevent the appearance of these pathologies. Our results demonstrate the existence of a previously unappreciated role for Th17 cell-intrinsic GSH coupling to promote mitochondrial function, IL-22 translation and signaling. These data reveal an axis that is essential for maintaining the integrity of the intestinal barrier and protecting it from damage caused by gastrointestinal infection.
Collapse
Affiliation(s)
- Lynn Bonetti
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Veronika Horkova
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Luana Guerra
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Henry Kurniawan
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Davide G. Franchina
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Leticia Soriano-Baguet
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Melanie Grusdat
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sabine Spath
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland
- Center for Fundamental Immunology, Benaroya Research Institute; Seattle, WA 98101, USA
| | - Eric Koncina
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Anouk Ewen
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Carole Binsfeld
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Charlène Verschueren
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Jean-Jacques Gérardy
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, L-3555, Luxembourg
| | - Takumi Kobayashi
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Catherine Dostert
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sophie Farinelle
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Janika Härm
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Isaac S. Harris
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty Heinrich Heine University Düsseldorf, Germany
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland
| | - Michel Mittelbronn
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, L-3555, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, L-4362, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Luxembourg, L-1526, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
13
|
Chen GY, Thorup NR, Miller AJ, Li YC, Ayres JS. Cooperation between physiological defenses and immune resistance produces asymptomatic carriage of a lethal bacterial pathogen. SCIENCE ADVANCES 2023; 9:eadg8719. [PMID: 37352357 PMCID: PMC10289649 DOI: 10.1126/sciadv.adg8719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/18/2023] [Indexed: 06/25/2023]
Abstract
Animals evolved two defense strategies to survive infections. Antagonistic strategies include immune resistance mechanisms that operate to kill invading pathogens. Cooperative or physiological defenses mediate host adaptation to the infected state, limiting physiological damage and disease, without killing the pathogen, and have been shown to cause asymptomatic carriage and transmission of lethal pathogens. Here, we demonstrate that physiological defenses cooperate with the adaptive immune response to generate long-term asymptomatic carriage of the lethal enteric murine pathogen, Citrobacter rodentium. Asymptomatic carriage of genetically virulent C. rodentium provided immune resistance against subsequent infections. Immune protection was dependent on systemic antibody responses and pathogen virulence behavior rather than the recognition of specific virulent antigens. Last, we demonstrate that an avirulent strain of C. rodentium in the field has background mutations in genes that are important for LPS structure. Our work reveals insight into how asymptomatic infections can arise mechanistically with immune resistance, mediating exclusion of phenotypically virulent enteric pathogen to promote asymptomatic carriage.
Collapse
Affiliation(s)
- Grischa Y. Chen
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Natalia R. Thorup
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Abigail J. Miller
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Yao-Cheng Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Janelle S. Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
14
|
Zhang W, Cao X, Zhong X, Wu H, Shi Y, Feng M, Wang YC, Ann D, Gwack Y, Yuan YC, Shang W, Sun Z. SRC2 controls CD4 + T cell activation via stimulating c-Myc-mediated upregulation of amino acid transporter Slc7a5. Proc Natl Acad Sci U S A 2023; 120:e2221352120. [PMID: 37094160 PMCID: PMC10160970 DOI: 10.1073/pnas.2221352120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/16/2023] [Indexed: 04/26/2023] Open
Abstract
T cell activation stimulates substantially increased protein synthesis activity to accumulate sufficient biomass for cell proliferation. The protein synthesis is fueled by the amino acids transported from the environment. Steroid nuclear receptor coactivator 2 (SRC2) is a member of a family of transcription coactivators. Here, we show that SRC2 recruited by c-Myc enhances CD4+ T cell activation to stimulate immune responses via upregulation of amino acid transporter Slc7a5. Mice deficient of SRC2 in T cells (SRC2fl/fl/CD4Cre) are resistant to the induction of experimental autoimmune encephalomyelitis (EAE) and susceptible to Citrobacter rodentium (C. rodentium) infection. Adoptive transfer of naive CD4+ T cells from SRC2fl/fl/CD4Cre mice fails to elicit EAE and colitis in Rag1/ recipients. Further, CD4+ T cells from SRC2fl/fl/CD4Cre mice display defective T cell proliferation, cytokine production, and differentiation both in vitro and in vivo. Mechanically, SRC2 functions as a coactivator to work together with c-Myc to stimulate the expression of amino acid transporter Slc7a5 required for T cell activation. Slc7a5 fails to be up-regulated in CD4+ T cells from SRC2fl/fl/CD4Cre mice, and forced expression of Slc7a5 rescues proliferation, cytokine production, and the ability of SRC2fl/fl/CD4Cre CD4+ T cells to induce EAE. Therefore, SRC2 is essential for CD4+ T cell activation and, thus, a potential drug target for controlling CD4+ T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Wencan Zhang
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - Xiancai Zhong
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - Hongmin Wu
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - Yun Shi
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - Yi-Chang Wang
- Department of Diabetes Complication and Metabolism, Arthur Rigs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - David Ann
- Department of Diabetes Complication and Metabolism, Arthur Rigs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA91010
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Yate-Ching Yuan
- Division of Translational Bioinformatic, Bioinformatics Core, City of Hope, Duarte, CA91010
| | - Weirong Shang
- Department of Gynecology and Obsterics, School of Medicine, Emory University, Atlanta, GA30322
| | - Zuoming Sun
- Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA91010
| |
Collapse
|
15
|
Chen GY, Thorup NR, Miller AJ, Li YC, Ayres JS. Cooperation between physiological defenses and immune resistance produces asymptomatic carriage of a lethal bacterial pathogen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525099. [PMID: 36711884 PMCID: PMC9882269 DOI: 10.1101/2023.01.22.525099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Animals have evolved two defense strategies to survive infections. Antagonistic strategies include mechanisms of immune resistance that operate to sense and kill invading pathogens. Cooperative or physiological defenses mediate host adaptation to the infected state, limiting physiological damage and disease, without killing the pathogen, and have been shown to cause asymptomatic carriage and transmission of lethal pathogens. Here we demonstrate that physiological defenses cooperate with the adaptive immune response to generate long-term asymptomatic carriage of the lethal enteric murine pathogen, Citrobacter rodentium. Asymptomatic carriage of genetically virulent C. rodentium provided immune resistance against subsequent infections. Host immune protection was dependent on systemic antibody responses and pathogen virulence behavior, rather than the recognition of specific virulent factor antigens. Finally, we demonstrate that an avirulent strain of C. rodentium in the field has background mutations in two genes that are important for LPS structure. Our work reveals novel insight into how asymptomatic infections can arise mechanistically with immune resistance, mediating exclusion of phenotypically virulent enteric pathogen to promote asymptomatic carriage.
Collapse
Affiliation(s)
- Grischa Y Chen
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Natalia R Thorup
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Abigail J Miller
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Yao-Cheng Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Janelle S Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| |
Collapse
|
16
|
Tsai K, Ma C, Han X, Allaire J, Healey GR, Crowley SM, Yu H, Jacobson K, Xia L, Priatel JJ, Vallance BA. Highly Sensitive, Flow Cytometry-Based Measurement of Intestinal Permeability in Models of Experimental Colitis. Cell Mol Gastroenterol Hepatol 2023; 15:425-438. [PMID: 36244647 PMCID: PMC9791122 DOI: 10.1016/j.jcmgh.2022.10.004] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND & AIMS Increased intestinal permeability is seen in a variety of inflammatory conditions such as enteric infections and inflammatory bowel disease. Because barrier function can provide a key biomarker of disease severity, it often is assayed in animal models. A common methodology involves gavaging mice with fluorescein isothiocyanate-conjugated dextran (FITC-D), followed by cardiac puncture to assay plasma fluorescence on a spectrophotometer. Although the FITC-D method is relatively simple, its sensitivity is limited and enables only a single measurement because the test requires killing the subject. Herein, we describe a novel flow cytometry-based method of intestinal permeability measurement based on detection of orally gavaged ovalbumin (OVA) that leaks out of the gut. Our approach uses minute blood volumes collected from the tail vein, permitting repeated testing of the same subject at multiple time points. By comparing this assay against the gold standard FITC-D method, we show the expanded utility of our OVA assay in measuring intestinal permeability. METHODS We directly compared our OVA assay against the FITC-D assay by co-administering both probes orally to the same animals and subsequently using their respective methodologies to measure intestinal permeability by detecting probe levels in the plasma. Permeability was assessed in mice genetically deficient in intestinal mucus production or glycosylation. In addition, wild-type mice undergoing dextran sodium sulfate-induced colitis or infected by the enteric bacterial pathogen Citrobacter rodentium also were tested. RESULTS The OVA assay showed very high efficacy in all animal models of intestinal barrier dysfunction tested. Besides identifying intestinal barrier dysfunction in mice with impaired mucin glycosylation, the assay also allowed for repeated tracking of intestinal permeability within the same animal over time, providing data that cannot be easily acquired with other currently applied methods. CONCLUSIONS The OVA assay is a highly sensitive and effective method of measuring intestinal permeability in mouse models of barrier dysfunction and experimental colitis.
