101
|
Abstract
Prevention of infections by vaccination remains a compelling goal to improve public health. Most infections involve the mucosae, but the development of vaccines against many of these pathogens has yet to be successful. Mucosal vaccines would make immunization procedures easier, be better suited for mass administration, and most efficiently induce immune exclusion - a term coined for non-inflammatory antibody shielding of internal body surfaces - mediated principally by secretory immunoglobulin A (SIgA). The exported antibodies are polymeric, mainly IgA dimers (pIgA) - produced by local plasma cells stimulated by antigens that target the mucosae. SIgA was early shown to be complexed with an epithelial glycoprotein - the secretory component (SC). In 1974, a common SC-dependent transport of pIgA and pentameric IgM was proposed. From the basolateral surface, pIg-SC complexes are taken up by endocytosis and finally extruded into the lumen. Membrane SC is now referred to as polymeric Ig receptor (pIgR). In 1980, it was shown to be synthesized as a larger transmembrane protein - first cloned from rabbit and then from human. Mice deficient for pIgR showed that this is the only receptor responsible for epithelial transport of IgA and IgM. In the gut, induction of B cells occurs in gut-associated lymphoid tissue, particularly the Peyer's patches, but also in mesenteric lymph nodes. Plasma cell differentiation is accomplished in the lamina propria to which the memory/effector cells home. The airways also receive such cells from nasopharynx-associated lymphoid tissue - but by different homing receptors. Such compartmentalization is a challenge for development of mucosal vaccines.
Collapse
Affiliation(s)
- P Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation, University of Oslo, Division and Institute of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| |
Collapse
|
102
|
Matharu KS, Mizoguchi E, Cotoner CA, Nguyen DD, Mingle B, Iweala OI, McBee ME, Stefka AT, Prioult G, Haigis KM, Bhan AK, Snapper SB, Murakami H, Schauer DB, Reinecker HC, Mizoguchi A, Nagler CR. Toll-like receptor 4-mediated regulation of spontaneous Helicobacter-dependent colitis in IL-10-deficient mice. Gastroenterology 2009; 137:1380-90.e1-3. [PMID: 19596011 PMCID: PMC2757440 DOI: 10.1053/j.gastro.2009.07.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Revised: 06/26/2009] [Accepted: 07/01/2009] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS The commensal microbiota is believed to have an important role in regulating immune responsiveness and preventing intestinal inflammation. Intestinal microbes produce signals that regulate inflammation via Toll-like receptor (TLR) signaling, but the mechanisms of this process are poorly understood. We investigated the role of the anti-inflammatory cytokine interleukin (IL)-10 in this signaling pathway using a mouse model of colitis. METHODS Clinical, histopathologic, and functional parameters of intestinal inflammation were evaluated in TLR4(-/-), IL-10(-/-), and TLR4(-/-) x IL-10(-/-) mice that were free of specific pathogens and in TLR4(-/-) x IL-10(-/-) mice following eradication and reintroduction of Helicobacter hepaticus. Regulatory T-cell (Treg) function was evaluated by crossing each of the lines with transgenic mice that express green fluorescent protein under control of the endogenous regulatory elements of Foxp3. Apoptotic cells in the colonic lamina propria were detected by a TUNEL assay. RESULTS TLR4-mediated signals have 2 interrelated roles in promoting inflammation in TLR4(-/-) x IL-10(-/-) mice. In the absence of TLR4-mediated signals, secretion of proinflammatory and immunoregulatory cytokines is dysregulated. Tregs (Foxp3(+)) that secrete interferon-gamma and IL-17 accumulate in the colonic lamina propria of TLR4(-/-) x IL-10(-/-) mice and do not prevent inflammation. Aberrant control of epithelial cell turnover results in the persistence of antigen-presenting cells that contain apoptotic epithelial fragments in the colonic lamina propria of Helicobacter-infected TLR4(-/-) mice. CONCLUSIONS In mice that lack both IL-10- and TLR4-mediated signals, aberrant regulatory T-cell function and dysregulated control of epithelial homeostasis combine to exacerbate intestinal inflammation.
