1
|
Fazelnia K, Fakhraei J, Yarahmadi HM, Amini K. Dietary Supplementation of Potential Probiotics Bacillus subtilis, Bacillus licheniformis, and Saccharomyces cerevisiae and Synbiotic Improves Growth Performance and Immune Responses by Modulation in Intestinal System in Broiler Chicks Challenged with Salmonella Typhimurium. Probiotics Antimicrob Proteins 2021; 13:1081-1092. [PMID: 33459998 DOI: 10.1007/s12602-020-09737-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2020] [Indexed: 11/28/2022]
Abstract
This study evaluates the effects of probiotics and synbiotics on the performance, immune responses, and intestinal morphology, and the expression of immunity-related genes of broiler chicks challenged with Salmonella typhimurium. Three hundred and sixty broiler chicks were divided into six groups, including broiler chicks challenged and non-challenged with S. typhimurium and fed with probiotic, synbiotic, and basal diet without additive. Growth performance (food intake, daily gain, feed conversion ratio, and mortality), immune responses (antibody titer against sheep red blood cells, immunoglobulins G and M), intestinal morphology, lactic acid bacteria population, and the expression of immunity-related genes (interferon-γ, interleukins 6 and 12, and tumor necrosis factor-α) were investigated. The administration of S. typhimurium decreased growth performance (P = 0.0001), immune responses (P = 0.0001), intestinal morphology (P = 0.0001), lactic acid bacteria population (P = 0.0001), and the expression of immunity-related genes (P = 0.0001) of broiler chickens. However, broiler chicks fed with probiotic (P = 0.001) and synbiotic (P = 0.0001) showed better growth performance, immune responses, intestinal morphology, lactic acid bacteria population, and the expression of immunity-related genes in comparison with infected broiler chicks fed with basal diet lack of probiotic and synbiotic. Feeding probiotics (P = 0.001) and synbiotics (P = 0.0001) showed positive effects for challenged and non-challenged broiler chicks. In sum, feeding synbiotic and probiotic alleviated the negative effects of S. typhimurium on growth and immunity of broiler chicks. It can be suggested to apply synbiotic and probiotics as benefit additive against infectious challenges, such as S. typhimurium.
Collapse
Affiliation(s)
- Kambiz Fazelnia
- Department of Animal Science, Arak Branch, Islamic Azad University, Arak, Iran
| | - Jafar Fakhraei
- Department of Animal Science, Arak Branch, Islamic Azad University, Arak, Iran.
| | | | - Kumarss Amini
- Department of Microbiology, Faculty of Basic Sciences, Saveh Branch, Islamic Azad University, Saveh, Iran
| |
Collapse
|
2
|
Dhingra D, Marathe SA, Sharma N, Marathe A, Chakravortty D. Modeling the immune response to Salmonella during typhoid. Int Immunol 2021; 33:281-298. [PMID: 33406267 DOI: 10.1093/intimm/dxab003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 11/13/2022] Open
Abstract
Several facets of the host immune response to Salmonella infection have been studied independently at great depths to understand the progress and pathogenesis of Salmonella infection. The circumstances under which a Salmonella-infected individual succumbs to an active disease, evolves as a persister or clears the infection are not understood in detail. We have adopted a system-level approach to develop a continuous-time mechanistic model. We considered key interactions of the immune system state variables with Salmonella in the mesenteric lymph node to determine the final disease outcome deterministically and exclusively temporally. The model accurately predicts the disease outcomes and immune response trajectories operational during typhoid. The results of the simulation confirm the role of anti-inflammatory (M2) macrophages as a site for persistence and relapsing infection. Global sensitivity analysis highlights the importance of both bacterial and host attributes in influencing the disease outcome. It also illustrates the importance of robust phagocytic and anti-microbial potential of M1 macrophages and dendritic cells (DCs) in controlling the disease. Finally, we propose therapeutic strategies for both antibiotic-sensitive and antibiotic-resistant strains (such as IFN-γ therapy, DC transfer and phagocytic potential stimulation). We also suggest prevention strategies such as improving the humoral response and macrophage carrying capacity, which could complement current vaccination schemes for enhanced efficiency.
Collapse
Affiliation(s)
- Divy Dhingra
- Department of Mechanical Engineering, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Sandhya Amol Marathe
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Nandita Sharma
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Amol Marathe
- Department of Mechanical Engineering, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| |
Collapse
|
3
|
Kerner G, Rosain J, Guérin A, Al-Khabaz A, Oleaga-Quintas C, Rapaport F, Massaad MJ, Ding JY, Khan T, Ali FA, Rahman M, Deswarte C, Martinez-Barricarte R, Geha RS, Jeanne-Julien V, Garcia D, Chi CY, Yang R, Roynard M, Fleckenstein B, Rozenberg F, Boisson-Dupuis S, Ku CL, Seeleuthner Y, Béziat V, Marr N, Abel L, Al-Herz W, Casanova JL, Bustamante J. Inherited human IFN-γ deficiency underlies mycobacterial disease. J Clin Invest 2020; 130:3158-3171. [PMID: 32163377 PMCID: PMC7260033 DOI: 10.1172/jci135460] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/04/2020] [Indexed: 12/30/2022] Open
Abstract
Mendelian susceptibility to mycobacterial disease (MSMD) is characterized by a selective predisposition to clinical disease caused by the Bacille Calmette-Guérin (BCG) vaccine and environmental mycobacteria. The known genetic etiologies of MSMD are inborn errors of IFN-γ immunity due to mutations of 15 genes controlling the production of or response to IFN-γ. Since the first MSMD-causing mutations were reported in 1996, biallelic mutations in the genes encoding IFN-γ receptor 1 (IFN-γR1) and IFN-γR2 have been reported in many patients of diverse ancestries. Surprisingly, mutations of the gene encoding the IFN-γ cytokine itself have not been reported, raising the remote possibility that there might be other agonists of the IFN-γ receptor. We describe 2 Lebanese cousins with MSMD, living in Kuwait, who are both homozygous for a small deletion within the IFNG gene (c.354_357del), causing a frameshift that generates a premature stop codon (p.T119Ifs4*). The mutant allele is loss of expression and loss of function. We also show that the patients' herpesvirus Saimiri-immortalized T lymphocytes did not produce IFN-γ, a phenotype that can be rescued by retrotransduction with WT IFNG cDNA. The blood T and NK lymphocytes from these patients also failed to produce and secrete detectable amounts of IFN-γ. Finally, we show that human IFNG has evolved under stronger negative selection than IFNGR1 or IFNGR2, suggesting that it is less tolerant to heterozygous deleterious mutations than IFNGR1 or IFNGR2. This may account for the rarity of patients with autosomal-recessive, complete IFN-γ deficiency relative to patients with complete IFN-γR1 and IFN-γR2 deficiencies.
