1
|
Chakraborty S, Dutta P, Pal A, Chakraborty S, Banik G, Halder P, Gope A, Miyoshi SI, Das S. Intranasal immunization of mice with chimera of Salmonella Typhi protein elicits protective intestinal immunity. NPJ Vaccines 2024; 9:24. [PMID: 38321067 PMCID: PMC10847434 DOI: 10.1038/s41541-024-00812-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/26/2024] [Indexed: 02/08/2024] Open
Abstract
Development of safe, highly effective and affordable enteric fever vaccines is a global health priority. Live, oral typhoid vaccines induce strong mucosal immunity and long-term protection, but safety remains a concern. In contrast, efficacy wears off rapidly for injectable, polysaccharide-based vaccines, which elicit poor mucosal response. We previously reported Salmonella Typhi outer membrane protein, T2544 as a potential candidate for bivalent (S. Typhi and S. Paratyphi A) vaccine development. Here, we show that intranasal immunization with a subunit vaccine (chimera of T2544 and cholera toxin B subunit) induced strong systemic and intestinal mucosal immunity and protection from S. Typhi challenge in a mouse model. CTB-T2544 augmented gut-homing receptor expression on lymphocytes that produced Th1 and Th17 cytokines, secretory IgA in stool that inhibited bacterial motility and epithelial attachment, antibody recall response and affinity maturation with increased number of follicular helper T cells and CD4+ central and effector memory cells.
Collapse
Affiliation(s)
- Suparna Chakraborty
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Pujarini Dutta
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
- Department of Pediatrics, Steele Children's Research Center, University of Arizona, Tuscon, AZ, USA
| | - Ananda Pal
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Swarnali Chakraborty
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - George Banik
- BD Biosciences, INDIA, Smart works Business Center, Victoria Park, 37/2 GN Block, Sector 5, Saltlake City, Kolkata, 700091, India
| | - Prolay Halder
- Division of Bacteriology, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Animesh Gope
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India
| | - Shin-Ichi Miyoshi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Collaborative Research Center of Okayama University for Infectious Diseases at Indian Council of Medical Research-National Institute of Cholera and Enteric Diseases, Kolkata, 700010, India
| | - Santasabuj Das
- Division of Clinical Medicine, ICMR- National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata, 700 010, India.
- ICMR-National Institute of Occupational Health, Meghaninagar, Ahmedabad, 3800016, Gujarat, India.
| |
Collapse
|
2
|
Pardy RD, Wallbank BA, Striepen B, Hunter CA. Immunity to Cryptosporidium: insights into principles of enteric responses to infection. Nat Rev Immunol 2024; 24:142-155. [PMID: 37697084 DOI: 10.1038/s41577-023-00932-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/13/2023]
Abstract
Cryptosporidium parasites replicate within intestinal epithelial cells and are an important cause of diarrhoeal disease in young children and in patients with primary and acquired defects in T cell function. This Review of immune-mediated control of Cryptosporidium highlights advances in understanding how intestinal epithelial cells detect this infection, the induction of innate resistance and the processes required for activation of T cell responses that promote parasite control. The development of a genetic tool set to modify Cryptosporidium combined with tractable mouse models provide new opportunities to understand the principles that govern the interface between intestinal epithelial cells and the immune system that mediate resistance to enteric pathogens.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Xu Y, Wang Y, Winner H, Yang H, He R, Wang J, Zhong G. Regulation of chlamydial spreading from the small intestine to the large intestine by IL-22-producing CD4 + T cells. Infect Immun 2024; 92:e0042123. [PMID: 38047677 PMCID: PMC10790816 DOI: 10.1128/iai.00421-23] [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: 10/13/2023] [Accepted: 11/14/2023] [Indexed: 12/05/2023] Open
Abstract
Following an oral inoculation, Chlamydia muridarum descends to the mouse large intestine for long-lasting colonization. However, a mutant C. muridarum that lacks the plasmid-encoded protein pGP3 due to an engineered premature stop codon (designated as CMpGP3S) failed to do so even following an intrajejunal inoculation. This was because a CD4+ T cell-dependent immunity prevented the spread of CMpGP3S from the small intestine to the large intestine. In the current study, we found that mice deficient in IL-22 (IL-22-/-) allowed CMpGP3S to spread from the small intestine to the large intestine on day 3 after intrajejunal inoculation, indicating a critical role of IL-22 in regulating the chlamydial spread. The responsible IL-22 is produced by CD4+ T cells since IL-22-/- mice were rescued to block the CMpGP3S spread by donor CD4+ T cells from C57BL/6J mice. Consistently, CD4+ T cells lacking IL-22 failed to block the spread of CMpGP3S in Rag2-/- mice, while IL-22-competent CD4+ T cells did block. Furthermore, mice deficient in cathelicidin-related antimicrobial peptide (CRAMP) permitted the CMpGP3S spread, but donor CD4+ T cells from CRAMP-/- mice were still sufficient for preventing the CMpGP3S spread in Rag2-/- mice, indicating a critical role of CRAMP in regulating chlamydial spreading, and the responsible CRAMP is not produced by CD4+ T cells. Thus, the IL-22-producing CD4+ T cell-dependent regulation of chlamydial spreading correlated with CRAMP produced by non-CD4+ T cells. These findings provide a platform for further characterizing the subset(s) of CD4+ T cells responsible for regulating bacterial spreading in the intestine.