Collapse
Affiliation(s)
- Kevin Tsai
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Caixia Ma
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiao Han
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joannie Allaire
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Genelle R Healey
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shauna M Crowley
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hongbing Yu
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevan Jacobson
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - John J Priatel
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce A Vallance
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
17
|
Latour YL, Allaman MM, Barry DP, Smith TM, Williams KJ, McNamara KM, Jacobse J, Goettel JA, Delgado AG, Piazuelo MB, Zhao S, Gobert AP, Wilson KT. Epithelial talin-1 protects mice from citrobacter rodentium-induced colitis by restricting bacterial crypt intrusion and enhancing t cell immunity. Gut Microbes 2023; 15:2192623. [PMID: 36951501 PMCID: PMC10038039 DOI: 10.1080/19490976.2023.2192623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/13/2023] [Indexed: 03/24/2023] Open
Abstract
Pathogenic enteric Escherichia coli present a significant burden to global health. Food-borne enteropathogenic E. coli (EPEC) and Shiga toxin-producing E. coli (STEC) utilize attaching and effacing (A/E) lesions and actin-dense pedestal formation to colonize the gastrointestinal tract. Talin-1 is a large structural protein that links the actin cytoskeleton to the extracellular matrix though direct influence on integrins. Here we show that mice lacking talin-1 in intestinal epithelial cells (Tln1Δepi) have heightened susceptibility to colonic disease caused by the A/E murine pathogen Citrobacter rodentium. Tln1Δepi mice exhibit decreased survival, and increased colonization, colon weight, and histologic colitis compared to littermate Tln1fl/fl controls. These findings were associated with decreased actin polymerization and increased infiltration of innate myeloperoxidase-expressing immune cells, confirmed as neutrophils by flow cytometry, but more bacterial dissemination deep into colonic crypts. Further evaluation of the immune population recruited to the mucosa in response to C. rodentium revealed that loss of Tln1 in colonic epithelial cells (CECs) results in impaired recruitment and activation of T cells. C. rodentium infection-induced colonic mucosal hyperplasia was exacerbated in Tln1Δepi mice compared to littermate controls. We demonstrate that this is associated with decreased CEC apoptosis and crowding of proliferating cells in the base of the glands. Taken together, talin-1 expression by CECs is important in the regulation of both epithelial renewal and the inflammatory T cell response in the setting of colitis caused by C. rodentium, suggesting that this protein functions in CECs to limit, rather than contribute to the pathogenesis of this enteric infection.
Collapse
Affiliation(s)
- Yvonne L. Latour
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Margaret M. Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel P. Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thaddeus M. Smith
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kamery J. Williams
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kara M. McNamara
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Justin Jacobse
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeremy A. Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G. Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alain P. Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T. Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
- Medical Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
18
|
Lecron JC, Charreau S, Jégou JF, Salhi N, Petit-Paris I, Guignouard E, Burucoa C, Favot-Laforge L, Bodet C, Barra A, Huguier V, Mcheik J, Dumoutier L, Garnier J, Bernard FX, Ryffel B, Morel F. IL-17 and IL-22 are pivotal cytokines to delay wound healing of S. aureus and P. aeruginosa infected skin. Front Immunol 2022; 13:984016. [PMID: 36275755 PMCID: PMC9585169 DOI: 10.3389/fimmu.2022.984016] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionAlthough the presence of pathogens in skin wounds is known to delay the wound healing process, the mechanisms underlying this delay remain poorly understood. In the present study, we have investigated the regulatory role of proinflammatory cytokines on the healing kinetics of infected wounds.MethodsWe have developed a mouse model of cutaneous wound healing, with or without wound inoculation with Staphylococcus aureus and Pseudomonas aeruginosa, two major pathogens involved in cutaneous wound bacterial infections.ResultsAseptic excision in C57BL/6 mouse skin induced early expression of IL-1β, TNFα and Oncostatin M (OSM), without detectable expression of IL-22 and IL-17A/F. S. aureus and P. aeruginosa wound inoculation not only increased the expression of IL-1β and OSM, but also induced a strong cutaneous expression of IL-22, IL-17A and IL-17F, along with an increased number of infiltrating IL-17A and/or IL-22-producing γδ T cells. The same cytokine expression pattern was observed in infected human skin wounds. When compared to uninfected wounds, mouse skin infection delayed the wound healing process. Injection of IL-1α, TNFα, OSM, IL-22 and IL-17 together in the wound edges induced delayed wound healing similar to that induced by the bacterial infection. Wound healing experiments in infected Rag2KO mice (deficient in lymphocytes) showed a wound healing kinetic similar to uninfected Rag2KO mice or WT mice. Rag2KO infected-skin lesions expressed lower levels of IL-17 and IL-22 than WT, suggesting that the expression of these cytokines is mainly dependent on γδ T cells in this model. Wound healing was not delayed in infected IL-17R/IL-22KO, comparable to uninfected control mice. Injection of recombinant IL-22 and IL-17 in infected wound edges of Rag2KO mice re-establish the delayed kinetic of wound healing, as in infected WT mice.ConclusionThese results demonstrate the synergistic and specific effects of IL-22 and IL-17 induced by bacterial infection delay the wound healing process, regardless of the presence of bacteria per se. Therefore, these cytokines play an unexpected role in delayed skin wound healing.
Collapse
Affiliation(s)
- Jean-Claude Lecron
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire Immunologie et Inflammation, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
- *Correspondence: Jean-Claude Lecron,
| | - Sandrine Charreau
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - Jean-François Jégou
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Nadjet Salhi
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Isabelle Petit-Paris
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Emmanuel Guignouard
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Christophe Burucoa
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire de Bactériologie, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Laure Favot-Laforge
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Anne Barra
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire Immunologie et Inflammation, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Vincent Huguier
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Service de Chirurgie Plastique, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Jiad Mcheik
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Service de Chirurgie Pédiatrique, Centre Hospitalier et Universitaire CHU) de Poitiers, Poitiers, France
| | - Laure Dumoutier
- De Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Julien Garnier
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - François-Xavier Bernard
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - Bernhard Ryffel
- Laboratoire d'Immunologie et Neurogénétique Expérimentales et Moléculaire (INEM) - Unité Mixte de Recherche (UMR) 7355, Centre National de la Recherche Scientifique (CNRS) et Université d’Orléans, Orléans, France
| | - Franck Morel
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| |
Collapse
|
19
|
Interferon regulatory factor 1 (IRF-1) promotes intestinal group 3 innate lymphoid responses during Citrobacter rodentium infection. Nat Commun 2022; 13:5730. [PMID: 36175404 PMCID: PMC9522774 DOI: 10.1038/s41467-022-33326-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are crucial mediators of immunity and epithelial barrier function during immune responses against extracellular bacteria. Here, we identify Interferon regulatory factor 1 (IRF-1), a transcription factor previously associated with type 1 immunity, as an essential regulator of intestinal ILC3 accumulation and effector cytokine production. We demonstrate that IRF-1 is upregulated in the context of infection with the enteropathogen Citrobacter rodentium and that its presence is central for anatomical containment and prevention of pathogen dissemination. We furthermore show that IRF-1 is required in order for intestinal ILC3s to produce large amounts of the protective effector cytokine IL-22 early in the course of infection. On a molecular level, our data indicate that IRF-1 controls ILC3 numbers and their activation by direct transcriptional regulation of the IL-12Rβ1 chain, thereby allowing ILCs to physiologically respond to IL-23 stimulation. Innate lymphoid cells (ILC) are involved with different immune responses. Here the authors show that Interferon regulatory factor 1 (IRF1) is important for intestinal ILC3 accumulation during Citrobacter rodentium infection and promotes release of the protective cytokine IL-22 and response to IL-23.
Collapse
|
20
|
Slack E, Diard M. Resistance is futile? Mucosal immune mechanisms in the context of microbial ecology and evolution. Mucosal Immunol 2022; 15:1188-1198. [PMID: 36329192 PMCID: PMC9705250 DOI: 10.1038/s41385-022-00574-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
In the beginning it was simple: we injected a protein antigen and studied the immune responses against the purified protein. This elegant toolbox uncovered thousands of mechanisms via which immune cells are activated. However, when we consider immune responses against real infectious threats, this elegant simplification misses half of the story: the infectious agents are typically evolving orders-of-magnitude faster than we are. Nowhere is this more pronounced than in the mammalian large intestine. A bacterium representing only 0.1% of the human gut microbiota will have a population size of 109 clones, each actively replicating. Moreover, the evolutionary pressure from other microbes is at least as profound as direct effects of the immune system. Therefore, to really understand intestinal immune mechanisms, we need to understand both the host response and how rapid microbial evolution alters the apparent outcome of the response. In this review we use the examples of intestinal inflammation and secretory immunoglobulin A (SIgA) to highlight what is already known (Fig. 1). Further, we will explore how these interactions can inform immunotherapy and prophylaxis. This has major implications for how we design effective mucosal vaccines against increasingly drug-resistant bacterial pathogens Fig. 1 THE IMMUNE RESPONSE SHAPES THE FITNESS LANDSCAPE IN THE GASTRO-INTESTINAL TRACT.: The red arrows depict possible evolutionary paths of a novel colonizer along adaptive peaks in the intestinal fitness landscapes that change with the status of the host immune system. The flat surfaces represent the non-null fitness baselines (values x or y) at which a bacterium can establish at minimum carrying capacity. a In the healthy gut, metabolic competence, resistance to aggressions by competitors and predators, swift adaptation to rapid fluctuations as well as surviving acidic pH and the flow of the intestinal content, represent potent selective pressures and as many opportunities for bacteria to increase fitness by phenotypic or genetic variations. b When pathogens trigger acute inflammation, bacteria must adapt to iron starvation, killing by immune cells and antimicrobial peptides, and oxidative stress, while new metabolic opportunities emerge. c When high-affinity SIgA are produced against a bacterium, e.g., after oral vaccination, escape of SIgA by altering or losing surface epitopes becomes crucial for maximum fitness. However, escaping polyvalent SIgA responses after vaccination with "evolutionary trap" vaccines leads to evolutionary trade-offs: A fitness maximum is reached in the vaccinated host gut that represents a major disadvantage for transmission into naïve hosts (fitness diminished below x) (d).
Collapse
Affiliation(s)
- Emma Slack
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland.
- Botnar Research Institute for Child Health, Basel, Switzerland.
| | - Médéric Diard
- Botnar Research Institute for Child Health, Basel, Switzerland.