Collapse
Affiliation(s)
- Gaspard Kerner
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Jérémie Rosain
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Antoine Guérin
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Ahmad Al-Khabaz
- Allergy and Clinical Immunology Unit, Pediatric Department, Mubarak Al-Kabeer Hospital, Kuwait University, Jabriya City, Kuwait
| | - Carmen Oleaga-Quintas
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Franck Rapaport
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - Michel J. Massaad
- Department of Experimental Pathology, Immunology and Microbiology, and
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Jing-Ya Ding
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | | | | | - Caroline Deswarte
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Rubén Martinez-Barricarte
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - Raif S. Geha
- Division of Immunology, Department of Pediatrics, Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Valentine Jeanne-Julien
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Diane Garcia
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Chih-Yu Chi
- Division of Infectious Diseases, Department of Internal Medicine and
- School of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - Manon Roynard
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Bernhard Fleckenstein
- Institute of Clinical and Molecular Virology, Erlangen-Nurnberg University, Erlangen, Germany
| | - Flore Rozenberg
- Department of Virology, University of Paris, Cochin Hospital, Assistance Publique – Hôpitaux de Paris (AP-HP), Paris, France
| | - Stéphanie Boisson-Dupuis
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - Cheng-Lung Ku
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yoann Seeleuthner
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Vivien Béziat
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - Nico Marr
- Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Laurent Abel
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
- Allergy and Clinical Immunology Unit, Pediatric Department, Al-Sabah Hospital, Kuwait City, Kuwait
| | - Jean-Laurent Casanova
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
- Pediatric Hematology and Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | - Jacinta Bustamante
- INSERM U1163, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM 1163, Paris, France
- Imagine Institute, University of Paris, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
- Center for the Study of Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| |
Collapse
|
4
|
Jordan SJ, Bakshi RK, Brown LT, Chi X, Geisler WM. Stimulated peripheral blood mononuclear cells from chlamydia-infected women release predominantly Th1-polarizing cytokines. Cytokine 2019; 113:458-461. [PMID: 29914793 PMCID: PMC6311343 DOI: 10.1016/j.cyto.2018.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/19/2018] [Accepted: 06/11/2018] [Indexed: 12/21/2022]
Abstract
Chlamydia trachomatis infection (chlamydia) is the most prevalent sexually transmitted bacterial infection and causes significant reproductive morbidity in women. Little is known about how immunity to chlamydia develops in women, though animal models of chlamydia indicate that T-helper type 1 (Th1) responses are important for chlamydia clearance and protective immunity, whereas T-helper type 2 (Th2) responses are associated with persisting infection. In chlamydia-infected women, whether the predominant immune response is Th1- or Th2-polarizing remains controversial. To determine the cytokine profiles elicited by peripheral blood mononuclear cells (PBMCs) from chlamydia-infected women, we stimulated PBMCs with C. trachomatis elementary bodies and recombinant C. trachomatis Pgp3 and measured supernatant levels of select cytokines spanning Th1- and Th2-polarizing responses. We found that stimulated PBMCs from chlamydia-infected women secreted cytokines that indicate strong Th1-polarizing responses, especially interferon-gamma, whereas Th2-polarizing cytokines were expressed at significantly lower levels. In chlamydia-infected women, the predominant cytokine responses elicited on stimulation of PBMCs with C. trachomatis antigens were Th1-polarizing, with interferon-gamma as the predominant cytokine.
Collapse
Affiliation(s)
- Stephen J Jordan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rakesh K Bakshi
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - LaDraka' T Brown
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaofei Chi
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - William M Geisler
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
5
|
Carlin AF, Plummer EM, Vizcarra EA, Sheets N, Joo Y, Tang W, Day J, Greenbaum J, Glass CK, Diamond MS, Shresta S. An IRF-3-, IRF-5-, and IRF-7-Independent Pathway of Dengue Viral Resistance Utilizes IRF-1 to Stimulate Type I and II Interferon Responses. Cell Rep 2018; 21:1600-1612. [PMID: 29117564 DOI: 10.1016/j.celrep.2017.10.054] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/25/2017] [Accepted: 10/13/2017] [Indexed: 01/19/2023] Open
Abstract
Interferon-regulatory factors (IRFs) are a family of transcription factors (TFs) that translate viral recognition into antiviral responses, including type I interferon (IFN) production. Dengue virus (DENV) and other clinically important flaviviruses are suppressed by type I IFN. While mice lacking the type I IFN receptor (Ifnar1-/-) succumb to DENV infection, we found that mice deficient in three transcription factors controlling type I IFN production (Irf3-/-Irf5-/-Irf7-/- triple knockout [TKO]) survive DENV challenge. DENV infection of TKO mice resulted in minimal type I IFN production but a robust type II IFN (IFN-γ) response. Using loss-of-function approaches for various molecules, we demonstrate that the IRF-3-, IRF-5-, IRF-7-independent pathway predominantly utilizes IFN-γ and, to a lesser degree, type I IFNs. This pathway signals via IRF-1 to stimulate interleukin-12 (IL-12) production and IFN-γ response. These results reveal a key antiviral role for IRF-1 by activating both type I and II IFN responses during DENV infection.
Collapse
Affiliation(s)
- Aaron F Carlin
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Emily M Plummer
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Edward A Vizcarra
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Nicholas Sheets
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Yunichel Joo
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - William Tang
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Jeremy Day
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Jay Greenbaum
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael S Diamond
- Departments of Medicine, Pathology and Immunology, and Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sujan Shresta
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA.
| |
Collapse
|
6
|
Silva AF, Matos MP, Ralph MT, Silva DL, de Alencar NM, Ramos MV, Lima-Filho JV. Comparison of immunomodulatory properties of mannose-binding lectins from Canavalia brasiliensis and Cratylia argentea in a mice model of Salmonella infection. Int Immunopharmacol 2016; 31:233-8. [DOI: 10.1016/j.intimp.2015.12.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/06/2015] [Accepted: 12/28/2015] [Indexed: 02/08/2023]
|
7
|
Hu JL, Yu H, Kulkarni RR, Sharif S, Cui SW, Xie MY, Nie SP, Gong J. Modulation of cytokine gene expression by selectedLactobacillusisolates in the ileum, caecal tonsils and spleen ofSalmonella-challenged broilers. Avian Pathol 2015; 44:463-9. [DOI: 10.1080/03079457.2015.1086725] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
8
|
Wei L, Jin Q, Chu Y, Wu S, Huang R. Suppression of dendritic cell and T-cell activation by the pRST⁹⁸ Salmonella plasmid. Mol Med Rep 2014; 11:2306-14. [PMID: 25385146 DOI: 10.3892/mmr.2014.2919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 10/24/2014] [Indexed: 11/06/2022] Open
Abstract
Salmonella evades host immune response via the expression of a variety of pathogenic factors. The 'pRST98' plasmid of Salmonella enterica serotype Typhi (S. Typhi) is involved in conferring the multidrug‑resistance and virulence of S. typhi. However, its specific effect on host‑cell function has remained elusive. Dendritic cells (DCs) are key regulators of immune responses. The present study therefore aimed to investigate whether pRST98 may target DCs involved in mediating the adaptive immune response. In vivo experiments with Salmonella enterica serotype Typhimurium χ3337 and χ3337/pRST98 revealed that pRST98 may influence multiple important functions of murine DCs, including maturation, survival and cytokine production. In addition, pRST98 markedly contributed to decreasing T‑cell activation. These data suggested that by targeting the aforementioned functions of DCs, pRST98 may partially overturn the adaptive immune defense mechanisms of the host, which are required for elimination of this pathogen from infected tissues. This may contribute to the evasion of host adaptive immune responses by S. Typhi and therefore provide a target for the prevention and treatment of typhoid fever.