Collapse
Affiliation(s)
- Ying Xu
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yihui Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Halah Winner
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Huijie Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Rongze He
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
Sreekantapuram S, Berens C, Barth SA, Methner U, Berndt A. Interaction of Salmonella Gallinarum and Salmonella Enteritidis with peripheral leucocytes of hens with different laying performance. Vet Res 2021; 52:123. [PMID: 34563266 PMCID: PMC8467188 DOI: 10.1186/s13567-021-00994-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/26/2021] [Indexed: 11/23/2022] Open
Abstract
Salmonella enterica ssp. enterica serovars Enteritidis (SE) and Gallinarum (SG) cause different diseases in chickens. However, both are able to reach the blood stream where heterophils and monocytes are potentially able to phagocytose and kill the pathogens. Using an ex vivo chicken whole blood infection model, we compared the complex interactions of the differentially host-adapted SE and SG with immune cells in blood samples of two White Leghorn chicken lines showing different laying performance (WLA: high producer; R11: low producer). In order to examine the dynamic interaction between peripheral blood leucocytes and the Salmonella serovars, we performed flow cytometric analyses and survival assays measuring (i) leucocyte numbers, (ii) pathogen association with immune cells, (iii) Salmonella viability and (iv) immune gene transcription in infected whole blood over a four-hour co-culture period. Inoculation of blood from the two chicken lines with Salmonella led primarily to an interaction of the bacteria with monocytes, followed by heterophils and thrombocytes. We found higher proportions of monocytes associated with SE than with SG. In blood samples of high producing chickens, a decrease in the numbers of both heterophils and Salmonella was observed. The Salmonella challenge induced transcription of interleukin-8 (IL-8) which was more pronounced in SG- than SE-inoculated blood of R11. In conclusion, the stronger interaction of monocytes with SE than SG and the better survivability of Salmonella in blood of low-producer chickens shows that the host-pathogen interaction and the strength of the immune defence depend on both the Salmonella serovar and the chicken line.
Collapse
Affiliation(s)
- Sravya Sreekantapuram
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Christian Berens
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, Jena, Germany
| | - Stefanie A Barth
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, Jena, Germany
| | - Ulrich Methner
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Jena, Germany
| | - Angela Berndt
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, Jena, Germany.
| |
Collapse
|
5
|
Roy U, de Oliveira RS, Galvez EJC, Gronow A, Basic M, Perez LG, Gagliani N, Bleich A, Huber S, Strowig T. Induction of IL-22-Producing CD4+ T Cells by Segmented Filamentous Bacteria Independent of Classical Th17 Cells. Front Immunol 2021; 12:671331. [PMID: 34566952 PMCID: PMC8456099 DOI: 10.3389/fimmu.2021.671331] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/11/2021] [Indexed: 01/13/2023] Open
Abstract
The intestinal microbiota modulates IL-22 production in the intestine, including the induction of IL-22-producing CD4+ T helper cells. Which specific bacteria are responsible for the induction of these cells is less well understood. Here, we demonstrate through the use of novel gnotobiotic knock-in reporter mice that segmented filamentous bacteria (SFB), which are known for their ability to induce Th17 cells, also induce distinct IL-17A negative CD4+ T cell populations in the intestine. A subset of these cells instead produces IL-22 upon restimulation ex vivo and also during enteric infections. Furthermore, they produce a distinct set of cytokines compared to Th17 cells including the differential expression of IL-17F and IFN-γ. Importantly, genetic models demonstrate that these cells, presumably Th22 cells, develop independently of intestinal Th17 cells. Together, our data identifies that besides Th17, SFB also induces CD4+ T cell populations, which serve as immediate source of IL-22 during intestinal inflammation.