Collapse
Affiliation(s)
- Kabir S. Matharu
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Emiko Mizoguchi
- Gastrointestinal Unit, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Carmen Alonso Cotoner
- Gastrointestinal Unit, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Deanna D. Nguyen
- Gastrointestinal Unit, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Bethany Mingle
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Onyinye I. Iweala
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Megan E. McBee
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Andrew T. Stefka
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Guenolee Prioult
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Nestle Research Center, Lausanne 26, Switzerland
| | - Kevin M. Haigis
- Molecular Pathology/Cancer Center, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Atul K. Bhan
- Experimental Pathology Unit, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Scott B. Snapper
- Gastrointestinal Unit, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Hidehiro Murakami
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Ehime University School of Medicine, Ehime 791 0295 Japan
| | - David B. Schauer
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Hans-Christian Reinecker
- Gastrointestinal Unit, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Atsushi Mizoguchi
- Experimental Pathology Unit, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| | - Cathryn R. Nagler
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston 02114 and Charlestown 02129 MA
| |
Collapse
|
103
|
Westendorf AM, Fleissner D, Hansen W, Buer J. T cells, dendritic cells and epithelial cells in intestinal homeostasis. Int J Med Microbiol 2009; 300:11-8. [PMID: 19781991 DOI: 10.1016/j.ijmm.2009.08.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mucosal immune system of the intestinal tract is continuously exposed to both potential pathogens and beneficial commensal microorganism. A variety of mechanisms contribute to the ability of the gut to either react or remain tolerant to antigen present in the intestinal lumen. Antigens of the gut commensals are not simply ignored, but rather trigger an active immunosuppressive process, which prevents the outcome of immunopathology. The aim of this review is to provide an update on the mechanism of intestinal homeostasis, with particular focus on the complex crosstalk between T cells, dendritic cells and intestinal epithelial cells.
Collapse
Affiliation(s)
- Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, Hufelandstrasse 55, D-45122 Essen, Germany.
| | | | | | | |
Collapse
|
104
|
Thomas C, Gupta V, Ahsan F. Influence of surface charge of PLGA particles of recombinant hepatitis B surface antigen in enhancing systemic and mucosal immune responses. Int J Pharm 2009; 379:41-50. [DOI: 10.1016/j.ijpharm.2009.06.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/28/2009] [Accepted: 06/03/2009] [Indexed: 11/30/2022]
|
105
|
Andreu-Ballester JC, Ballester F, Pérez-Griera J, Amigo V, Peñarroja-Otero C, Colomer-Rubio E, Ortiz-Tarín I, Pelayo V, García-Hernández P, Rodero M, Cuéllar C. Differential effect of appendectomy and tonsillectomy on anti-Kudoa sp. antibodies in patients with MALTectomy. Parasitol Int 2009; 58:401-5. [PMID: 19683065 DOI: 10.1016/j.parint.2009.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 07/31/2009] [Accepted: 08/04/2009] [Indexed: 12/22/2022]
Abstract
We found an association between tonsillectomized patients and subsequent appendicitis. We also observed that MALTectomy significantly decreased secretory IgA levels in serum of patients, being this decrease more pronounced when both operations (tonsillectomy and appendectomy) had been performed. The elevated humoral responses detected previously by us in BALB/c mice immunized with Kudoa sp. pseudocyst extracts and the high IgG1 and IgE levels induced by the oral administration of Kudoa sp. pseudocysts to BALB/c mice showed the possible immunopathological effects in man from the ingestion of Kudoa sp. infected fish. We use the ELISA method to investigate the possible relationship between MALTectomy (tonsillectomy and appendectomy) and specific antibody levels to Kudoa sp. Both anti-Kudoa sp. specific antibody levels and the number of patients that recognized Kudoa sp. antigens were greater in tonsillectomy patients when compared to the control and the other studied groups (appendectomized and appendectomized+tonsillectomies patients). Tonsillectomy was associated to a switch in the class of immunoglobulins involved in these responses and these responses may be abrogated by appendectomy. Tonsils and appendix may respond in different ways to Kudoa sp. antigens and these different reactions may be involved in some immunopathological reactions.