Collapse
Affiliation(s)
- Li Wei
- Department of Microbiology, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Qili Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233040, P.R. China
| | - Yuanyuan Chu
- Department of Microbiology, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Shuyan Wu
- Department of Microbiology, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Rui Huang
- Department of Microbiology, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
9
|
Wittkopf N, Neurath MF, Becker C. Immune-epithelial crosstalk at the intestinal surface. J Gastroenterol 2014; 49:375-87. [PMID: 24469679 DOI: 10.1007/s00535-013-0929-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 12/18/2013] [Indexed: 02/04/2023]
Abstract
The intestinal tract is one of the most complex organs of the human body. It has to exercise various functions including food and water absorption, as well as barrier and immune regulation. These functions affect not only the gut itself, but influence the overall health of the organism. Diseases involving the gastrointestinal tract such as inflammatory bowel disease and colorectal cancer therefore severely affect the patient's quality of life and can become life-threatening. Intestinal epithelial cells (IECs) play an important role in intestinal inflammation, infection, and cancer development. IECs not only constitute the first barrier in the gut against the lumen, they also constantly signal information about the gut lumen to immune cells, thereby influencing their behaviour. In contrast, by producing various antimicrobial peptides, IECs shape the microbial community within the gut. IECs also respond to cytokines and other mediators of immune cells in the lamina propria. Interactions between epithelial cells and immune cells in the intestine are responsible for gut homeostasis, and modulations of this crosstalk have been reported in studies of gut diseases. This review discusses the wide field of immune-epithelial interactions and shows the importance of immune-epithelial crosstalk in the intestine to gut homeostasis and the overall health status.
Collapse
Affiliation(s)
- Nadine Wittkopf
- Department of Medicine 1, Friedrich-Alexander-University, 91052, Erlangen, Germany,
| | | | | |
Collapse
|
10
|
Baron J, Bin-Tarif A, Herbert R, Frost L, Taylor G, Baron MD. Early changes in cytokine expression in peste des petits ruminants disease. Vet Res 2014; 45:22. [PMID: 24559207 PMCID: PMC3941941 DOI: 10.1186/1297-9716-45-22] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/14/2014] [Indexed: 12/25/2022] Open
Abstract
Peste des petits ruminants is a viral disease of sheep and goats that has spread through most of Africa as well as the Middle East and the Indian subcontinent. Although, the spread of the disease and its economic impact has made it a focus of international concern, relatively little is known about the nature of the disease itself. We have studied the early stages of pathogenesis in goats infected with six different isolates of Peste des petits ruminants virus representing all four known lineages of the virus. No lineage-specific difference in the pathogenicity of the virus isolates was observed, although there was evidence that even small numbers of cell culture passages could affect the degree of pathogenicity of an isolate. A consistent reduction in CD4+ T cells was observed at 4 days post infection (dpi). Measurement of the expression of various cytokines showed elements of a classic inflammatory response but also a relatively early induction of interleukin 10, which may be contributing to the observed disease.
Collapse
|
11
|
Bielinska AU, Makidon PE, Janczak KW, Blanco LP, Swanson B, Smith DM, Pham T, Szabo Z, Kukowska-Latallo JF, Baker JR. Distinct pathways of humoral and cellular immunity induced with the mucosal administration of a nanoemulsion adjuvant. THE JOURNAL OF IMMUNOLOGY 2014; 192:2722-33. [PMID: 24532579 DOI: 10.4049/jimmunol.1301424] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nasal administration of an oil-in-water nanoemulsion (NE) adjuvant W805EC produces potent systemic and mucosal, Th-1- and Th-17-balanced cellular responses. However, its molecular mechanism of action has not been fully characterized and is of particular interest because NE does not contain specific ligands for innate immune receptors. In these studies, we demonstrate that W805EC NE adjuvant activates innate immunity, induces specific gene transcription, and modulates NF-κB activity via TLR2 and TLR4 by a mechanism that appears to be distinct from typical TLR agonists. Nasal immunization with NE-based vaccine showed that the TLR2, TLR4, and MyD88 pathways and IL-12 and IL-12Rβ1 expression are not required for an Ab response, but they are essential for the induction of balanced Th-1 polarization and Th-17 cellular immunity. NE adjuvant induces MHC class II, CD80, and CD86 costimulatory molecule expression and dendritic cell maturation. Further, upon immunization with NE, adjuvant mice deficient in the CD86 receptor had normal Ab responses but significantly reduced Th-1 cellular responses, whereas animals deficient in both CD80 and CD86 or lacking CD40 failed to produce either humoral or cellular immunity. Overall, our data show that intranasal administration of Ag with NE induces TLR2 and TLR4 activation along with a MyD88-independent Ab response and a MyD88-dependent Th-1 and Th-17 cell-mediated immune response. These findings suggest that the unique properties of NE adjuvant may offer novel opportunities for understanding previously unrecognized mechanisms of immune activation important for generating effective mucosal and systemic immune responses.
Collapse
Affiliation(s)
- Anna U Bielinska
- Division of Allergy, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Crane MJ, Gaddi PJ, Salazar-Mather TP. UNC93B1 mediates innate inflammation and antiviral defense in the liver during acute murine cytomegalovirus infection. PLoS One 2012; 7:e39161. [PMID: 22723955 PMCID: PMC3377622 DOI: 10.1371/journal.pone.0039161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 05/21/2012] [Indexed: 12/14/2022] Open
Abstract
Antiviral defense in the liver during acute infection with the hepatotropic virus murine cytomegalovirus (MCMV) involves complex cytokine and cellular interactions. However, the mechanism of viral sensing in the liver that promotes these cytokine and cellular responses has remained unclear. Studies here were undertaken to investigate the role of nucleic acid-sensing Toll-like receptors (TLRs) in initiating antiviral immunity in the liver during infection with MCMV. We examined the host response of UNC93B1 mutant mice, which do not signal properly through TLR3, TLR7 and TLR9, to acute MCMV infection to determine whether liver antiviral defense depends on signaling through these molecules. Infection of UNC93B1 mutant mice revealed reduced production of systemic and liver proinflammatory cytokines including IFN-α, IFN-γ, IL-12 and TNF-α when compared to wild-type. UNC93B1 deficiency also contributed to a transient hepatitis later in acute infection, evidenced by augmented liver pathology and elevated systemic alanine aminotransferase levels. Moreover, viral clearance was impaired in UNC93B1 mutant mice, despite intact virus-specific CD8+ T cell responses in the liver. Altogether, these results suggest a combined role for nucleic acid-sensing TLRs in promoting early liver antiviral defense during MCMV infection.