Collapse
Affiliation(s)
- Urmi Roy
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Rômulo S. de Oliveira
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Eric J. C. Galvez
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Achim Gronow
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Laura Garcia Perez
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
6
|
Characteristics of Chlamydia suis Ocular Infection in Pigs. Pathogens 2021; 10:pathogens10091103. [PMID: 34578134 PMCID: PMC8470092 DOI: 10.3390/pathogens10091103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022] Open
Abstract
Chlamydia (C.) suis can often be isolated from conjunctival swab specimens from pigs with conjunctivitis or keratoconjunctivitis. In the field, it is assumed to be a multifactorial disease triggered by immunosuppressing factors. This is the first experimental study to provoke clinical signs of conjunctivitis in pigs after C. suis primary mono-infection. Five six-week-old male piglets, free of ocular chlamydia shedding and seronegative for Chlamydia, were conjunctivally infected with the C. suis-type strain S45 (1 × 109 inclusion forming units), while four piglets served as negative controls. The infection group developed clinical signs of conjunctivitis with a peak in the first week post-infection. Immunohistochemical evaluation revealed the presence of Chlamydia not only in the conjunctival epithelium, but also in the enlarged lacrimal glands, lungs, and intestine. No circulating antibodies could be detected during the whole study period of three weeks, although three different test systems were applied as follows: the complement fixation test, MOMP-based Chlamydiaceae ELISA, and PmpC-based C. suis ELISA. Meanwhile, high numbers of IFN-γ-producing lymphocytes within PBMC were seen after C. suis re-stimulation 14 days post-infection. Hence, these data suggest that entry via the eye may not elicit immunological responses comparable to other routes of chlamydial infections.
Collapse
|
7
|
Burlacu A, Crisan-Dabija R, Popa IV, Covic A. The Second Wave of COVID-19 Pandemic Strikes during the Flu Season: An Awareness Perspective. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:E707. [PMID: 33352889 PMCID: PMC7766640 DOI: 10.3390/medicina56120707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/02/2020] [Accepted: 12/16/2020] [Indexed: 11/16/2022]
Abstract
Coinfection with both SARS-CoV-2 and influenza viruses seems to be a real and severe problem. However, coinfection is far from a simple matter, and cannot be considered having more unfavorable outcomes as a direct consequence. In reality, the aftermath is powerfully nuanced by the presence of risk factors and specific molecular mechanisms. Our objective was to raise awareness around the unpredictable association between COVID-19 pandemics and the upcoming flu season, and make arguments about the need to develop new routine testing protocols for both viruses, at least during the period with an expected high incidence. Our reasoning is built around the various impacts that the whole range of risk groups, common immunological mechanisms, and complex interactions, such as influenza vaccination, will have on patients' prognosis. We show that the more flawed clinical course is due to managing only one of the infections (and, subsequently, neglecting the other condition).
Collapse
Affiliation(s)
- Alexandru Burlacu
- Department of Interventional Cardiology—Cardiovascular Diseases Institute, 700503 Iasi, Romania;
- Department of Internal Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.C.-D.); (A.C.)
| | - Radu Crisan-Dabija
- Department of Internal Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.C.-D.); (A.C.)
- Pulmonology Department, Clinic of Pulmonary Diseases, 700115 Iasi, Romania
| | - Iolanda Valentina Popa
- Department of Internal Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.C.-D.); (A.C.)
| | - Adrian Covic
- Department of Internal Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.C.-D.); (A.C.)
- Nephrology Clinic, Dialysis, and Renal Transplant Center—‘C.I. Parhon’ University Hospital, 700503 Iasi, Romania
| |
Collapse
|
8
|
Affiliation(s)
- Ajay Gupta
- Department of Orthopaedics, Maulana Azad Medical College and Associated LN Hospital, New Delhi, 110002 India
| |
Collapse
|
9
|
Clay SL, Bravo-Blas A, Wall DM, MacLeod MKL, Milling SWF. Regulatory T cells control the dynamic and site-specific polarization of total CD4 T cells following Salmonella infection. Mucosal Immunol 2020; 13:946-957. [PMID: 32457450 PMCID: PMC7567643 DOI: 10.1038/s41385-020-0299-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 04/10/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
FoxP3+ regulatory T cells (Tregs) control inflammation and maintain mucosal homeostasis, but their functions during infection are poorly understood. Th1, Th2, and Th17 cells can be identified by master transcription factors (TFs) T-bet, GATA3, and RORγT; Tregs also express these TFs. While T-bet+ Tregs can selectively suppress Th1 cells, it is unclear whether distinct Treg populations can alter Th bias. To address this, we used Salmonella enterica serotype Typhimurium to induce nonlethal colitis. Following infection, we observed an early colonic Th17 response within total CD4 T cells, followed by a Th1 bias. The early Th17 response, which contains both Salmonella-specific and non-Salmonella-specific cells, parallels an increase in T-bet+ Tregs. Later, Th1 cells and RORγT+ Tregs dominate. This reciprocal dynamic may indicate that Tregs selectively suppress Th cells, shaping the immune response. Treg depletion 1-2 days post-infection shifted the early Th17 response to a Th1 bias; however, Treg depletion 6-7 days post-infection abrogated the Th1 bias. Thus, Tregs are necessary for the early Th17 response, and for a maximal Th1 response later. These data show that Tregs shape the overall tissue CD4 T cell response and highlight the potential for subpopulations of Tregs to be used in targeted therapeutic approaches.