Collapse
|
106
|
Iweala OI, Smith DW, Matharu KS, Sada-Ovalle I, Nguyen DD, Dekruyff RH, Umetsu DT, Behar SM, Nagler CR. Vaccine-induced antibody isotypes are skewed by impaired CD4 T cell and invariant NKT cell effector responses in MyD88-deficient mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:2252-60. [PMID: 19620295 DOI: 10.4049/jimmunol.0804011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The requirement for TLR signaling in the initiation of an Ag-specific Ab response is controversial. In this report we show that a novel OVA-expressing recombinant Salmonella vaccine (Salmonella-OVA) elicits a Th1-biased cell-mediated and serum Ab response upon oral or i.p. immunization of C57BL/6 mice. In MyD88(-/-) mice, Th1-dependent Ab responses are greatly reduced while Th2-dependent Ab isotypes are elevated in response to oral and i.p., but not s.c. footpad, immunization. When the T effector response to oral vaccination is examined we find that activated, adoptively transferred Ag-specific CD4(+) T cells accumulate in the draining lymph nodes, but fail to produce IFN-gamma, in MyD88(-/-) mice. Moreover, CD1d tetramer staining shows that invariant NKT cells are activated in response to oral Salmonella-OVA vaccination in wild-type, but not MyD88(-/-), mice. Treatment with neutralizing Ab to CD1d reduces the OVA-specific Ab response only in MyD88-sufficient wild-type mice, suggesting that both Ag-specific CD4 T cell and invariant NKT cell effector responses to Salmonella-OVA vaccination are MyD88 dependent. Taken together, our data indicate that the type of adaptive immune response generated to this live attenuated vaccine is regulated by both the presence of MyD88-mediated signals and vaccination route, which may have important implications for future vaccine design.
Collapse
Affiliation(s)
- Onyinye I Iweala
- Center for Immunology and Inflammatory Disease, Division of Rheumatology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Wood C, Keeling S, Bradley S, Johnson-Green P, Green-Johnson JM. Interactions in the mucosal microenvironment: vasoactive intestinal peptide modulates the down-regulatory action ofLactobacillus rhamnosuson LPS-induced interleukin-8 production by intestinal epithelial cells. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.1080/08910600701278722] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Christine Wood
- School of Nutrition and Dietetics, Acadia University, Wolfville, NS
| | - Suzanne Keeling
- Faculty of Science, University of Ontario Institute of Technology, Oshawa, ON, Canada
| | - Shannon Bradley
- Faculty of Science, University of Ontario Institute of Technology, Oshawa, ON, Canada
| | - Perry Johnson-Green
- School of Nutrition and Dietetics, Acadia University, Wolfville, NS
- Faculty of Science, University of Ontario Institute of Technology, Oshawa, ON, Canada
| | | |
Collapse
|
108
|
Reynoso ED, Elpek KG, Francisco L, Bronson R, Bellemare-Pelletier A, Sharpe AH, Freeman GJ, Turley SJ. Intestinal tolerance is converted to autoimmune enteritis upon PD-1 ligand blockade. THE JOURNAL OF IMMUNOLOGY 2009; 182:2102-12. [PMID: 19201863 DOI: 10.4049/jimmunol.0802769] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The B7 family member programmed death-1 ligand (PD-L1) has been shown to play an inhibitory role in the regulation of T cell responses in several organs. However, the role of PD-L1 in regulating tolerance to self-Ags of the small intestine has not been previously addressed. In this study, we investigated the role of PD-L1 in CD8(+) T cell tolerance to an intestinal epithelium-specific Ag using the iFABP-tOVA transgenic mouse model, in which OVA is expressed as a self-Ag throughout the small intestine. Using adoptive transfer of naive OVA-specific CD8(+) T cells, we show that loss of PD-1:PD-L1 signaling, by either Ab-mediated PD-L1 blockade or transfer of PD-1(-/-) T cells, leads to considerable expansion of OVA-specific CD8(+) T cells and their differentiation into effector cells capable of producing proinflammatory cytokines. A fatal CD8(+) T cell-mediated inflammatory response develops rapidly against the small bowel causing destruction of the epithelial barrier, severe blunting of intestinal villi, and recruitment and activation of myeloid cells. This response is highly specific because immune destruction selectively targets the small intestine but not other organs. Collectively, these results indicate that loss of the PD-1:PD-L1 inhibitory pathway breaks CD8(+) T cell tolerance to intestinal self-Ag, thus leading to severe enteric autoimmunity.