Collapse
Affiliation(s)
- Meredith J. Crane
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| | - Pamela J. Gaddi
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| | - Thais P. Salazar-Mather
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
13
|
Inflammation induced by phytomodulatory proteins from the latex of Calotropis procera (Asclepiadaceae) protects against Salmonella infection in a murine model of typhoid fever. Inflamm Res 2012; 61:689-98. [DOI: 10.1007/s00011-012-0460-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 02/26/2012] [Accepted: 03/04/2012] [Indexed: 10/28/2022] Open
|
14
|
Flores-Langarica A, Marshall JL, Bobat S, Mohr E, Hitchcock J, Ross EA, Coughlan RE, Khan M, Van Rooijen N, Henderson IR, Maclennan ICM, Cunningham AF. T-zone localized monocyte-derived dendritic cells promote Th1 priming to Salmonella. Eur J Immunol 2011; 41:2654-65. [PMID: 21630252 DOI: 10.1002/eji.201141440] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 05/04/2011] [Accepted: 05/25/2011] [Indexed: 12/24/2022]
Abstract
Control of intracellular Salmonella infection requires Th1 priming and IFN-γ production. Here, we show that efficient Th1 priming after Salmonella infection requires CD11c(+) CD11b(hi) F4/80(+) monocyte-derived dendritic cells (moDCs). In non-infected spleens, moDCs are absent from T-cell zones (T zones) of secondary lymphoid tissues, but by 24 h post-infection moDCs are readily discernible in these sites. The accumulation of moDCs is more dependent upon bacterial viability than bacterial virulence. Kinetic studies showed that moDCs were necessary to prime but not sustain Th1 responses, while ex vivo studies showed that antigen-experienced moDCs were sufficient to induce T-cell proliferation and IFN-γ production via a TNF-α-dependent mechanism. Importantly, moDCs and cDCs when co-cultured induced superior Th1 differentiation than either subset alone, and this activity was independent of TNF-α. Thus, optimal Th1 development to Salmonella requires the rapid accumulation of moDCs within T zones and their collaboration with cDCs.
Collapse
Affiliation(s)
- Adriana Flores-Langarica
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yoon WS, Choi HJ, Park YK. Salmonella typhimurium harboring plasmid expressing interleukin-12 induced attenuation of infection and protective immune responses. J GEN APPL MICROBIOL 2011; 57:115-22. [PMID: 21606612 DOI: 10.2323/jgam.57.115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
IL-12 is known to be an essential cytokine which appears to provide protective immunity against intracellular bacteria, such as Salmonella. In this study, we investigated the possibility of developing a vaccine using IL-12 against virulent Salmonella. We used the host defense system activated by cytokine IL-12. The highly virulent Salmonella strain (Salmonella typhimurium UK-1) was transformed with cytokine-expressing plasmids. These live, wild-type pathogens were used as vaccine strains without undergoing any other biological or genetic attenuating processes. The newly developed strains induced partial protection from infections (30-40%). Of note, the interleukin-12-transformed pathogen was safe upon immunization with low doses (10(3) cfu), induced IgG responses, and stimulated protective immune responses against Salmonella typhimurium in mice (80-100%). These results suggest that IL-12 induced attenuation of wild-type Salmonella in the host infection stage and vaccine development using the wild-type strain harboring plasmid-secreting IL-12 may be considered as an alternative process for intracellular bacterial vaccine development without the inconvenience of time-consuming attenuation processes.
Collapse
Affiliation(s)
- Won Suck Yoon
- School of Life Sciences and Biotechnology, Korea University, Seoul
| | | | | |
Collapse
|
16
|
Early interferon-γ production in human lymphocyte subsets in response to nontyphoidal Salmonella demonstrates inherent capacity in innate cells. PLoS One 2010; 5:e13667. [PMID: 21048923 PMCID: PMC2965112 DOI: 10.1371/journal.pone.0013667] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 08/16/2010] [Indexed: 12/05/2022] Open
Abstract
Background Nontyphoidal Salmonellae frequently cause life-threatening bacteremia in sub-Saharan Africa. Young children and HIV-infected adults are particularly susceptible. High case-fatality rates and increasing antibiotic resistance require new approaches to the management of this disease. Impaired cellular immunity caused by defects in the T helper 1 pathway lead to intracellular disease with Salmonella that can be countered by IFNγ administration. This report identifies the lymphocyte subsets that produce IFNγ early in Salmonella infection. Methodology Intracellular cytokine staining was used to identify IFNγ production in blood lymphocyte subsets of ten healthy adults with antibodies to Salmonella (as evidence of immunity to Salmonella), in response to stimulation with live and heat-killed preparations of the D23580 invasive African isolate of Salmonella Typhimurium. The absolute number of IFNγ-producing cells in innate, innate-like and adaptive lymphocyte subpopulations was determined. Principal Findings Early IFNγ production was found in the innate/innate-like lymphocyte subsets: γδ-T cells, NK cells and NK-like T cells. Significantly higher percentages of such cells produced IFNγ compared to adaptive αβ-T cells (Student's t test, P<0.001 and ≤0.02 for each innate subset compared, respectively, with CD4+- and CD8+-T cells). The absolute numbers of IFNγ-producing cells showed similar differences. The proportion of IFNγ-producing γδ-T cells, but not other lymphocytes, was significantly higher when stimulated with live compared with heat-killed bacteria (P<0.0001). Conclusion/Significance Our findings indicate an inherent capacity of innate/innate-like lymphocyte subsets to produce IFNγ early in the response to Salmonella infection. This may serve to control intracellular infection and reduce the threat of extracellular spread of disease with bacteremia which becomes life-threatening in the absence of protective antibody. These innate cells may also help mitigate against the effect on IFNγ production of depletion of Salmonella-specific CD4+-T lymphocytes in HIV infection.