Collapse
Affiliation(s)
- Slater L. Clay
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK ,grid.38142.3c000000041936754XDepartment of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115 USA
| | - Alberto Bravo-Blas
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK ,grid.23636.320000 0000 8821 5196Institute of Cancer Sciences, University of Glasgow and Cancer Research UK Beatson Institute, Glasgow, G61 1BD UK
| | - Daniel M. Wall
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK
| | - Megan K. L. MacLeod
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK
| | - Simon W. F. Milling
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK
| |
Collapse
|
10
|
Patankar YR, Sutiwisesak R, Boyce S, Lai R, Lindestam Arlehamn CS, Sette A, Behar SM. Limited recognition of Mycobacterium tuberculosis-infected macrophages by polyclonal CD4 and CD8 T cells from the lungs of infected mice. Mucosal Immunol 2020; 13:140-148. [PMID: 31636345 PMCID: PMC7161428 DOI: 10.1038/s41385-019-0217-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 02/04/2023]
Abstract
Immune responses following Mycobacterium tuberculosis (Mtb) infection or vaccination are frequently assessed by measuring T-cell recognition of crude Mtb antigens, recombinant proteins, or peptide epitopes. We previously showed that not all Mtb-specific T cells recognize Mtb-infected macrophages. Thus, an important question is what proportion of T cells elicited by Mtb infection recognize Mtb-infected macrophages. We address this question by developing a modified elispot assay using viable Mtb-infected macrophages, a low multiplicity of infection and purified T cells. In C57BL/6 mice, CD4 and CD8 T cells were classically MHC restricted. Comparable frequencies of T cells that recognize Mtb-infected macrophages were determined using interferon-γ elispot and intracellular cytokine staining, and lung CD4 T cells more sensitively recognized Mtb-infected macrophages than lung CD8 T cells. Compared to the relatively high frequencies of T cells specific for antigens such as ESAT-6 and TB10.4, low frequencies of total pulmonary T cells elicited by aerosolized Mtb infection recognize Mtb-infected macrophages. Finally, we demonstrate that BCG vaccination elicits T cells that recognize Mtb-infected macrophages. We propose that the frequency of T cells that recognize infected macrophages could correlate with protective immunity and may be an alternative approach to measuring T-cell responses to Mtb antigens.
Collapse
Affiliation(s)
- Yash R. Patankar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rujapak Sutiwisesak
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Shayla Boyce
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rocky Lai
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Cecilia S. Lindestam Arlehamn
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Alessandro Sette
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA ,0000 0001 2107 4242grid.266100.3Department of Medicine, University of California San Diego, La Jolla, CA 92093 USA
| | - Samuel M. Behar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| |
Collapse
|
11
|
van Langelaar J, Rijvers L, Janssen M, Wierenga-Wolf AF, Melief MJ, Siepman TA, de Vries HE, Unger PPA, van Ham SM, Hintzen RQ, van Luijn MM. Induction of brain-infiltrating T-bet-expressing B cells in multiple sclerosis. Ann Neurol 2019; 86:264-278. [PMID: 31136008 PMCID: PMC6771938 DOI: 10.1002/ana.25508] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/24/2022]
Abstract
Objective Results from anti‐CD20 therapies demonstrate that B‐ and T‐cell interaction is a major driver of multiple sclerosis (MS). The local presence of B‐cell follicle‐like structures and oligoclonal bands in MS patients indicates that certain B cells infiltrate the central nervous system (CNS) to mediate pathology. Which peripheral triggers underlie the development of CNS‐infiltrating B cells is not fully understood. Methods Ex vivo flow cytometry was used to assess chemokine receptor profiles of B cells in blood, cerebrospinal fluid, meningeal, and brain tissues of MS patients (n = 10). Similar analyses were performed for distinct memory subsets in the blood of untreated and natalizumab‐treated MS patients (n = 38). To assess T‐bet(CXCR3)+ B‐cell differentiation, we cultured B cells from MS patients (n = 21) and healthy individuals (n = 34) under T helper 1‐ and TLR9‐inducing conditions. Their CNS transmigration capacity was confirmed using brain endothelial monolayers. Results CXC chemokine receptor 3 (CXCR3)‐expressing B cells were enriched in different CNS compartments of MS patients. Treatment with the clinically effective drug natalizumab prevented the recruitment of CXCR3high IgG1+ subsets, corresponding to their increased ability to cross CNS barriers in vitro. Blocking of interferon‐γ (IFNγ) reduced the transmigration potential and antigen‐presenting function of these cells. IFNγ‐induced B cells from MS patients showed increased T‐bet expression and plasmablast development. Additional TLR9 triggering further upregulated T‐bet and CXCR3, and was essential for IgG1 switching. Interpretation This study demonstrates that T‐bethigh IgG1+ B cells are triggered by IFNγ and TLR9 signals, likely contributing to enhanced CXCR3‐mediated recruitment and local reactivity in the CNS of MS patients. ANN NEUROL 2019;86:264–278