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
21
|
Transcriptional Profiling of the Small Intestine and the Colon Reveals Modulation of Gut Infection with Citrobacter rodentium According to the Vitamin A Status. Nutrients 2022; 14:nu14081563. [PMID: 35458125 PMCID: PMC9026425 DOI: 10.3390/nu14081563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/10/2022] Open
Abstract
Vitamin A (VA) deficiency and diarrheal diseases are both serious public health issues worldwide. VA deficiency is associated with impaired intestinal barrier function and increased risk of mucosal infection-related mortality. The bioactive form of VA, retinoic acid, is a well-known regulator of mucosal integrity. Using Citrobacter rodentium-infected mice as a model for diarrheal diseases in humans, previous studies showed that VA-deficient (VAD) mice failed to clear C. rodentium as compared to their VA-sufficient (VAS) counterparts. However, the distinct intestinal gene responses that are dependent on the host’s VA status still need to be discovered. The mRNAs extracted from the small intestine (SI) and the colon were sequenced and analyzed on three levels: differential gene expression, enrichment, and co-expression. C. rodentium infection interacted differentially with VA status to alter colon gene expression. Novel functional categories downregulated by this pathogen were identified, highlighted by genes related to the metabolism of VA, vitamin D, and ion transport, including improper upregulation of Cl− secretion and disrupted HCO3− metabolism. Our results suggest that derangement of micronutrient metabolism and ion transport, together with the compromised immune responses in VAD hosts, may be responsible for the higher mortality to C. rodentium under conditions of inadequate VA.
Collapse
|
22
|
Zindl CL, Witte SJ, Laufer VA, Gao M, Yue Z, Janowski KM, Cai B, Frey BF, Silberger DJ, Harbour SN, Singer JR, Turner H, Lund FE, Vallance BA, Rosenberg AF, Schoeb TR, Chen JY, Hatton RD, Weaver CT. A nonredundant role for T cell-derived interleukin 22 in antibacterial defense of colonic crypts. Immunity 2022; 55:494-511.e11. [PMID: 35263568 PMCID: PMC9126440 DOI: 10.1016/j.immuni.2022.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 11/11/2021] [Accepted: 02/04/2022] [Indexed: 02/05/2023]
Abstract
Interleukin (IL)-22 is central to immune defense at barrier sites. We examined the contributions of innate lymphoid cell (ILC) and T cell-derived IL-22 during Citrobacter rodentium (C.r) infection using mice that both report Il22 expression and allow lineage-specific deletion. ILC-derived IL-22 activated STAT3 in C.r-colonized surface intestinal epithelial cells (IECs) but only temporally restrained bacterial growth. T cell-derived IL-22 induced a more robust and extensive activation of STAT3 in IECs, including IECs lining colonic crypts, and T cell-specific deficiency of IL-22 led to pathogen invasion of the crypts and increased mortality. This reflected a requirement for T cell-derived IL-22 for the expression of a host-protective transcriptomic program that included AMPs, neutrophil-recruiting chemokines, and mucin-related molecules, and it restricted IFNγ-induced proinflammatory genes. Our findings demonstrate spatiotemporal differences in the production and action of IL-22 by ILCs and T cells during infection and reveal an indispensable role for IL-22-producing T cells in the protection of the intestinal crypts.
Collapse
Affiliation(s)
- Carlene L Zindl
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Steven J Witte
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Vincent A Laufer
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Min Gao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Informatics Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zongliang Yue
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Informatics Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen M Janowski
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Baiyi Cai
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Blake F Frey
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel J Silberger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stacey N Harbour
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeffrey R Singer
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Henrietta Turner
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Frances E Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Bruce A Vallance
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3V4, Canada
| | - Alexander F Rosenberg
- Informatics Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Trenton R Schoeb
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jake Y Chen
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Informatics Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robin D Hatton
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
23
|
Khoobchandani M, Khan A, Katti KK, Thipe VC, Al-Yasiri AY, MohanDoss DKD, Nicholl MB, Lugão AB, Hans CP, Katti KV. Green nanotechnology of MGF-AuNPs for immunomodulatory intervention in prostate cancer therapy. Sci Rep 2021; 11:16797. [PMID: 34408231 PMCID: PMC8373987 DOI: 10.1038/s41598-021-96224-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Men with castration-resistant prostate cancer (CRPC) face poor prognosis and increased risk of treatment-incurred adverse effects resulting in one of the highest mortalities among patient population globally. Immune cells act as double-edged sword depending on the tumor microenvironment, which leads to increased infiltration of pro-tumor (M2) macrophages. Development of new immunomodulatory therapeutic agents capable of targeting the tumor microenvironment, and hence orchestrating the transformation of pro-tumor M2 macrophages to anti-tumor M1, would substantially improve treatment outcomes of CRPC patients. We report, herein, Mangiferin functionalized gold nanoparticulate agent (MGF-AuNPs) and its immunomodulatory characteristics in treating prostate cancer. We provide evidence of immunomodulatory intervention of MGF-AuNPs in prostate cancers through observations of enhanced levels of anti-tumor cytokines (IL-12 and TNF-α) with concomitant reductions in the levels of pro-tumor cytokines (IL-10 and IL-6). In the MGF-AuNPs treated groups, IL-12 was elevated to ten-fold while TNF-α was elevated to about 50-fold, while IL-10 and IL-6 were reduced by two-fold. Ability of MGF-AuNPs to target splenic macrophages is invoked via targeting of NF-kB signaling pathway. Finally, therapeutic efficacy of MGF-AuNPs, in treating prostate cancer in vivo in tumor bearing mice, is described taking into consideration various immunomodulatory interventions triggered by this green nanotechnology-based nanomedicine agent.
Collapse
Affiliation(s)
- Menka Khoobchandani
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO, 65212, USA
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA
| | - Aslam Khan
- Department of Biochemistry, University of Missouri, Columbia, MO, 65212, USA
| | - Kavita K Katti
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO, 65212, USA
| | - Velaphi C Thipe
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear, IPEN/CNEN-SP, Butantã, São Paulo, SP, Brasil
| | - Amal Y Al-Yasiri
- Nuclear Science and Engineering Institute (NSEI), University of Missouri, Columbia, MO, 65211, USA
- College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Darsha K D MohanDoss
- Dhanvantari Nano Ayushadi Pvt Ltd, No. 8/34, Neelakanta Mehta Street, T. Nagar, Chennai, 600017, India
| | | | - Ademar B Lugão
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear, IPEN/CNEN-SP, Butantã, São Paulo, SP, Brasil
| | - Chetan P Hans
- Department of Medicine-Cardiology, University of Missouri, Columbia, MO, 65212, USA
| | - Kattesh V Katti
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO, 65212, USA.
- Department of Physics, University of Missouri, Columbia, MO, 65212, USA.
- University of Missouri Research Reactor (MURR), University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
24
|
DCLK1 isoforms and aberrant Notch signaling in the regulation of human and murine colitis. Cell Death Discov 2021; 7:169. [PMID: 34226497 PMCID: PMC8257684 DOI: 10.1038/s41420-021-00526-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/10/2021] [Accepted: 04/07/2021] [Indexed: 01/19/2023] Open
Abstract
Alternative promoter usage generates long and short isoforms (DCLK1-L and DCLK1-S) of doublecortin-like kinase-1 (DCLK1). Tight control of Notch signaling is important to prevent and restitute inflammation in the intestine. Our aim was to investigate whether Notch1–DCLK1 axis regulates the mucosal immune responses to infection and whether this is phenocopied in human models of colitis. In the FFPE (formalin-fixed paraffin-embedded) sections prepared from the colons of ulcerative colitis (UC) and immune-mediated colitis (IRAEC) patients, expression of DCLK1 isoforms correlated positively with Notch1 and negatively with a transcriptional repressor, FoxD3 (Forkhead Box D3). DCLK1 protein staining in these sections was predominantly sub-epithelial (stromal) wherein DCLK1 co-localized with NICD, CD68, CD11c, and neutrophil elastase (NE). NE also co-stained with Citrullinated-H3 indicating the presence of neutrophil extracellular traps. In human neutrophils, elevated levels of DCLK1-S, CXCL-10, Ly6G, MPO, NE, and Notch1/2 in LPS-treated cells were inhibited when LPS was added in conjunction with Notch blocker dibenzazepine (DBZ; LPS + DBZ group). In CR-infected Rag1−/− mice, higher levels of DCLK1 in the colonic crypts were inhibited when mice received DBZ for 10 days coincident with significant dysbiosis, barrier disruption, and colitis. Concurrently, DCLK1 immunoreactivity shifted toward the stroma in CR + DBZ mice with predominance of DCLK1-S that coincided with higher Notch1 levels. Upon antibiotic treatment, partial restoration of crypt DCLK1, reduction in MPO activity, and increased survival followed. When intestinal epithelial cell-specific Dclk1-knockout (Dclk1ΔIEC) or Dclk1ΔIEC;Rag1−/− double knockout (DKO) mice were infected with CR and given a single dose of DBZ, they developed barrier defect and severe colitis with higher levels of stromal DCLK1-S, Ly6G, NE, and Notch1. We therefore propose that, by regulating the mucosal immune responses, the Notch–DCLK1 axis may be integral to the development of murine or human colitis.