Collapse
|
17
|
Lima-Filho JV, Patriota JM, Silva AFB, Filho NT, Oliveira RSB, Alencar NMN, Ramos MV. Proteins from latex of Calotropis procera prevent septic shock due to lethal infection by Salmonella enterica serovar Typhimurium. JOURNAL OF ETHNOPHARMACOLOGY 2010; 129:327-334. [PMID: 20371281 DOI: 10.1016/j.jep.2010.03.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/11/2010] [Accepted: 03/27/2010] [Indexed: 05/29/2023]
Abstract
AIM OF THE STUDY The latex of Calotropis procera has been used in traditional medicine to treat different inflammatory diseases. The anti-inflammatory activity of latex proteins (LP) has been well documented using different inflammatory models. In this work the anti-inflammatory protein fraction was evaluated in a true inflammatory process by inducing a lethal experimental infection in the murine model caused by Salmonella enterica Subsp. enterica serovar Typhimurium. MATERIALS AND METHODS Experimental Swiss mice were given 0.2 ml of LP (30 or 60 mg/kg) by the intraperitoneal route 24 h before or after lethal challenge (0.2 ml) containing 10(6) CFU/ml of Salmonella Typhimurium using the same route of administration. RESULTS All the control animals succumbed to infection within 6 days. When given before bacterial inoculums LP prevented the death of mice, which remained in observation until day 28. Even, LP-treated animals exhibited only discrete signs of infection which disappeared latter. LP fraction was also protective when given orally or by subcutaneous route. Histopathological examination revealed that necrosis and inflammatory infiltrates were similar in both the experimental and control groups on days 1 and 5 after infection. LP activity did not clear Salmonella Typhimurium, which was still present in the spleen at approximately 10(4) cells/g of organ 28 days after challenge. However, no bacteria were detected in the liver at this stage. LP did not inhibit bacterial growth in culture medium at all. In the early stages of infection bacteria population was similar in organs and in the peritoneal fluid but drastically reduced in blood. Titration of TNF-alpha in serum revealed no differences between experimental and control groups on days 1 and 5 days after infection while IL-12 was only discretely diminished in serum of experimental animals on day 5. Moreover, cultured macrophages treated with LP and stimulated by LPS released significantly less IL-1beta. CONCLUSIONS LP-treated mice did not succumb to septic shock when submitted to a lethal infection. LP did not exhibit in vitro bactericidal activity. It is thought that protection of LP-treated mice against Salmonella Typhimurium possibly involves down-regulation of pro-inflammatory cytokines (other than TNF-alpha). LP inhibited IL-1beta release in cultured macrophages and discretely reduced IL-12 in serum of animals given LP. Results reported here support the folk use of latex to treat skin infections by topic application.
Collapse
Affiliation(s)
- José V Lima-Filho
- Departamento de Biologia, Universidade Federal Rural de Pernambuco, Campus Dois Irmãos, Recife, PE, Brazil.
| | | | | | | | | | | | | |
Collapse
|
18
|
John B, Harris TH, Tait ED, Wilson EH, Gregg B, Ng LG, Mrass P, Roos DS, Dzierszinski F, Weninger W, Hunter CA. Dynamic Imaging of CD8(+) T cells and dendritic cells during infection with Toxoplasma gondii. PLoS Pathog 2009; 5:e1000505. [PMID: 19578440 PMCID: PMC2700268 DOI: 10.1371/journal.ppat.1000505] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 06/05/2009] [Indexed: 12/21/2022] Open
Abstract
To better understand the initiation of CD8+ T cell responses during infection, the primary response to the intracellular parasite Toxoplasma gondii was characterized using 2-photon microscopy combined with an experimental system that allowed visualization of dendritic cells (DCs) and parasite specific CD8+ T cells. Infection with T. gondii induced localization of both these populations to the sub-capsular/interfollicular region of the draining lymph node and DCs were required for the expansion of the T cells. Consistent with current models, in the presence of cognate antigen, the average velocity of CD8+ T cells decreased. Unexpectedly, infection also resulted in modulation of the behavior of non-parasite specific T cells. This TCR-independent process correlated with the re-modeling of the lymph node micro-architecture and changes in expression of CCL21 and CCL3. Infection also resulted in sustained interactions between the DCs and CD8+ T cells that were visualized only in the presence of cognate antigen and were limited to an early phase in the response. Infected DCs were rare within the lymph node during this time frame; however, DCs presenting the cognate antigen were detected. Together, these data provide novel insights into the earliest interaction between DCs and CD8+ T cells and suggest that cross presentation by bystander DCs rather than infected DCs is an important route of antigen presentation during toxoplasmosis. Toxoplasma gondii is a protozoan parasite that can infect a wide range of hosts, including humans. Infection with T. gondii is potentially life threatening in immuno-compromised individuals and it can be detrimental during pregnancy, often leading to abortion of the fetus. Dendritic cells are thought to play a vital role in the development of protective immunity to Toxoplasma gondii through their ability to produce immunological signals such as cytokines and also process and present parasite derived peptides to T cells. However, little is known about the actual interactions between these cell types in an intact organ, such as the lymph node, during infection. Using the technology of live imaging by 2-photon microscopy we have identified a very early window of time during infection when dendritic cells and T cells make sustained contacts with one another, which appears crucial for the generation of protective responses. We also show that substantial changes are induced in the lymph node micro-architecture as a result of infection, which in turn could have effects on immune responses to secondary pathogens. Understanding the interaction between these immune cells in vivo that leads to resistance to active infection would help in the design of better strategies to develop protective immune responses against this pathogen in immuno-compromised individuals.
Collapse
Affiliation(s)
- Beena John
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tajie H. Harris
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elia D. Tait
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Emma H. Wilson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Division of Biomedical Sciences, University of California, Riverside, California, United States of America
| | - Beth Gregg
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lai Guan Ng
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Paulus Mrass
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - David S. Roos
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Wolfgang Weninger
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
19
|
Le Negrate G, Faustin B, Welsh K, Loeffler M, Krajewska M, Hasegawa P, Mukherjee S, Orth K, Krajewski S, Godzik A, Guiney DG, Reed JC. Salmonella Secreted Factor L Deubiquitinase of Salmonella typhimurium Inhibits NF-κB, Suppresses IκBα Ubiquitination and Modulates Innate Immune Responses. THE JOURNAL OF IMMUNOLOGY 2008; 180:5045-56. [DOI: 10.4049/jimmunol.180.7.5045] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Abstract
Interferon-gamma (IFN-gamma) is crucial for immunity against intracellular pathogens and for tumor control. However, aberrant IFN-gamma expression has been associated with a number of autoinflammatory and autoimmune diseases. This cytokine is produced predominantly by natural killer (NK) and natural killer T (NKT) cells as part of the innate immune response, and by Th1 CD4 and CD8 cytotoxic T lymphocyte (CTL) effector T cells once antigen-specific immunity develops. Herein, we briefly review the functions of IFN-gamma, the cells that produce it, the cell extrinsic signals that induce its production and influence the differentiation of naïve T cells into IFN-gamma-producing effector T cells, and the signaling pathways and transcription factors that facilitate, induce, or repress production of this cytokine. We then review and discuss recent insights regarding the molecular regulation of IFN-gamma, focusing on work that has led to the identification and characterization of distal regulatory elements and epigenetic modifications with the IFN-gamma locus (Ifng) that govern its expression. The epigenetic modifications and three-dimensional structure of the Ifng locus in naive CD4 T cells, and the modifications they undergo as these cells differentiate into effector T cells, suggest a model whereby the chromatin architecture of Ifng is poised to facilitate either rapid opening or silencing during Th1 or Th2 differentiation, respectively.