Collapse
Affiliation(s)
- Jamie van Langelaar
- Department of Immunology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Liza Rijvers
- Department of Immunology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Malou Janssen
- Department of Immunology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Marie-José Melief
- Department of Immunology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Theodora A Siepman
- Department of Neurology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Peter-Paul A Unger
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Rogier Q Hintzen
- Department of Immunology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Hoggarth A, Weaver A, Pu Q, Huang T, Schettler J, Chen F, Yuan X, Wu M. Mechanistic research holds promise for bacterial vaccines and phage therapies for Pseudomonas aeruginosa. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:909-924. [PMID: 30936684 PMCID: PMC6431001 DOI: 10.2147/dddt.s189847] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vaccines for Pseudomonas aeruginosa have been of longstanding interest to immunologists, bacteriologists, and clinicians, due to the widespread prevalence of hospital-acquired infection. As P. aeruginosa becomes increasingly antibiotic resistant, there is a dire need for novel treatments and preventive vaccines. Despite intense efforts, there currently remains no vaccine on the market to combat this dangerous pathogen. This article summarizes current and past vaccines under development that target various constituents of P. aeruginosa. Targeting lipopolysaccharides and O-antigens have shown some promise in preventing infection. Recombinant flagella and pili that target TLR5 have been utilized to combat P. aeruginosa by blocking its motility and adhesion. The type 3 secretion system components, such as needle-like structure PcrV or exotoxin PopB, are also potential vaccine targets. Outer membrane proteins including OprF and OprI are newer representatives of vaccine candidates. Live attenuated vaccines are a focal point in this review, and are also considered for novel vaccines. In addition, phage therapy is revived as an effective option for treating refractory infections after failure with antibiotic treatment. Many of the aforementioned vaccines act on a single target, thus lacking a broad range of protection. Recent studies have shown that mixtures of vaccines and combination approaches may significantly augment immunogenicity, thereby increasing their preventive and therapeutic potential.
Collapse
Affiliation(s)
- Austin Hoggarth
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Andrew Weaver
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Qinqin Pu
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Ting Huang
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA, .,Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Jacob Schettler
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Feng Chen
- Pulmonary and Allergy Institute, Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - Xiefang Yuan
- Pulmonary and Allergy Institute, Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - Min Wu
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| |
Collapse
|
13
|
Whiteside SK, Snook JP, Ma Y, Sonderegger FL, Fisher C, Petersen C, Zachary JF, Round JL, Williams MA, Weis JJ. IL-10 Deficiency Reveals a Role for TLR2-Dependent Bystander Activation of T Cells in Lyme Arthritis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1457-1470. [PMID: 29330323 PMCID: PMC5809275 DOI: 10.4049/jimmunol.1701248] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/13/2017] [Indexed: 11/19/2022]
Abstract
T cells predominate the immune responses in the synovial fluid of patients with persistent Lyme arthritis; however, their role in Lyme disease remains poorly defined. Using a murine model of persistent Lyme arthritis, we observed that bystander activation of CD4+ and CD8+ T cells leads to arthritis-promoting IFN-γ, similar to the inflammatory environment seen in the synovial tissue of patients with posttreatment Lyme disease. TCR transgenic mice containing monoclonal specificity toward non-Borrelia epitopes confirmed that bystander T cell activation was responsible for disease development. The microbial pattern recognition receptor TLR2 was upregulated on T cells following infection, implicating it as marker of bystander T cell activation. In fact, T cell-intrinsic expression of TLR2 contributed to IFN-γ production and arthritis, providing a mechanism for microbial-induced bystander T cell activation during infection. The IL-10-deficient mouse reveals a novel TLR2-intrinsic role for T cells in Lyme arthritis, with potentially broad application to immune pathogenesis.