Collapse
|
25
|
Cox CB, Storm EE, Kapoor VN, Chavarria-Smith J, Lin DL, Wang L, Li Y, Kljavin N, Ota N, Bainbridge TW, Anderson K, Roose-Girma M, Warming S, Arron JR, Turley SJ, de Sauvage FJ, van Lookeren Campagne M. IL-1R1-dependent signaling coordinates epithelial regeneration in response to intestinal damage. Sci Immunol 2021; 6:eabe8856. [PMID: 33963061 DOI: 10.1126/sciimmunol.abe8856] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 04/08/2021] [Indexed: 12/29/2022]
Abstract
Repair of the intestinal epithelium is tightly regulated to maintain homeostasis. The response after epithelial damage needs to be local and proportional to the insult. How different types of damage are coupled to repair remains incompletely understood. We report that after distinct types of intestinal epithelial damage, IL-1R1 signaling in GREM1+ mesenchymal cells increases production of R-spondin 3 (RSPO3), a Wnt agonist required for intestinal stem cell self-renewal. In parallel, IL-1R1 signaling regulates IL-22 production by innate lymphoid cells and promotes epithelial hyperplasia and regeneration. Although the regulation of both RSPO3 and IL-22 is critical for epithelial recovery from Citrobacter rodentium infection, IL-1R1-dependent RSPO3 production by GREM1+ mesenchymal cells alone is sufficient and required for recovery after dextran sulfate sodium-induced colitis. These data demonstrate how IL-1R1-dependent signaling orchestrates distinct repair programs tailored to the type of injury sustained that are required to restore intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Christian B Cox
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Elaine E Storm
- Department of Molecular Oncology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Varun N Kapoor
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | | | - David L Lin
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Lifen Wang
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Yun Li
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Noelyn Kljavin
- Department of Molecular Oncology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Naruhisa Ota
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Travis W Bainbridge
- Department of Protein Chemistry, Genentech Inc., South San Francisco, CA 94080, USA
| | - Keith Anderson
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Søren Warming
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Joseph R Arron
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Frederic J de Sauvage
- Department of Molecular Oncology, Genentech Inc., South San Francisco, CA 94080, USA.
| | | |
Collapse
|
26
|
Mamareli P, Kruse F, Lu CW, Guderian M, Floess S, Rox K, Allan DSJ, Carlyle JR, Brönstrup M, Müller R, Berod L, Sparwasser T, Lochner M. Targeting cellular fatty acid synthesis limits T helper and innate lymphoid cell function during intestinal inflammation and infection. Mucosal Immunol 2021; 14:164-176. [PMID: 32355319 DOI: 10.1038/s41385-020-0285-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 03/13/2020] [Accepted: 03/24/2020] [Indexed: 02/04/2023]
Abstract
CD4+ T cells contribute critically to a protective immune response during intestinal infections, but have also been implicated in the aggravation of intestinal inflammatory pathology. Previous studies suggested that T helper type (Th)1 and Th17 cells depend on de novo fatty acid (FA) synthesis for their development and effector function. Here, we report that T-cell-specific targeting of the enzyme acetyl-CoA carboxylase 1 (ACC1), a major checkpoint controlling FA synthesis, impaired intestinal Th1 and Th17 responses by limiting CD4+ T-cell expansion and infiltration into the lamina propria in murine models of colitis and infection-associated intestinal inflammation. Importantly, pharmacological inhibition of ACC1 by the natural compound soraphen A mirrored the anti-inflammatory effects of T-cell-specific targeting, but also enhanced susceptibility toward infection with C. rodentium. Further analysis revealed that deletion of ACC1 in RORγt+ innate lymphoid cells (ILC), but not dendritic cells or macrophages, decreased resistance to infection by interfering with IL-22 production and intestinal barrier function. Together, our study suggests pharmacological targeting of ACC1 as an effective approach for metabolic immune modulation of T-cell-driven intestinal inflammatory responses, but also reveals an important role of ACC1-mediated lipogenesis for the function of RORγt+ ILC.
Collapse
Affiliation(s)
- Panagiota Mamareli
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany.,Institute of Medical Microbiology and Hygiene, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Friederike Kruse
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Chia-Wen Lu
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany.,Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Melanie Guderian
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany.,Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Stefan Floess
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - David S J Allan
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - James R Carlyle
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research, Helmholtz Centre for Infection Research, Saarland University, Saarbrücken, Germany.,Department of Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | - Luciana Berod
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany.,Institute of Medical Microbiology and Hygiene, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany. .,Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
27
|
Wu Y, Wang J, He Q, Yu L, Pham Q, Cheung L, Zhang Z, Kim YS, Smith AD, Wang TTY. Dietary Indole-3-Carbinol Alleviated Spleen Enlargement, Enhanced IgG Response in C3H/HeN Mice Infected with Citrobacter rodentium. Nutrients 2020; 12:E3148. [PMID: 33076301 PMCID: PMC7602481 DOI: 10.3390/nu12103148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/06/2020] [Accepted: 10/11/2020] [Indexed: 12/21/2022] Open
Abstract
Enteropathogenic and enterohemorrhagic Escherichia coli are important enteric pathogens that induce hemorrhagic colitis or even fatal hemolytic uremic syndrome. Emerging evidence shows that some bio-actives derived from fruits and vegetables may serve as alternatives to antibiotics for overcoming multidrug resistant E. coli infections. In this study, the Citrobacter rodentium (Cr) infection model was utilized to mimic E. coli-induced acute intestinal inflammation, and the effects of a cruciferous vegetable-derived cancer protective compound, indole-3-carbinol (I3C), on the immune responses of Cr-susceptible C3H/HeN mice were investigated. Dietary I3C significantly inhibited the loss of body weight and the increase in spleen size in Cr infected mice. In addition, I3C treatment reduced the inflammatory response to Cr infection by maintaining anti-inflammatory cytokine IL-22 mRNA levels while reducing expression of other pro-inflammatory cytokines including IL17A, IL6, IL1β, TNF-α, and IFN-γ. Moreover, the serum cytokine levels of IL17, TNF-α, IL12p70, and G-CSF also were down-regulated by I3C in Cr-infected mice. Additionally, dietary I3C specifically enhanced the Cr-specific IgG response to Cr infection. In general, dietary I3C reduced the Cr-induced pro-inflammatory response in susceptible C3H/HeN mice and alleviated the physiological changes and tissue damage induced by Cr infection but not Cr colonization.
Collapse
Affiliation(s)
- Yanbei Wu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology & Business University, Beijing 100048, China; (Y.W.); (J.W.)
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA; (L.Y.); (Z.Z.)
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China;
| | - Jing Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology & Business University, Beijing 100048, China; (Y.W.); (J.W.)
| | - Qiang He
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China;
| | - Liangli Yu
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA; (L.Y.); (Z.Z.)
| | - Quynhchi Pham
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| | - Lumei Cheung
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| | - Zhi Zhang
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA; (L.Y.); (Z.Z.)
| | - Young S. Kim
- Nutritional Science Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Allen D. Smith
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| | - Thomas T. Y. Wang
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| |
Collapse
|
28
|
Wu Y, He Q, Yu L, Pham Q, Cheung L, Kim YS, Wang TTY, Smith AD. Indole-3-Carbinol Inhibits Citrobacter rodentium Infection through Multiple Pathways Including Reduction of Bacterial Adhesion and Enhancement of Cytotoxic T Cell Activity. Nutrients 2020; 12:E917. [PMID: 32230738 PMCID: PMC7230886 DOI: 10.3390/nu12040917] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/24/2022] Open
Abstract
Intestinal inflammation is associated with an increased risk of developing colorectal cancer and may result from dysregulated responses to commensal bacteria or exposure to bacterial pathogens. Dietary modulation of intestinal inflammation may protect against development of colon cancer. However, the precise diet-derived components and underlying mechanisms remain elusive. Citrobacter rodentium (Cr) induces acute intestinal inflammation and has been used to study the role of inflammation in the susceptibility to colon cancer. Here we examine the effects of indole-3-carbinol (I3C), a dietary compound with anticarcinogenic properties, on intestinal immune and inflammatory responses to Cr infection and adhesion to colonic cells in vitro. C57BL/6J mice were fed a diet with/without 1 μmol/g I3C and infected with Cr. Compared to infected mice fed with a control diet, consumption of a 1 μmol I3C/g diet significantly reduced fecal excretion of Cr, Cr colonization of the colon, and reduced colon crypt hyperplasia. Furthermore, expression of Cr-induced inflammatory markers such as IL-17A, IL-6, and IL1β were attenuated in infected mice fed with the I3C diet, compared to mice fed a control diet. The expression of cytotoxic T cell markers CD8 and FasL mRNA were increased in I3C-fed infected mice. In-vitro, I3C inhibited Cr growth and adhesion to Caco-2 cells. I3C alleviates Cr-induced murine colitis through multiple mechanisms including inhibition of Cr growth and adhesion to colonic cells in vitro and enhancement of cytotoxic T cell activity.
Collapse
Affiliation(s)
- Yanbei Wu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology & Business University, Beijing 100048, China;
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University, Beijing 100048, China
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| | - Qiang He
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China;
| | - Liangli Yu
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA;
| | - Quynhchi Pham
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| | - Lumei Cheung
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| | - Young S. Kim
- Nutritional Science Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Thomas T. Y. Wang
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| | - Allen D. Smith
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, USDA-ARS, Beltsville, MD 20705, USA; (Q.P.); (L.C.)