Collapse
|
21
|
Haghighi HR, Abdul-Careem MF, Dara RA, Chambers JR, Sharif S. Cytokine gene expression in chicken cecal tonsils following treatment with probiotics and Salmonella infection. Vet Microbiol 2008; 126:225-33. [PMID: 17681719 DOI: 10.1016/j.vetmic.2007.06.026] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Revised: 06/26/2007] [Accepted: 06/27/2007] [Indexed: 12/31/2022]
Abstract
Probiotics are currently employed for control of pathogens and enhancement of immune response in chickens. In this study, we investigated the underlying immunological mechanisms of the action of probiotics against colonization of the chicken intestine by Salmonella enterica subsp. enterica serovar Typhimurium (Salmonella serovar Typhimurium). Birds received probiotics by oral gavage on day 1 of age and, subsequently, received Salmonella serovar Typhimurium on day 2 of age. Cecal tonsils were removed on days 1, 3 and 5 post-infection (p.i.), RNA was extracted and subjected to real-time quantitative RT-PCR for measurement of interleukin (IL)-6, IL-10, IL-12 and interferon (IFN)-gamma gene expression. There was no significant difference in IL-6 and IL-10 gene expression in cecal tonsils of chickens belonging to various treatment groups. Salmonella serovar Typhimurium infection resulted in a significant increase in IL-12 expression in cecal tonsils on days 1 and 5p.i. However, when chickens were treated with probiotics prior to experimental infection with Salmonella, the level of IL-12 expression was similar to that observed in uninfected control chickens. Treatment of birds with probiotics resulted in a significant decrease in IFN-gamma gene expression in cecal tonsils of chickens infected with Salmonella compared to the Salmonella-infected birds not treated with probiotics. These findings reveal that repression of IL-12 and IFN-gamma expression is associated with probiotic-mediated reduction in intestinal colonization with Salmonella serovar Typhimurium.
Collapse
Affiliation(s)
- Hamid R Haghighi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | | | | | | | |
Collapse
|
22
|
Price JD, Simpfendorfer KR, Mantena RR, Holden J, Heath WR, van Rooijen N, Strugnell RA, Wijburg OLC. Gamma interferon-independent effects of interleukin-12 on immunity to Salmonella enterica serovar Typhimurium. Infect Immun 2007; 75:5753-62. [PMID: 17875635 PMCID: PMC2168367 DOI: 10.1128/iai.00971-07] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 08/20/2007] [Accepted: 09/02/2007] [Indexed: 11/20/2022] Open
Abstract
Interleukin-12 (IL-12) and IL-18 are both central to the induction of gamma interferon (IFN-gamma), and various roles for IL-12 and IL-18 in control of intracellular microbial infections have been demonstrated. We used IL-12p40(-/-) and IL-18(-/-) mice to further investigate the role of IL-12 and IL-18 in control of Salmonella enterica serovar Typhimurium. While C57BL/6 and IL-18(-/-) mice were able to resolve attenuated S. enterica serovar Typhimurium infections, the IL-12p40(-/-) mice succumbed to a high bacterial burden after 60 days. Using ovalbumin (OVA)-specific T-cell receptor transgenic T cells (OT-II cells), we demonstrated that following oral infection with recombinant S. enterica serovar Typhimurium expressing OVA, the OT-II cells proliferated in the mesenteric lymph nodes of C57BL/6 and IL-18(-/-) mice but not in IL-12p40(-/-) mice. In addition, we demonstrated by flow cytometry that equivalent or increased numbers of T cells produced IFN-gamma in IL-12p40(-/-) mice compared with the numbers of T cells that produced IFN-gamma in C57BL/6 and IL-18(-/-) mice. Finally, we demonstrated that removal of macrophages from S. enterica serovar Typhimurium-infected C57BL/6 and IL-12p40(-/-) mice did not affect the bacterial load, suggesting that impaired control of S. enterica serovar Typhimurium infection in the absence of IL-12p40 is not due to reduced macrophage bactericidal activities, while IL-18(-/-) mice did rely on the presence of macrophages for control of the infection. Our results suggest that IL-12p40, but not IL-18, is critical to resolution of infections with attenuated S. enterica serovar Typhimurium and that especially the effects of IL-12p40 on proliferative responses of CD4+ T cells, but not the ability of these cells to produce IFN-gamma, are important in the resolution of infection by this intracellular bacterial pathogen.
Collapse
Affiliation(s)
- Jason D Price
- CRC for Vaccine Technology and Department of Microbiology & Immunology, The University of Melbourne, Parkville VIC3010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Rodenburg W, Keijer J, Kramer E, Roosing S, Vink C, Katan MB, van der Meer R, Bovee-Oudenhoven IMJ. Salmonella induces prominent gene expression in the rat colon. BMC Microbiol 2007; 7:84. [PMID: 17850650 PMCID: PMC2048963 DOI: 10.1186/1471-2180-7-84] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Accepted: 09/12/2007] [Indexed: 11/25/2022] Open
Abstract
Background Salmonella enteritidis is suggested to translocate in the small intestine. In vivo it induces gene expression changes in the ileal mucosa and Peyer's patches. Stimulation of Salmonella translocation by dietary prebiotics fermented in colon suggests involvement of the colon as well. However, effects of Salmonella on colonic gene expression in vivo are largely unknown. We aimed to characterize time dependent Salmonella-induced changes of colonic mucosal gene expression in rats using whole genome microarrays. For this, rats were orally infected with Salmonella enteritidis to mimic a foodborne infection and colonic gene expression was determined at days 1, 3 and 6 post-infection (n = 8 rats per time-point). As fructo-oligosaccharides (FOS) affect colonic physiology, we analyzed colonic mucosal gene expression of FOS-fed versus cellulose-fed rats infected with Salmonella in a separate experiment. Colonic mucosal samples were isolated at day 2 post-infection. Results Salmonella affected transport (e.g. Chloride channel calcium activated 6, H+/K+ transporting Atp-ase), antimicrobial defense (e.g. Lipopolysaccharide binding protein, Defensin 5 and phospholipase A2), inflammation (e.g. calprotectin), oxidative stress related genes (e.g. Dual oxidase 2 and Glutathione peroxidase 2) and Proteolysis (e.g. Ubiquitin D and Proteosome subunit beta type 9). Furthermore, Salmonella translocation increased serum IFNγ and many interferon-related genes in colonic mucosa. The gene most strongly induced by Salmonella infection was Pancreatitis Associated Protein (Pap), showing >100-fold induction at day 6 after oral infection. Results were confirmed by Q-PCR in individual rats. Stimulation of Salmonella translocation by dietary FOS was accompanied by enhancement of the Salmonella-induced mucosal processes, not by induction of other processes. Conclusion We conclude that the colon is a target tissue for Salmonella, considering the abundant changes in mucosal gene expression.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Gene Expression
- Intestine, Small/metabolism
- Intestine, Small/microbiology
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Oligonucleotide Array Sequence Analysis
- Pancreatitis-Associated Proteins
- Rats
- Salmonella Infections, Animal/microbiology
- Salmonella enteritidis/chemistry
- Salmonella enteritidis/genetics
- Salmonella enteritidis/immunology
- Salmonella enteritidis/physiology
Collapse
Affiliation(s)
- Wendy Rodenburg
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
- RIKILT Institute of Food Safety, PO Box 230, 6700 AE, Wageningen, The Netherlands
- NIZO Food Research, PO Box 20, 6710 BA, Ede, The Netherlands
| | - Jaap Keijer
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
- RIKILT Institute of Food Safety, PO Box 230, 6700 AE, Wageningen, The Netherlands
| | - Evelien Kramer
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
- RIKILT Institute of Food Safety, PO Box 230, 6700 AE, Wageningen, The Netherlands
| | - Susanne Roosing
- RIKILT Institute of Food Safety, PO Box 230, 6700 AE, Wageningen, The Netherlands
| | - Carolien Vink
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