Collapse
Affiliation(s)
- Sarah K Whiteside
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - Jeremy P Snook
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - Ying Ma
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - F Lynn Sonderegger
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - Colleen Fisher
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - Charisse Petersen
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - James F Zachary
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802
| | - June L Round
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - Matthew A Williams
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| | - Janis J Weis
- Department of Pathology, University of Utah, Salt Lake City, UT 84112; and
| |
Collapse
|
14
|
Herrera-Uribe J, Zaldívar-López S, Aguilar C, Luque C, Bautista R, Carvajal A, Claros MG, Garrido JJ. Regulatory role of microRNA in mesenteric lymph nodes after Salmonella Typhimurium infection. Vet Res 2018; 49:9. [PMID: 29391047 PMCID: PMC5796392 DOI: 10.1186/s13567-018-0506-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/23/2017] [Indexed: 12/14/2022] Open
Abstract
Salmonellosis is a gastrointestinal disease caused by non-typhoidal Salmonella serovars such as Salmonella Typhimurium. This pathology is a zoonosis, and food animals with subclinical infection constitute a vast reservoir for disease. After intestinal colonization, Salmonella Typhimurium reaches mesenteric lymph nodes (MLN), where infection is controlled avoiding systemic spread. Although the molecular basis of this infection has been extensively studied, little is known about how microRNA (miRNA) regulate the expression of proteins involved in the Salmonella-host interaction. Using small RNA-seq, we examined expression profiles of MLN 2 days after infection with Salmonella Typhimurium, and we found 110 dysregulated miRNA. Among them, we found upregulated miR-21, miR-155, miR-150, and miR-221, as well as downregulated miR-143 and miR-125, all of them previously linked to other bacterial infections. Integration with proteomic data revealed 30 miRNA potentially regulating the expression of 15 proteins involved in biological functions such as cell death and survival, inflammatory response and antigenic presentation. The inflammatory response was found increased via upregulation of miRNA such as miR-21 and miR-155. Downregulation of miR-125a/b, miR-148 and miR-1 were identified as potential regulators of MHC-class I components PSMB8, HSP90B1 and PDIA3, respectively. Furthermore, we confirmed that miR-125a is a direct target of immunoproteasome component PSMB8. Since we also found miR-130 downregulation, which is associated with upregulation of HSPA8, we suggest induction of both MHC-I and MHC-II antigen presentation pathways. In conclusion, our study identifies miRNA that could regulate critical networks for antigenic presentation, inflammatory response and cytoskeletal rearrangements.
Collapse
Affiliation(s)
- Juber Herrera-Uribe
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain
| | - Sara Zaldívar-López
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain.
| | - Carmen Aguilar
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain.,Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Cristina Luque
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain
| | - Rocío Bautista
- Plataforma Andaluza de Bioinformática, Universidad de Málaga, 29590, Málaga, Spain
| | - Ana Carvajal
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, 24071, León, Spain
| | - M Gonzalo Claros
- Plataforma Andaluza de Bioinformática, Universidad de Málaga, 29590, Málaga, Spain.,Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071, Málaga, Spain
| | - Juan J Garrido
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain
| |
Collapse
|
15
|
Pham OH, O’Donnell H, Al-Shamkhani A, Kerrinnes T, Tsolis RM, McSorley SJ. T cell expression of IL-18R and DR3 is essential for non-cognate stimulation of Th1 cells and optimal clearance of intracellular bacteria. PLoS Pathog 2017; 13:e1006566. [PMID: 28817719 PMCID: PMC5574617 DOI: 10.1371/journal.ppat.1006566] [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: 03/28/2017] [Revised: 08/29/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023] Open
Abstract
Th1 cells can be activated by TCR-independent stimuli, but the importance of this pathway in vivo and the precise mechanisms involved require further investigation. Here, we used a simple model of non-cognate Th1 cell stimulation in Salmonella-infected mice to examine these issues. CD4 Th1 cell expression of both IL-18R and DR3 was required for optimal IFN-γ induction in response to non-cognate stimulation, while IL-15R expression was dispensable. Interestingly, effector Th1 cells generated by immunization rather than live infection had lower non-cognate activity despite comparable IL-18R and DR3 expression. Mice lacking T cell intrinsic expression of MyD88, an important adapter molecule in non-cognate T cell stimulation, exhibited higher bacterial burdens upon infection with Salmonella, Chlamydia or Brucella, suggesting that non-cognate Th1 stimulation is a critical element of efficient bacterial clearance. Thus, IL-18R and DR3 are critical players in non-cognate stimulation of Th1 cells and this response plays an important role in protection against intracellular bacteria.