| |
Collapse
|
29
|
Sayed IM, Suarez K, Lim E, Singh S, Pereira M, Ibeawuchi SR, Katkar G, Dunkel Y, Mittal Y, Chattopadhyay R, Guma M, Boland BS, Dulai PS, Sandborn WJ, Ghosh P, Das S. Host engulfment pathway controls inflammation in inflammatory bowel disease. FEBS J 2020; 287:3967-3988. [PMID: 32003126 DOI: 10.1111/febs.15236] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 12/20/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022]
Abstract
Chronic diseases, including inflammatory bowel disease (IBD) urgently need new biomarkers as a significant proportion of patients, do not respond to current medications. Inflammation is a common factor in these diseases, and microbial sensing in the intestinal tract is critical to initiate the inflammation. We have identified ELMO1 (engulfment and cell motility protein 1) as a microbial sensor in epithelial and phagocytic cells that turns on inflammatory signals. Using a stem cell-based 'gut-in-a-dish' coculture model, we studied the interactions between microbes, epithelium, and monocytes in the context of IBD. To mimic the in vivo cell physiology, enteroid-derived monolayers (EDMs) were generated from the organoids isolated from WT and ELMO1-/- mice and colonic biopsies of IBD patients. The EDMs were infected with the IBD-associated microbes to monitor the inflammatory responses. ELMO1-depleted EDMs displayed a significant reduction in bacterial internalization, a decrease in pro-inflammatory cytokine productions and monocyte recruitment. The expression of ELMO1 is elevated in the colonic epithelium and in the inflammatory infiltrates within the lamina propria of IBD patients where the higher expression is positively correlated with the elevated expression of pro-inflammatory cytokines, MCP-1 and TNF-α. MCP-1 is released from the epithelium and recruits monocytes to the site of inflammation. Once recruited, monocytes require ELMO1 to engulf the bacteria and propagate a robust TNF-α storm. These findings highlight that the dysregulated epithelial ELMO1 → MCP-1 axis can serve as an early biomarker in the diagnostics of IBD and other inflammatory disorders.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, CA, USA
| | - Katherine Suarez
- Department of Pathology, University of California, San Diego, CA, USA
| | - Eileen Lim
- Department of Pathology, University of California, San Diego, CA, USA
| | - Sujay Singh
- Department of Pathology, University of California, San Diego, CA, USA
| | - Matheus Pereira
- Department of Pathology, University of California, San Diego, CA, USA
| | | | - Gajanan Katkar
- Department of Cellular & Molecular Medicine, University of California, San Diego, CA, USA
| | - Ying Dunkel
- Department of Medicine, University of California, San Diego, CA, USA
| | - Yash Mittal
- Department of Medicine, University of California, San Diego, CA, USA
| | - Ranajoy Chattopadhyay
- Department of Cellular & Molecular Medicine, University of California, San Diego, CA, USA
| | - Monica Guma
- Department of Medicine, University of California, San Diego, CA, USA
| | - Brigid S Boland
- Department of Medicine, University of California, San Diego, CA, USA
| | - Parambir S Dulai
- Department of Medicine, University of California, San Diego, CA, USA
| | | | - Pradipta Ghosh
- Department of Medicine, University of California, San Diego, CA, USA.,Department of Cellular & Molecular Medicine, University of California, San Diego, CA, USA
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA, USA
| |
Collapse
|
30
|
Amadou Amani S, Lang ML. Bacteria That Cause Enteric Diseases Stimulate Distinct Humoral Immune Responses. Front Immunol 2020; 11:565648. [PMID: 33042146 PMCID: PMC7524877 DOI: 10.3389/fimmu.2020.565648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
Bacterial enteric pathogens individually and collectively represent a serious global health burden. Humoral immune responses following natural or experimentally-induced infections are broadly appreciated to contribute to pathogen clearance and prevention of disease recurrence. Herein, we have compared observations on humoral immune mechanisms following infection with Citrobacter rodentium, the model for enteropathogenic Escherichia coli, Vibrio cholerae, Shigella species, Salmonella enterica species, and Clostridioides difficile. A comparison of what is known about the humoral immune responses to these pathogens reveals considerable variance in specific features of humoral immunity including establishment of high affinity, IgG class-switched memory B cell and long-lived plasma cell compartments. This article suggests that such variance could be contributory to persistent and recurrent disease.
Collapse
|
31
|
Luu M, Romero R, Bazant J, Abass E, Hartmann S, Leister H, Fischer F, Mahdavi R, Plaza-Sirvent C, Schmitz I, Steinhoff U, Visekruna A. The NF-κB transcription factor c-Rel controls host defense against Citrobacter rodentium. Eur J Immunol 2019; 50:292-294. [PMID: 31724737 DOI: 10.1002/eji.201948314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/14/2019] [Accepted: 11/12/2019] [Indexed: 01/03/2023]
Abstract
Mice lacking CD4+ T cells or B cells are highly susceptible to Citrobacter rodentium infection. In this study, we show that the activity of the transcription factor c-Rel in lymphocytes is crucial for clearance of C. rodentium. Mice deficient for c-Rel fail to generate protective antibodies and to eradicate the pathogen.
Collapse
Affiliation(s)
- Maik Luu
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Rossana Romero
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Jasmin Bazant
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Elfadil Abass
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sabrina Hartmann
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Hanna Leister
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Florence Fischer
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Rouzbeh Mahdavi
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Carlos Plaza-Sirvent
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ingo Schmitz
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Systems-Oriented Immunology and Inflammation Research Group, Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ulrich Steinhoff
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
32
|
Saha P, Yeoh BS, Xiao X, Golonka RM, Singh V, Wang Y, Vijay-Kumar M. PAD4-dependent NETs generation are indispensable for intestinal clearance of Citrobacter rodentium. Mucosal Immunol 2019; 12:761-771. [PMID: 30710097 PMCID: PMC6519124 DOI: 10.1038/s41385-019-0139-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 02/04/2023]
Abstract
Peptidyl arginine deiminase-4 (PAD4) is indispensable for generation of neutrophil extracellular traps (NETs), which can provide antimicrobial effects during host innate immune response; however, the role of PAD4 against gastrointestinal infection is largely unknown. Herein, we challenged PAD4-deficient (Pad4-/-) mice and wild-type (WT) littermates with Citrobacter rodentium (CR), and investigated bacteria clearance and gut pathology. Luminal colonization of CR in Pad4-/- mice peaked between 11-14 days post-infection, whereas WT mice suppressed the infection by 14 days. We demonstrated that Pad4-/- mice were unable to form NETs, whereas WT mice showed increased NETs formation in the colon during infection. Pad4-/- mice showed aggravated CR-associated inflammation as indicated by elevated systemic and colonic pro-inflammatory markers. Histological analysis revealed that transmissible colonic hyperplasia, goblet cell depletion, and apoptotic cell death were more pronounced in the colon of CR-infected Pad4-/- mice. Treating WT mice with deoxyribonuclease I, which can disrupt NETs generation, recapitulated the exacerbated CR infection and gut pathology associated with the loss of PAD4. Administration of the PAD4 inhibitor, Cl-amidine also aggravated CR infection, but to a lesser extent. Taken together, our findings highlight the importance of PAD4 in the mucosal clearance of CR and in resolving gut-associated inflammation.
Collapse
Affiliation(s)
- Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Beng San Yeoh
- Graduate Program in Immunology & Infectious Disease, The Pennsylvania State University, University Park, Philadelphia, PA, 16802, USA
| | - Xia Xiao
- Division of Nephrology, MGH, Harvard Medical School, Boston, MA, 02114, USA
| | - Rachel M Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Vishal Singh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Yanming Wang
- College of Life Sciences & Medicine, Henan University, Kaifeng, 475004, China
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| |
Collapse
|
33
|
Gaudino SJ, Kumar P. Cross-Talk Between Antigen Presenting Cells and T Cells Impacts Intestinal Homeostasis, Bacterial Infections, and Tumorigenesis. Front Immunol 2019; 10:360. [PMID: 30894857 PMCID: PMC6414782 DOI: 10.3389/fimmu.2019.00360] [Citation(s) in RCA: 274] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/12/2019] [Indexed: 11/21/2022] Open
Abstract
Innate immunity is maintained in part by antigen presenting cells (APCs) including dendritic cells, macrophages, and B cells. APCs interact with T cells to link innate and adaptive immune responses. By displaying bacterial and tumorigenic antigens on their surface via major histocompatibility complexes, APCs can directly influence the differentiation of T cells. Likewise, T cell activation, differentiation, and effector functions are modulated by APCs utilizing multiple mechanisms. The objective of this review is to describe how APCs interact with and influence the activation of T cells to maintain innate immunity during exposure to microbial infection and malignant cells. How bacteria and cancer cells take advantage of some of these interactions for their own benefit will also be discussed. While this review will cover a broad range of topics, a general focus will be held around pathogens, cancers, and interactions that typically occur within the gastrointestinal tract.
Collapse
Affiliation(s)
- Stephen J Gaudino
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| | - Pawan Kumar
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
34
|
Solaymani-Mohammadi S, Berzofsky JA. Interleukin 21 collaborates with interferon-γ for the optimal expression of interferon-stimulated genes and enhances protection against enteric microbial infection. PLoS Pathog 2019; 15:e1007614. [PMID: 30818341 PMCID: PMC6413951 DOI: 10.1371/journal.ppat.1007614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 03/12/2019] [Accepted: 02/03/2019] [Indexed: 12/18/2022] Open
Abstract
The mucosal surface of the intestinal tract represents a major entry route for many microbes. Despite recent progress in the understanding of the IL-21/IL-21R signaling axis in the generation of germinal center B cells, the roles played by this signaling pathway in the context of enteric microbial infections is not well-understood. Here, we demonstrate that Il21r-/- mice are more susceptible to colonic microbial infection, and in the process discovered that the IL-21/IL-21R signaling axis surprisingly collaborates with the IFN-γ/IFN-γR signaling pathway to enhance the expression of interferon-stimulated genes (ISGs) required for protection, via amplifying activation of STAT1 in mucosal CD4+ T cells in a murine model of Citrobacter rodentium colitis. As expected, conditional deletion of STAT3 in CD4+ T cells indicated that STAT3 also contributed importantly to host defense against C. rodentium infection in the colon. However, the collaboration between IL-21 and IFN-γ to enhance the phosphorylation of STAT1 and upregulate ISGs was independent of STAT3. Unveiling this previously unreported crosstalk between these two cytokine networks and their downstream genes induced will provide insight into the development of novel therapeutic targets for colonic infections, inflammatory bowel disease, and promotion of mucosal vaccine efficacy.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail: (SSM); (JAB)
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail: (SSM); (JAB)
| |
Collapse
|
35
|
Wang B, Lim JH, Kajikawa T, Li X, Vallance BA, Moutsopoulos NM, Chavakis T, Hajishengallis G. Macrophage β2-Integrins Regulate IL-22 by ILC3s and Protect from Lethal Citrobacter rodentium-Induced Colitis. Cell Rep 2019; 26:1614-1626.e5. [PMID: 30726742 PMCID: PMC6404229 DOI: 10.1016/j.celrep.2019.01.054] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/07/2018] [Accepted: 01/15/2019] [Indexed: 12/13/2022] Open
Abstract
β2-integrins promote neutrophil recruitment to infected tissues and are crucial for host defense. Neutrophil recruitment is defective in leukocyte adhesion deficiency type-1 (LAD1), a condition caused by mutations in the CD18 (β2-integrin) gene. Using a model of Citrobacter rodentium (CR)-induced colitis, we show that CD18-/- mice display increased intestinal damage and systemic bacterial burden, compared to littermate controls, ultimately succumbing to infection. This phenotype is not attributed to defective neutrophil recruitment, as it is shared by CXCR2-/- mice that survive CR infection. CR-infected CD18-/- mice feature prominent upregulation of IL-17 and downregulation of IL-22. Exogenous IL-22 administration, but not endogenous IL-17 neutralization, protects CD18-/- mice from lethal colitis. β2-integrin expression on macrophages is mechanistically linked to Rac1/ROS-mediated induction of noncanonical-NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome-dependent IL-1β production, which promotes ILC3-derived IL-22. Therefore, β2-integrins are required for protective IL-1β-dependent IL-22 responses in colitis, and the identified mechanism may underlie the association of human LAD1 with colitis.