- NIZO Food Research, PO Box 20, 6710 BA, Ede, The Netherlands
| | - Martijn B Katan
- Vrije Universiteit, Institute of Health Sciences, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Roelof van der Meer
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
- NIZO Food Research, PO Box 20, 6710 BA, Ede, The Netherlands
- Nutrigenomics Consortium, TIFN, PO Box 557, 6700 AN, Wageningen, The Netherlands
| | - Ingeborg MJ Bovee-Oudenhoven
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
- NIZO Food Research, PO Box 20, 6710 BA, Ede, The Netherlands
| |
Collapse
|
24
|
Wick MJ. Monocyte and dendritic cell recruitment and activation during oral Salmonella infection. Immunol Lett 2007; 112:68-74. [PMID: 17720254 DOI: 10.1016/j.imlet.2007.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Revised: 07/13/2007] [Accepted: 07/15/2007] [Indexed: 12/24/2022]
Abstract
Immunity to bacterial infection involves the joint effort of the innate and adaptive immune systems. The innate immune response is triggered when the body senses bacterial components, such as lipopolysaccharide, that alarm the body of the invader. An array of cell types function in the innate response. These cells are rapidly recruited to the infection site and activated to optimally perform their functions. The adaptive immune response follows the innate response, and one cell type in particular, dendritic cells (DCs), are the critical link between the innate and adaptive responses. This review will summarize recent data concerning the events that occur early during oral infection with the intracellular pathogen Salmonella, with emphasis on the phagocytic cells involved in combating the infection in the gut-associated lymphoid tissues. In particular, recent findings concerning the recruitment and activation of mononuclear phagocyte populations and dendritic cell subsets will be presented after an overview of the Salmonella infection model.
Collapse
Affiliation(s)
- Mary Jo Wick
- Department of Microbiology and Immunology, Göteborg University, Box 435, S-405 30 Göteborg, Sweden.
| |
Collapse
|
25
|
Wang Y, Qu L, Uthe JJ, Bearson SMD, Kuhar D, Lunney JK, Couture OP, Nettleton D, Dekkers JCM, Tuggle CK. Global transcriptional response of porcine mesenteric lymph nodes to Salmonella enterica serovar Typhimurium. Genomics 2007; 90:72-84. [PMID: 17499962 DOI: 10.1016/j.ygeno.2007.03.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 03/20/2007] [Accepted: 03/23/2007] [Indexed: 10/23/2022]
Abstract
To elucidate the host transcriptional response to Salmonella enterica serovar Typhimurium, Affymetrix porcine GeneChip analysis of pig mesenteric lymph nodes was used to identify 848 genes showing differential expression across different times after inoculation or when compared to non-inoculated controls. Annotation analyses showed that a high proportion of these differentially expressed (DE) genes are involved in immune and inflammatory responses. T helper 1, innate/inflammatory, and antigen-processing pathways were induced at 24 h post-inoculation (hpi) and/or 48 hpi, while apoptosis and antigen presentation/dendritic cell function pathways were downregulated at 8 hpi. Cluster analyses revealed that most DE genes annotated as NFkappaB targets were grouped into a specific induced subcluster, while many translation-related DE genes were found in a repressed subcluster. Quantitative polymerase chain reaction analyses confirmed the Affymetrix results, revealing transcriptional induction of NFkappaB target genes at 24 hpi and suppression of the NFkappaB pathway from 24 to 48 hpi. We propose that such NFkappaB suppression in antigen-presenting cells may be the mechanism by which S. Typhimurium eludes a strong inflammatory response to establish a carrier status in pigs.
Collapse
Affiliation(s)
- Yanfang Wang
- Department of Animal Science and Center for Integrated Animal Genomics Iowa State University, Ames, IA 50010, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Salem ML, Gillanders WE, Kadima AN, El-Naggar S, Rubinstein MP, Demcheva M, Vournakis JN, Cole DJ. Review: novel nonviral delivery approaches for interleukin-12 protein and gene systems: curbing toxicity and enhancing adjuvant activity. J Interferon Cytokine Res 2006; 26:593-608. [PMID: 16978064 DOI: 10.1089/jir.2006.26.593] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
It has become increasingly apparent that the ability to generate an optimal host immune response requires effective cross talk between the innate and adaptive components of the immune system. Pro-inflammatory cytokines, in particular those that can induce a danger signal, often called signal 3, are crucial in this role of initiating and augmenting the presentation of exogenous antigen to T cells by dendritic cells. Interleukin-12 (IL-12) in particular has been defined as a "signal 3" cytokine required for the antigen cross priming. Given this unique interactive function, a significant amount of work has been performed to define possible therapeutic applications for IL-12. Systemic IL-12 administration can clearly act as a potent adjuvant for postvaccination T cell responses in a variety of diseases. As an example, in the cancer setting, systemic IL-12 is capable of suppressing tumor growth, metastasis, and angiogenesis in vivo. IL-12, however, has been associated with significant dose- and schedule-dependent toxicity in early clinical trials, results that have proven to be a major obstacle to its clinical application. Recent research has focused on decreasing the toxicity of IL-12 using different delivery approaches, including virus-based and gene-modified cell-based delivery. Although effective, these approaches also have limitations, including the generation of neutralizing antibodies, in addition to lacking the simplicity and versatility required for universal clinical application. Thus, there is a significant interest in the development of alternative delivery approaches for IL-12 administration that can overcome these issues. Several nonviral delivery approaches for IL-12 protein or gene expression vectors are being defined, including alum, liposomes, and polymer-based delivery. These developing approaches have shown promising adjuvant effects with significantly lessened systemic toxicity. This article discusses the potential capabilities of these nonvirus-based IL-12 delivery systems in different disease settings, including allergy, infection, and cancer.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Department of Surgery, Section of Surgical Oncology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Roy MF, Larivière L, Wilkinson R, Tam M, Stevenson MM, Malo D. Incremental expression of Tlr4 correlates with mouse resistance to Salmonella infection and fine regulation of relevant immune genes. Genes Immun 2006; 7:372-83. [PMID: 16738669 DOI: 10.1038/sj.gene.6364309] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The mouse response to Salmonella Typhimurium infection is partly controlled through detection of the bacterium lipopolysaccharide by the host pattern recognition receptor, Toll-like receptor 4 (Tlr4). Mice deficient in Tlr4 signaling are extremely susceptible to Salmonella infection with a 1,000-fold reduction in LD(50). In a previous study, we showed, using transgenic mice carrying one, three, six and >30 copies of Tlr4, that the level of expression of this gene influences the outcome of Salmonella infection, with a plateau effect starting at three copies. In the present study, we further investigate the impact of Tlr4 during Salmonella infection in mice expressing Tlr4 at slightly sub-normal, normal and slightly supra-normal levels by comparing host responses in mice carrying one, two and three copies of Tlr4 on the same genetic background. We describe in detail the in vivo host response to pathogenic Salmonella and show for the first time, in this narrow range of Tlr4 expression, an incremental protective effect against Salmonella due to improved control of bacterial growth in target organs and increased expression of important immune response genes in the spleen.