Collapse
Affiliation(s)
- Oanh H. Pham
- Center for Comparative Medicine and Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Hope O’Donnell
- Center for Comparative Medicine and Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Aymen Al-Shamkhani
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Tobias Kerrinnes
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Stephen J. McSorley
- Center for Comparative Medicine and Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
16
|
van Aalst S, Ludwig IS, van der Zee R, van Eden W, Broere F. Bystander activation of irrelevant CD4+ T cells following antigen-specific vaccination occurs in the presence and absence of adjuvant. PLoS One 2017; 12:e0177365. [PMID: 28489886 PMCID: PMC5425230 DOI: 10.1371/journal.pone.0177365] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 01/01/2023] Open
Abstract
Autoimmune and other chronic inflammatory diseases (AID) are prevalent diseases which can severely impact the quality of life of those that suffer from the disease. In most cases, the etiology of these conditions have remained unclear. Immune responses that take place e.g. during natural infection or after vaccination are often linked with the development or exacerbation of AID. It is highly debated if vaccines induce or aggravate AID and in particular adjuvants are mentioned as potential cause. Since vaccines are given on a large scale to healthy individuals but also to elderly and immunocompromised individuals, more research is warranted. Non-specific induction of naïve or memory autoreactive T cells via bystander activation is one of the proposed mechanisms of how vaccination might be involved in AID. During bystander activation, T cells unrelated to the antigen presented can be activated without (strong) T cell receptor (TCR) ligation, but via signals derived from the ongoing response directed against the vaccine-antigen or adjuvant at hand. In this study we have set up a TCR transgenic T cell transfer mouse model by which we were able to measure local bystander activation of transferred and labeled CD4+ T cells. Intramuscular injection with the highly immunogenic Complete Freund's Adjuvant (CFA) led to local in vivo proliferation and activation of intravenously transferred CD4+ T cells in the iliac lymph node. This local bystander activation was also observed after CFA prime and Incomplete Freund's Adjuvant (IFA) boost injection. Furthermore, we showed that an antigen specific response is sufficient for the induction of a bystander activation response and the general, immune stimulating effect of CFA or IFA does not appear to increase this effect. In other words, no evidence was obtained that adjuvation of antigen specific responses is essential for bystander activation.
Collapse
Affiliation(s)
- Susan van Aalst
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Irene S. Ludwig
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Ruurd van der Zee
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Femke Broere
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
17
|
Salmonella typhimurium Infection Reduces Schistosoma japonicum Worm Burden in Mice. Sci Rep 2017; 7:1349. [PMID: 28465515 PMCID: PMC5430953 DOI: 10.1038/s41598-017-00992-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/17/2017] [Indexed: 11/09/2022] Open
Abstract
Coinfection of microorganisms is a common phenomenon in humans and animals. In order to further our understanding of the progress of coinfection and the possible interaction between different pathogens, we have built a coinfection mouse model with Schistosoma japonicum and Salmonella typhimurium, and used this model to investigate the systemic metabolic and immune responses using NMR-based metabonomics and immunological techniques. Our results show that Salmonella typhimurium (ATCC14028) infection reduces the number of adult schistosomal worms and eggs, relieves symptoms of schistosomiasis and also abates the mortality of mice infected by Schistosoma japonicum. In addition, Salmonella typhimurium infection counteracts the metabolic disturbances associated with schistosomiasis, which was reflected by the reverted levels of metabolites in coinfected mice, compared with the Schistosoma japonicum infected mice. Furthermore, immune analyses also indicate that shift of the immune response to different pathogens is a result of indirect interactions between Schistosoma japonicum and Salmonella typhimurium within the host. Salmonella typhimurium infection can ameliorate Schistosoma japonicum-caused schistosomiasis in BALB/c mice, which is most likely due to inverse immune polarization. Our work provides an insight into coinfection between Schistosoma japonicum and Salmonella typhimurium, and may further contribute to the development of new tools for controlling Schistosoma japonicum-associated diseases.