Collapse
Affiliation(s)
- Baomei Wang
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA.
| | - Jong-Hyung Lim
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA
| | - Tetsuhiro Kajikawa
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA
| | - Xiaofei Li
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA
| | - Bruce A Vallance
- Department of Pediatrics, Division of Gastroenterology, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | | | - Triantafyllos Chavakis
- Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - George Hajishengallis
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Ahmed I, Roy BC, Raach RMT, Owens SM, Xia L, Anant S, Sampath V, Umar S. Enteric infection coupled with chronic Notch pathway inhibition alters colonic mucus composition leading to dysbiosis, barrier disruption and colitis. PLoS One 2018; 13:e0206701. [PMID: 30383855 PMCID: PMC6211731 DOI: 10.1371/journal.pone.0206701] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/12/2018] [Indexed: 12/19/2022] Open
Abstract
Intestinal mucus layer disruption and gut microflora modification in conjunction with tight junction (TJ) changes can increase colonic permeability that allows bacterial dissemination and intestinal and systemic disease. We showed previously that Citrobacter rodentium (CR)-induced colonic crypt hyperplasia and/or colitis is regulated by a functional cross-talk between the Notch and Wnt/β-catenin pathways. In the current study, mucus analysis in the colons of CR-infected (108 CFUs) and Notch blocker Dibenzazepine (DBZ, i.p.; 10μmol/Kg b.w.)-treated mice revealed significant alterations in the composition of trace O-glycans and complex type and hybrid N-glycans, compared to CR-infected mice alone that preceded/accompanied alterations in 16S rDNA microbial community structure and elevated EUB338 staining. While mucin-degrading bacterium, Akkermansia muciniphila (A. muciniphila) along with Enterobacteriaceae belonging to Proteobacteria phyla increased in the feces, antimicrobial peptides Angiogenin-4, Intelectin-1 and Intelectin-2, and ISC marker Dclk1, exhibited dramatic decreases in the colons of CR-infected/DBZ-treated mice. Also evident was a loss of TJ and adherens junction protein immuno-staining within the colonic crypts that negatively impacted paracellular barrier. These changes coincided with the loss of Notch signaling and exacerbation of mucosal injury. In response to a cocktail of antibiotics (Metronidazole/ciprofloxacin) for 10 days, there was increased survival that coincided with: i) decreased levels of Proteobacteria, ii) elevated Dclk1 levels in the crypt and, iii) reduced paracellular permeability. Thus, enteric infections that interfere with Notch activity may promote mucosal dysbiosis that is preceded by changes in mucus composition. Controlled use of antibiotics seems to alleviate gut dysbiosis but may be insufficient to promote colonic crypt regeneration.
Collapse
Affiliation(s)
- Ishfaq Ahmed
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Badal C. Roy
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Rita-Marie T. Raach
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Sarah M. Owens
- Biosciences Division, Argonne National Laboratory, Lemont, Illinois, United States of America
| | - Lijun Xia
- Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center Oklahoma City, Oklahoma, United States of America
| | - Shrikant Anant
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Venkatesh Sampath
- Division of Neonatology, Children’s Mercy Hospital, Kansas City, Missouri, United States of America
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
37
|
Kang E, Crouse A, Chevallier L, Pontier SM, Alzahrani A, Silué N, Campbell-Valois FX, Montagutelli X, Gruenheid S, Malo D. Enterobacteria and host resistance to infection. Mamm Genome 2018; 29:558-576. [PMID: 29785663 DOI: 10.1007/s00335-018-9749-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Enterobacteriaceae are a large family of Gram-negative, non-spore-forming bacteria. Although many species exist as part of the natural flora of animals including humans, some members are associated with both intestinal and extraintestinal diseases. In this review, we focus on members of this family that have important roles in human disease: Salmonella, Escherichia, Shigella, and Yersinia, providing a brief overview of the disease caused by these bacteria, highlighting the contribution of animal models to our understanding of their pathogenesis and of host genetic determinants involved in susceptibility or resistance to infection.
Collapse
Affiliation(s)
- Eugene Kang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
| | - Alanna Crouse
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Lucie Chevallier
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, École Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
- Mouse Genetics Laboratory, Department of Genomes and Genetics, Institut Pasteur, Paris, France
| | - Stéphanie M Pontier
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - Ashwag Alzahrani
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - Navoun Silué
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - François-Xavier Campbell-Valois
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Xavier Montagutelli
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, École Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
| | - Danielle Malo
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
38
|
Kang E, Zhou G, Yousefi M, Cayrol R, Xia J, Gruenheid S. Loss of disease tolerance during Citrobacter rodentium infection is associated with impaired epithelial differentiation and hyperactivation of T cell responses. Sci Rep 2018; 8:847. [PMID: 29339782 PMCID: PMC5770458 DOI: 10.1038/s41598-017-17386-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023] Open
Abstract
Citrobacter rodentium is an intestinal mouse pathogen widely used as a model to study the mucosal response to infection. Inbred mouse strains suffer one of two fates following infection: self-limiting colitis or fatal diarrheal disease. We previously reported that Rspo2 is a major genetic determinant of the outcome of C. rodentium infection; Rspo2 induction during infection of susceptible mice leads to loss of intestinal function and mortality. Rspo2 induction does not impact bacterial colonization, but rather, impedes the ability of the host to tolerate C. rodentium infection. Here, we performed deep RNA sequencing and systematically analyzed the global gene expression profiles of C. rodentium-infected colon tissues from susceptible and resistant congenic mice strains to determine the common responses to infection and the Rspo2-mediated dysfunction pathway signatures associated with loss of disease tolerance. Our results highlight changes in metabolism, tissue remodeling, and host defence as common responses to infection. Conversely, increased Wnt and stem cell signatures, loss of epithelial differentiation, and exaggerated CD4+ T cell activation through increased antigen processing and presentation were specifically associated with the response to infection in susceptible mice. These data provide insights into the molecular mechanisms underlying intestinal dysfunction and disease tolerance during C. rodentium infection.
Collapse
Affiliation(s)
- Eugene Kang
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Guangyan Zhou
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
| | - Mitra Yousefi
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Romain Cayrol
- Department of Pathology and Cellular Biology, University of Montreal, Montreal, Quebec, Canada
| | - Jianguo Xia
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Department of Animal Science, McGill University, Sainte Anne de Bellevue, Quebec, Canada
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
39
|
Bouladoux N, Harrison OJ, Belkaid Y. The Mouse Model of Infection with Citrobacter rodentium. ACTA ACUST UNITED AC 2017; 119:19.15.1-19.15.25. [PMID: 29091261 DOI: 10.1002/cpim.34] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Citrobacter rodentium is a murine mucosal pathogen used as a model to elucidate the molecular and cellular pathogenesis of infection with two clinically important human gastrointestinal pathogens, enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC). C. rodentium infection provides an excellent model to study different aspects of host-pathogen interaction in the gut, including intestinal inflammatory responses during bacteria-induced colitis, mucosal healing and epithelial repair, the induction of mucosal immune responses, and the role of the intestinal microbiota in mediating resistance to colonization by enteric pathogens. This unit provides detailed protocols for growing this bacterium, infecting mice by intragastric inoculation, measuring bacterial loads in feces and organs, and monitoring intestinal pathology induced by infection. Additional protocols describe steps needed to create frozen stocks, establish a growth curve, perform ex vivo organ cultures, isolate immune cells from the large intestine, and measure immune response by flow cytometry. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Nicolas Bouladoux
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland.,NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Oliver J Harrison
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland.,NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| |
Collapse
|
40
|
Antigen-specific regulatory T-cell responses to intestinal microbiota. Mucosal Immunol 2017; 10:1375-1386. [PMID: 28766556 PMCID: PMC5939566 DOI: 10.1038/mi.2017.65] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 06/07/2017] [Indexed: 02/07/2023]
Abstract
The mammalian gastrointestinal tract can harbor both beneficial commensal bacteria important for host health, but also pathogenic bacteria capable of intestinal damage. It is therefore important that the host immune system mount the appropriate immune response to these divergent groups of bacteria-promoting tolerance in response to commensal bacteria and sterilizing immunity in response to pathogenic bacteria. Failure to induce tolerance to commensal bacteria may underlie immune-mediated diseases such as human inflammatory bowel disease. At homeostasis, regulatory T (Treg) cells are a key component of the tolerogenic response by adaptive immunity. This review examines the mechanisms by which intestinal bacteria influence colonic T-cells and B-cell immunoglobulin A (IgA) induction, with an emphasis on Treg cells and the role of antigen-specificity in these processes. In addition to discussing key primary literature, this review highlights current controversies and important future directions.