Collapse
Affiliation(s)
- M-F Roy
- Department of Human Genetics, McGill University Health Center, Montréal, Québec, Canada
| | | | | | | | | | | |
Collapse
|
28
|
Klagge IM, Abt M, Fries B, Schneider-Schaulies S. Impact of measles virus dendritic-cell infection on Th-cell polarization in vitro. J Gen Virol 2004; 85:3239-3247. [PMID: 15483237 DOI: 10.1099/vir.0.80125-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Interference of measles virus (MV) with dendritic-cell (DC) functions and deregulation of T-cell differentiation have been proposed to be central to the profound suppression of immune responses to secondary infections up to several weeks after the acute disease. To address the impact of MV infection on the ability of DCs to promote Th-cell differentiation, an in vitro system was used where uninfected, tumour necrosis factor alpha/interleukin (IL) 1 beta-primed DCs were co-cultured with CD45RO(-) T cells in the presence of conditioned media from MV-infected DCs primed under neutral or DC-polarizing conditions. It was found that supernatants of DCs infected with an MV vaccine strain strongly promoted Th1 differentation, whereas those obtained from wild-type MV-infected DCs generated a mixed Th1/Th0 response, irrespective of the conditions used for DC priming. Th-cell commitment in this system did not correlate with the production of IL12 p70, IL18 or IL23. Thus, a combination of these or other, as yet undefined, soluble factors is produced upon MV infection of DCs that strongly promotes Th1/Th0 differentiation.
Collapse
Affiliation(s)
- Ingo M Klagge
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| | - Marion Abt
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| | - Bianca Fries
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| | - Sibylle Schneider-Schaulies
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| |
Collapse
|
29
|
Cytokines in Salmonellosis. EcoSal Plus 2004; 1. [PMID: 26443358 DOI: 10.1128/ecosalplus.8.8.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The recruitment and activation of phagocytic cells in infected tissues and the induction of T-cell- and B-cell-dependent acquired immunity are crucial for the control and resolution of Salmonella infections. These complex processes require the interaction of bacteria with a multitude of cell surface receptors and the controlled production of soluble mediators. The mechanisms of cytokine induction in response to Salmonella and the role of cytokine networks in Salmonella infections are the main foci of this review. Pathogen-associated molecular pattern receptors play an important role in recognition of bacteria by the host. Effective immunity against the bacterium therefore relies on the ability of the host to recruit phagocytes in the tissues and to enhance the antibacterial functions of these inflammatory cells. TNF-a, IFN-?, IL12, IL15, and IL18 are needed for the full expression of innate host resistance to Salmonella. The genes for mammalian cytokines can be cloned into suitable vectors and expressed in Salmonella as functional proteins. The in vivo production of cytokines by Salmonella carriers can have therapeutic applications and can modulate immune functions in the host. The possibility to modulate antigen-specific immune responses by expressing cytokines in Salmonella is illustrated by the increase in Salmonella-specific IgA responses induced by administration of IL-5-expressing bacteria. The same cytokines that are responsible for endotoxic shock are elevated in the late stages of lethal Salmonella infections, indicating that the toxicity of Salmonella lipopolysaccharide (LPS) may actually be contributing to the death of the host.
Collapse
|
30
|
Babu U, Dalloul RA, Okamura M, Lillehoj HS, Xie H, Raybourne RB, Gaines D, Heckert RA. Salmonella enteritidis clearance and immune responses in chickens following Salmonella vaccination and challenge. Vet Immunol Immunopathol 2004; 101:251-7. [PMID: 15350755 DOI: 10.1016/j.vetimm.2004.05.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2003] [Revised: 05/06/2004] [Accepted: 05/20/2004] [Indexed: 11/27/2022]
Abstract
Our previous work showed that the cell-mediated immunity (CMI) was enhanced by live Salmonella vaccine (LV). The objective of this study was to evaluate the impact of live and killed Salmonella vaccines on Salmonella enteritidis (SE) clearance and to determine if the clearance was mediated by cell-mediated and/or humoral immunity. Chickens were first immunized at 2 weeks of age followed by a booster dose at 4 weeks, challenged with live SE 2 weeks later (6-week-old) and tested for CMI, antibody response and SE clearance 1-week post SE-challenge (7-week-old). Spleen cell proliferation induced by SE-flagella and Concanavalin A (Con A) were significantly higher and SE shedding was significantly lower in the LV group. The splenic CD3 population was significantly lower and B cells were higher in the control group compared to all the SE-challenged groups (with and without vaccination). Serum antibody to SE-flagella and envelope were significantly higher in the KV group compared to all the other groups. These results suggest that LV protects against SE infection, probably by enhancing the CMI.
Collapse
Affiliation(s)
- U Babu
- Center for Food Safety and Applied Nutrition, US Food and Drug Administration, HFS-326, 8301 Muirkirk Road, Laurel, MD 20708, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Norimatsu M, Chance V, Dougan G, Howard CJ, Villarreal-Ramos B. Live Salmonella enterica serovar Typhimurium (S. Typhimurium) elicit dendritic cell responses that differ from those induced by killed S. Typhimurium. Vet Immunol Immunopathol 2004; 98:193-201. [PMID: 15010228 DOI: 10.1016/j.vetimm.2003.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Revised: 12/05/2003] [Accepted: 12/05/2003] [Indexed: 11/16/2022]
Abstract
The immune response of bovine monocytes-derived dendritic cells (DC) exposed to either live or killed Salmonella enterica serovar Typhimurium was compared. Both live and killed bacteria induced changes in morphology with distinctive formation of processes and up-regulation of the ability of DC to stimulate allogeneic T-cell proliferation. Also, both live and killed bacteria up-regulated the expression of MHC-I, MHC-II and CD80. However, live bacteria induced greater up-regulation of the expression of CD40 and CD86 than killed bacteria. Live bacteria also induced greater up-regulation of transcription for IL-6, IL-12 and GM-CSF than killed bacteria as measured by quantitative RT-PCR. These data suggest that blood-monocyte-derived DC may follow distinct maturation pathways following exposure to live or killed bacteria. These differences are likely to have consequences for the priming of the adaptive immune responses.
Collapse
Affiliation(s)
- Mari Norimatsu
- Institute for Animal Health, Compton, Newbury, Berkshire RG20 7NN, UK.
| | | | | | | | | |
Collapse
|