Collapse
|
18
|
Arleevskaya MI, Kravtsova OA, Lemerle J, Renaudineau Y, Tsibulkin AP. How Rheumatoid Arthritis Can Result from Provocation of the Immune System by Microorganisms and Viruses. Front Microbiol 2016; 7:1296. [PMID: 27582741 PMCID: PMC4987382 DOI: 10.3389/fmicb.2016.01296] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 08/05/2016] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of rheumatoid arthritis (RA), similar to development of a majority of inflammatory and autoimmune disorders, is largely due to an inappropriate or inadequate immune response to environmental challenges. Among these challenges, infectious agents are the undisputed leaders. Since the 1870s, an impressive list of microorganisms suspected of provoking RA has formed, and the list is still growing. Although a definite causative link between a specific infectious agent and the disease has not been established, several arguments support such a possibility. First, in the absence of a defined pathogen, the spectrum of triggering agents may include polymicrobial communities or the cumulative effect of several bacterial/viral factors. Second, the range of infectious episodes (i.e., clinical manifestations caused by pathogens) may vary in the process of RA development from preclinical to late-stage disease. Third, infectious agents might not trigger RA in all cases, but trigger it in a certain subset of the cases, or the disease onset may arise from an unfortunate combination of infections along with, for example, psychological stress and/or chronic joint tissue microtrauma. Fourth, genetic differences may have a role in the disease onset. In this review, two aspects of the problem of “microorganisms and RA” are debated. First, is there an acquired immune deficiency and, in turn, susceptibility to infections in RA patients due to the too frequent and too lengthy infections, which at last break the tolerance of self antigens? Or, second, is there a congenital deficiency in tolerance and inflammation control, which may occur even with ordinary infection frequency and duration?
Collapse
Affiliation(s)
- Marina I Arleevskaya
- Central Research Laboratory, Department of Clinical Laboratory Diagnostics, Kazan State Medical Academy Kazan, Russia
| | - Olga A Kravtsova
- Department of Biochemistry and Biotechnology, Kazan Federal University Kazan, Russia
| | - Julie Lemerle
- Laboratory of Immunology and Immunotherapy, CHU Morvan Brest, France
| | - Yves Renaudineau
- Laboratory of Immunology and Immunotherapy, CHU Morvan Brest, France
| | - Anatoly P Tsibulkin
- Central Research Laboratory, Department of Clinical Laboratory Diagnostics, Kazan State Medical Academy Kazan, Russia
| |
Collapse
|
19
|
O'Donnell H, Pham OH, Benoun JM, Ravesloot-Chávez MM, McSorley SJ. Contaminated water delivery as a simple and effective method of experimental Salmonella infection. Future Microbiol 2015; 10:1615-27. [PMID: 26439708 DOI: 10.2217/fmb.15.93] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
AIM In most infectious disease models, it is assumed that gavage needle infection is the most reliable means of pathogen delivery to the GI tract. However, this methodology can cause esophageal tearing and induces stress in experimental animals, both of which have the potential to impact early infection and the subsequent immune response. MATERIALS & METHODS C57BL/6 mice were orally infected with virulent Salmonella Typhimurium SL1344 either by intragastric gavage preceded by sodium bicarbonate, or by contamination of drinking water. RESULTS We demonstrate that water contamination delivery of Salmonella is equivalent to gavage inoculation in providing a consistent model of infection. Furthermore, exposure of mice to contaminated drinking water for as little as 4 h allowed maximal mucosal and systemic infection, suggesting an abbreviated window exists for natural intestinal entry. CONCLUSION Together, these data question the need for gavage delivery for infection with oral pathogens.
Collapse
Affiliation(s)
- Hope O'Donnell
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA.,Yersinia Research Unit, Microbiology Department, Pasteur Institute, Paris, France
| | - Oanh H Pham
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| | - Joseph M Benoun
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| | - Marietta M Ravesloot-Chávez
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| | - Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
20
|
López-Macías C, Cunningham AF. Editorial: How Salmonella Infection can Inform on Mechanisms of Immune Function and Homeostasis. Front Immunol 2015; 6:451. [PMID: 26388874 PMCID: PMC4558537 DOI: 10.3389/fimmu.2015.00451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/20/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Constantino López-Macías
- Medical Research Unit on Immunochemistry, National Medical Centre "Siglo XXI", Mexican Institute for Social Security, Specialties Hospital , Mexico City , Mexico
| | - Adam F Cunningham
- Institute for Biomedical Research, School of Immunity and Infection, University of Birmingham , Birmingham , UK
| |
Collapse
|