Collapse
|
41
|
Acuff NV, Li X, Latha K, Nagy T, Watford WT. Tpl2 Promotes Innate Cell Recruitment and Effector T Cell Differentiation To Limit Citrobacter rodentium Burden and Dissemination. Infect Immun 2017; 85:e00193-17. [PMID: 28760932 PMCID: PMC5607429 DOI: 10.1128/iai.00193-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/22/2017] [Indexed: 01/22/2023] Open
Abstract
Tumor progression locus 2 (Tpl2) is a serine-threonine kinase that regulates Th1 differentiation, secretion of the inflammatory cytokine gamma interferon (IFN-γ), and host defense against the intracellular pathogens Toxoplasma gondii, Listeria monocytogenes, and Mycobacterium tuberculosis However, relatively little is known about the contribution of Tpl2 to Th17 differentiation and immune cell function during infection with an extracellular pathogen. The goal of this study was to determine whether Tpl2 influences the immune response generated to the extracellular bacterium Citrobacter rodentium, which induces a mixed Th1 and Th17 response. During peak infection with C. rodentium, Tpl2-/- mice experienced greater bacterial burdens with evidence of dissemination to the liver and spleen but ultimately cleared the bacteria within 3 weeks postinfection, similar to the findings for wild-type mice. Tpl2-/- mice also recruited fewer neutrophils and monocytes to the colon during peak infection, which correlated with increased bacterial burdens. In mixed bone marrow chimeras, Tpl2 was shown to play a T cell-intrinsic role in promoting both IFN-γ and interleukin-17A production during infection with C. rodentium However, upon CD4 T cell transfer into Rag-/- mice, Tpl2-/- CD4 T cells were as protective as wild-type CD4 T cells against the dissemination of bacteria and mortality. These data indicate that the enhanced bacterial burdens in Tpl2-/- mice are not caused primarily by impairments in CD4 T cell function but result from defects in innate immune cell recruitment and function.
Collapse
Affiliation(s)
- Nicole V Acuff
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Xin Li
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Krishna Latha
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Tamas Nagy
- Department of Pathology, University of Georgia, Athens, Georgia, USA
| | - Wendy T Watford
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
42
|
Citrobacter rodentium: a model enteropathogen for understanding the interplay of innate and adaptive components of type 3 immunity. Mucosal Immunol 2017; 10:1108-1117. [PMID: 28612839 PMCID: PMC5969517 DOI: 10.1038/mi.2017.47] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 04/13/2017] [Indexed: 02/07/2023]
Abstract
Citrobacter rodentium is a natural murine intestinal pathogen that shares a core set of virulence factors with the related human pathogens enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC). C. rodentium is now the most widely used small animal model for studying the molecular underpinnings of EPEC and EHEC infections in vivo, including: enterocyte attachment; virulence; colonization resistance; and mucosal immunity. In this review, we discuss type 3 immunity in the context of C. rodentium infection and discuss recent publications that use this model to understand how the innate and adaptive components of immunity intersect to mediate host protection against enteric pathogens and maintain homeostasis with the microbiota.
Collapse
|
43
|
Lu Y, Zhang X, Bouladoux N, Kaul SN, Jin K, Sant'Angelo D, Belkaid Y, Kovalovsky D. Zbtb1 controls NKp46 + ROR-gamma-T + innate lymphoid cell (ILC3) development. Oncotarget 2017; 8:55877-55888. [PMID: 28915559 PMCID: PMC5593530 DOI: 10.18632/oncotarget.19645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/14/2017] [Indexed: 11/25/2022] Open
Abstract
Innate lymphoid cells (ILCs) play a central role conferring protection at the mucosal frontier. In this study, we have identified a requirement of the transcription factor Zbtb1 for the development of RORγt+ ILCs (ILC3s). Zbtb1-deficient mice lacked NKp46+ ILC3 cells in the lamina propria of the small and large intestine. This requirement of Zbtb1 was cell intrinsic, as NKp46+ ILC3s were not generated from Zbtb1-deficient progenitors in bone marrow chimeras and Zbtb1-deficient RORγt+ CCR6−NKp46− ILC3s didn't generate NKp46+ ILC3s in co-cultures with OP9-DL1 stroma. In correlation with this impairment, Zbtb1-deficient ILC3 cells failed to upregulate T-bet expression, and to acquire IFN-γ production characteristic of NKp46+ cells. Finally, absence of NKp46+ILC3 cells combined with the absence of T-cells in Zbtb1-deficient mice, led to a transient susceptibility to C. rodentium infections. Altogether, these results establish that Zbtb1 is essential for the development of NKp46+ ILC3 cells.
Collapse
Affiliation(s)
- Ying Lu
- Experimental Immunology Branch, NCI, NIH, Bethesda, MD, USA
| | - Xianyu Zhang
- Experimental Immunology Branch, NCI, NIH, Bethesda, MD, USA
| | - Nicolas Bouladoux
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | | | - Kangxin Jin
- Zhongshan Ophthalmic Center, State Key Laboratory for Ophthalmic Researches, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Derek Sant'Angelo
- Cancer Metabolism and Growth Program, Rutgers, Child Health Institute of New Jersey, New Brunswick, NJ, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Damian Kovalovsky
- Experimental Immunology Branch, NCI, NIH, Bethesda, MD, USA.,Experimental Transplantation and Immunology Branch, NCI, NIH, Bethesda, MD, USA
| |
Collapse
|
44
|
Buschor S, Cuenca M, Uster SS, Schären OP, Balmer ML, Terrazos MA, Schürch CM, Hapfelmeier S. Innate immunity restricts Citrobacter rodentium A/E pathogenesis initiation to an early window of opportunity. PLoS Pathog 2017; 13:e1006476. [PMID: 28662171 PMCID: PMC5507559 DOI: 10.1371/journal.ppat.1006476] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/12/2017] [Accepted: 06/16/2017] [Indexed: 11/30/2022] Open
Abstract
Citrobacter rodentium infection is a mouse model for the important human diarrheal infection caused by enteropathogenic E. coli (EPEC). The pathogenesis of both species is very similar and depends on their unique ability to form intimately epithelium-adherent microcolonies, also known as "attachment/effacement" (A/E) lesions. These microcolonies must be dynamic and able to self-renew by continuous re-infection of the rapidly regenerating epithelium. It is unknown whether sustained epithelial A/E lesion pathogenesis is achieved through re-infection by planktonic bacteria from the luminal compartment or local spread of sessile bacteria without a planktonic phase. Focusing on the earliest events as C. rodentium becomes established, we show here that all colonic epithelial A/E microcolonies are clonal bacterial populations, and thus depend on local clonal growth to persist. In wild-type mice, microcolonies are established exclusively within the first 18 hours of infection. These early events shape the ongoing intestinal geography and severity of infection despite the continuous presence of phenotypically virulent luminal bacteria. Mechanistically, induced resistance to A/E lesion de-novo formation is mediated by TLR-MyD88/Trif-dependent signaling and is induced specifically by virulent C. rodentium in a virulence gene-dependent manner. Our data demonstrate that the establishment phase of C. rodentium pathogenesis in vivo is restricted to a very short window of opportunity that determines both disease geography and severity.
Collapse
Affiliation(s)
- Stefanie Buschor
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Miguelangel Cuenca
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Stephanie S. Uster
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Olivier P. Schären
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Maria L. Balmer
- Department of Biomedicine, Immunobiology, University of Basel, Basel, Switzerland
| | - Miguel A. Terrazos
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
45
|
MyD88 signaling in dendritic cells and the intestinal epithelium controls immunity against intestinal infection with C. rodentium. PLoS Pathog 2017; 13:e1006357. [PMID: 28520792 PMCID: PMC5433783 DOI: 10.1371/journal.ppat.1006357] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 04/18/2017] [Indexed: 12/29/2022] Open
Abstract
MyD88-mediated signaling downstream of Toll-like receptors and the IL-1 receptor family is critically involved in the induction of protective host responses upon infections. Although it is known that MyD88-deficient mice are highly susceptible to a wide range of bacterial infections, the cell type-specific contribution of MyD88 in protecting the host against intestinal bacterial infection is only poorly understood. In order to investigate the importance of MyD88 in specific immune and nonimmune cell types during intestinal infection, we employed a novel murine knock-in model for MyD88 that enables the cell type-specific reactivation of functional MyD88 expression in otherwise MyD88-deficient mice. We report here that functional MyD88 signaling in CD11c+ cells was sufficient to activate intestinal dendritic cells (DC) and to induce the early group 3 innate lymphoid cell (ILC3) response as well as the development of colonic Th17/Th1 cells in response to infection with the intestinal pathogen C. rodentium. In contrast, restricting MyD88 signaling to several other cell types, including macrophages (MO), T cells or ILC3 did not induce efficient intestinal immune responses upon infection. However, we observed that the functional expression of MyD88 in intestinal epithelial cells (IEC) also partially protected the mice during intestinal infection, which was associated with enhanced epithelial barrier integrity and increased expression of the antimicrobial peptide RegIIIγ and the acute phase protein SAA1 by epithelial cells. Together, our data suggest that MyD88 signaling in DC and IEC is both essential and sufficient to induce a full spectrum of host responses upon intestinal infection with C. rodentium. MyD88-dependent signaling pathways play a critical role in the protective immune response during intestinal infections. However, the significance of MyD88-mediated signaling in specific intestinal immune and nonimmune cell types for the activation of the early innate, adaptive and epithelial host responses upon infection remains poorly understood. Using a novel knock-in mouse model for MyD88, we report here that MyD88 signaling in CD11c+ dendritic cells (DC) is sufficient to activate RORγt+ group 3 innate lymphoid cells (ILC3) as well as Th17/Th1 cells in response to infection with C. rodentium. In contrast, restricting functional MyD88 signaling to several other immune cell types, including macrophages (MO), T cells and ILC3 did not result in intestinal immunity, while expression of MyD88 in intestinal epithelial cells (IEC) mainly enhanced epithelial barrier integrity. Together, our data suggest that MyD88 signaling in DC and IEC is both essential and sufficient to induce a full spectrum of host responses upon intestinal infection with C. rodentium.
Collapse
|
46
|
Lactobacillus delbrueckii subsp. lactis (strain CIDCA 133) stimulates murine macrophages infected with Citrobacter rodentium. World J Microbiol Biotechnol 2017; 33:48. [PMID: 28176201 DOI: 10.1007/s11274-017-2219-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/28/2017] [Indexed: 10/20/2022]
Abstract
Citrobacter rodentium is a specific murine enteropathogen which causes diarrheal disease characterized by colonic hyperplasia and intestinal inflammation. Recruitment of neutrophils and macrophages constitute a key step to control the infection. Since modulation of the activity of professional phagocytic cells could contribute to improve host´s defences against C. rodentium, we investigated the effect of Lactobacillus delbrueckii subsp. lactis (strain CIDCA 133) on the interaction between murine macrophages (RAW 264.7) and C. rodentium. Phagocytosis, surface molecules and inducible nitric oxide synthase (iNOs) expression were determined by flow cytometry. Reactive oxygen species (ROS) were assessed by fluorescence microscopy. The presence of lactobacilli increased phagocytosis of C. rodentium whereas C. rodentium had no effect on lactobacilli internalization. Survival of internalized C. rodentium diminished when strain CIDCA 133 was present. CD-86, MHCII, iNOs expression and nitrite production were increased when C. rodentium and lactobacilli were present even though strain CIDCA 133 alone had no effect. Strain CIDCA 133 led to a strong induction of ROS activity which was not modified by C. rodentium. Lactobacillus delbrueckii subsp. lactis (strain CIDCA 133) is able to increase the activation of murine macrophages infected with C. rodentium. The sole presence of lactobacilli is enough to modify some stimulation markers (e.g. ROS induction) whereas other markers require the presence of both bacteria; thus, indicating a synergistic effect.
Collapse
|
47
|
Lin X, Yang J, Wang J, Huang H, Wang HX, Chen P, Wang S, Pan Y, Qiu YR, Taylor GA, Vallance BA, Gao J, Zhong XP. mTOR is critical for intestinal T-cell homeostasis and resistance to Citrobacter rodentium. Sci Rep 2016; 6:34939. [PMID: 27731345 PMCID: PMC5059740 DOI: 10.1038/srep34939] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/20/2016] [Indexed: 11/25/2022] Open
Abstract
T-cells play an important role in promoting mucosal immunity against pathogens, but the mechanistic basis for their homeostasis in the intestine is still poorly understood. We report here that T-cell-specific deletion of mTOR results in dramatically decreased CD4 and CD8 T-cell numbers in the lamina propria of both small and large intestines under both steady-state and inflammatory conditions. These defects result in defective host resistance against a murine enteropathogen, Citrobacter rodentium, leading to the death of the animals. We further demonstrated that mTOR deficiency reduces the generation of gut-homing effector T-cells in both mesenteric lymph nodes and Peyer’s patches without obviously affecting expression of gut-homing molecules on those effector T-cells. Using mice with T-cell-specific ablation of Raptor/mTORC1 or Rictor/mTORC2, we revealed that both mTORC1 and, to a lesser extent, mTORC2 contribute to both CD4 and CD8 T-cell accumulation in the gastrointestinal (GI) tract. Additionally, mTORC1 but not mTORC2 plays an important role regulating the proliferative renewal of both CD4 and CD8 T-cells in the intestines. Our data thus reveal that mTOR is crucial for T-cell accumulation in the GI tract and for establishing local adaptive immunity against pathogens.
Collapse
Affiliation(s)
- Xingguang Lin
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jialong Yang
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jinli Wang
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongxiang Huang
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hong-Xia Wang
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Pengcheng Chen
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shang Wang
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yun Pan
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yu-Rong Qiu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Gregory A Taylor
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Durham, NC 27705, USA.,Department of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development, Duke University Medical Center, Durham NC 27710, USA.,Department of Molecular Genetics and Microbiology Duke University Medical Center, Durham NC 27710, USA
| | - Bruce A Vallance
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute and the University of British Columbia, Vancouver, British Columbia V6H 3V4, Canada
| | - Jimin Gao
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiao-Ping Zhong
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Immunology, Medical Center, Durham, NC 27710, USA.,Hematologic Malignancies and Cellular Therapies Program, Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
48
|
Kim M, Kim CH. Colonization and effector functions of innate lymphoid cells in mucosal tissues. Microbes Infect 2016; 18:604-614. [PMID: 27365193 PMCID: PMC5050099 DOI: 10.1016/j.micinf.2016.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/11/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
Abstract
Innate lymphoid cells (ILCs) protect mucosal barrier tissues to fight infection and maintain tissue integrity. ILCs and their progenitors are developmentally programmed to migrate, differentiate and populate various mucosal tissues and associated lymphoid tissues. Functionally mature ILC subsets respond to diverse pathogens such as bacteria, viruses, fungi and parasites in subset-specific manners. In this review, we will discuss how ILCs populate mucosal tissues and regulate immune responses to distinct pathogens to protect the host and maintain tissue integrity.
Collapse
Affiliation(s)
- Myunghoo Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Weldon School of Biomedical Engineering, Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Chang H Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Weldon School of Biomedical Engineering, Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
49
|
Lassen KG, McKenzie CI, Mari M, Murano T, Begun J, Baxt LA, Goel G, Villablanca EJ, Kuo SY, Huang H, Macia L, Bhan AK, Batten M, Daly MJ, Reggiori F, Mackay CR, Xavier RJ. Genetic Coding Variant in GPR65 Alters Lysosomal pH and Links Lysosomal Dysfunction with Colitis Risk. Immunity 2016; 44:1392-405. [PMID: 27287411 DOI: 10.1016/j.immuni.2016.05.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/19/2016] [Accepted: 03/21/2016] [Indexed: 12/28/2022]
Abstract
Although numerous polymorphisms have been associated with inflammatory bowel disease (IBD), identifying the function of these genetic factors has proved challenging. Here we identified a role for nine genes in IBD susceptibility loci in antibacterial autophagy and characterized a role for one of these genes, GPR65, in maintaining lysosome function. Mice lacking Gpr65, a proton-sensing G protein-coupled receptor, showed increased susceptibly to bacteria-induced colitis. Epithelial cells and macrophages lacking GPR65 exhibited impaired clearance of intracellular bacteria and accumulation of aberrant lysosomes. Similarly, IBD patient cells and epithelial cells expressing an IBD-associated missense variant, GPR65 I231L, displayed aberrant lysosomal pH resulting in lysosomal dysfunction, impaired bacterial restriction, and altered lipid droplet formation. The GPR65 I231L polymorphism was sufficient to confer decreased GPR65 signaling. Collectively, these data establish a role for GPR65 in IBD susceptibility and identify lysosomal dysfunction as a potentially causative element in IBD pathogenesis with effects on cellular homeostasis and defense.
Collapse
Affiliation(s)
- Kara G Lassen
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Craig I McKenzie
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Muriel Mari
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, 3713 AV Groningen, the Netherlands; Department of Cell Biology, University Medical Center Utrecht, 3564 CX Utrecht, the Netherlands
| | - Tatsuro Murano
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jakob Begun
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Mater Research Institute and School of Medicine, University of Queensland, Brisbane, QLD 4101, Australia
| | - Leigh A Baxt
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gautam Goel
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eduardo J Villablanca
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Szu-Yu Kuo
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hailiang Huang
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Laurence Macia
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Atul K Bhan
- Pathology Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marcel Batten
- Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Mark J Daly
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Fulvio Reggiori
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, 3713 AV Groningen, the Netherlands; Department of Cell Biology, University Medical Center Utrecht, 3564 CX Utrecht, the Netherlands
| | - Charles R Mackay
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Ramnik J Xavier
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
50
|
Solaymani-Mohammadi S, Lakhdari O, Minev I, Shenouda S, Frey BF, Billeskov R, Singer SM, Berzofsky JA, Eckmann L, Kagnoff MF. Lack of the programmed death-1 receptor renders host susceptible to enteric microbial infection through impairing the production of the mucosal natural killer cell effector molecules. J Leukoc Biol 2015; 99:475-82. [PMID: 26467188 DOI: 10.1189/jlb.4a0115-003rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 09/25/2015] [Indexed: 12/26/2022] Open
Abstract
The programmed death-1 receptor is expressed on a wide range of immune effector cells, including T cells, natural killer T cells, dendritic cells, macrophages, and natural killer cells. In malignancies and chronic viral infections, increased expression of programmed death-1 by T cells is generally associated with a poor prognosis. However, its role in early host microbial defense at the intestinal mucosa is not well understood. We report that programmed death-1 expression is increased on conventional natural killer cells but not on CD4(+), CD8(+) or natural killer T cells, or CD11b(+) or CD11c(+) macrophages or dendritic cells after infection with the mouse pathogen Citrobacter rodentium. Mice genetically deficient in programmed death-1 or treated with anti-programmed death-1 antibody were more susceptible to acute enteric and systemic infection with Citrobacter rodentium. Wild-type but not programmed death-1-deficient mice infected with Citrobacter rodentium showed significantly increased expression of the conventional mucosal NK cell effector molecules granzyme B and perforin. In contrast, natural killer cells from programmed death-1-deficient mice had impaired expression of those mediators. Consistent with programmed death-1 being important for intracellular expression of natural killer cell effector molecules, mice depleted of natural killer cells and perforin-deficient mice manifested increased susceptibility to acute enteric infection with Citrobacter rodentium. Our findings suggest that increased programmed death-1 signaling pathway expression by conventional natural killer cells promotes host protection at the intestinal mucosa during acute infection with a bacterial gut pathogen by enhancing the expression and production of important effectors of natural killer cell function.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Omar Lakhdari
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Ivelina Minev
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Steve Shenouda
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Blake F Frey
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Rolf Billeskov
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Steven M Singer
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Jay A Berzofsky
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Lars Eckmann
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| | - Martin F Kagnoff
- *Laboratory of Mucosal Immunology and Department of Medicine, University of California, San Diego, La Jolla, California, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and Department of Biology and Center for Infectious Disease, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|