Collapse
Affiliation(s)
- Erika D Reynoso
- Division of Medical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Che C, Pang X, Hua X, Zhang B, Shen J, Zhu J, Wei H, Sun L, Chen P, Cui L, Zhao L, Yang Q. Effects of Human Fecal Flora on Intestinal Morphology and Mucosal Immunity in Human Flora-associated Piglet. Scand J Immunol 2009; 69:223-33. [DOI: 10.1111/j.1365-3083.2008.02211.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
110
|
Utilization of the Porcine System to Study Lymphotoxin-β Regulation in Intestinal Lymphoid Tissue. Biochem Genet 2009; 47:126-36. [DOI: 10.1007/s10528-008-9212-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Accepted: 07/22/2008] [Indexed: 10/21/2022]
|
111
|
Peripheral tolerance induction by lymph node stroma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 633:113-27. [PMID: 19209685 DOI: 10.1007/978-0-387-79311-5_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review we have highlighted the role of LNSCs in the regulation of CD8+ T cell immune responses in peripheral lymph nodes, thereby adding another layer of protection, in addition to the role of resting DCs, against autoimmunity. LNSCs have recently been implicated in the induction of peripheral CD8+ T cell tolerance due to their ability to endogenously express, process, and present PTAs. Furthermore, LNSCs express surface molecules, such as MHC class II and PD-L1, similar to those expressed by mTECs in the thymus and APCs. For future studies it will be important to address some of the new questions that have emerged with respect to the biology and function of LNSCs. Further work will help us to (1) dissect the specific roles that DCs and LNSCs have in the induction and maintenance of tolerance to intestinal antigens, (2) gain a more in-depth understanding of the molecular mechanisms underlying self-tolerance induction by LNSCs and the impact of inflammation on this function, (3) evaluate the relationship of LNSCs to the FRN, and (4) determine if the APC function of LNSCs extends to the acquisition and presentation of exogenous antigens. Finally, it is important to mention that so far the studies done on LNSCs have focused on their role in CD8+ T cell tolerance. At the moment, we do not know if presentation of PTAs by LNSCs can also induce tolerance of CD4+ T cells. Based on the finding that LNSCs express MHC class II (I-A(b)) molecules it is possible that they may present self-antigens to CD4+ T cells and induce tolerance. However, this has yet to be elucidated.
Collapse
|
112
|
Bonner A, Almogren A, Furtado PB, Kerr MA, Perkins SJ. The nonplanar secretory IgA2 and near planar secretory IgA1 solution structures rationalize their different mucosal immune responses. J Biol Chem 2008; 284:5077-87. [PMID: 19109255 DOI: 10.1074/jbc.m807529200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Secretory IgA (SIgA) is the most prevalent human antibody and is central to mucosal immunity. It exists as two subclasses, SIgA1 and SIgA2, where SIgA2 has a shorter hinge joining the Fab and Fc regions. Both forms of SIgA are predominantly dimeric and contain an additional protein called the secretory component (SC) that is attached during the secretory process and is believed to protect SIgA in harsh mucosal conditions. Here we locate the five SC domains relative to dimeric IgA2 within SIgA2 using constrained scattering modeling. The x-ray and sedimentation parameters showed that SIgA2 has an extended solution structure. The constrained modeling of SIgA2 was initiated using two IgA2 monomers that were positioned according to our best fit solution structure for dimeric IgA1. SC was best located along the convex edge of the Fc-Fc region. The best fit models showed that SIgA2 is significantly nonplanar in its structure, in distinction to our previous near planar SIgA1 structure. Both the shorter IgA2 hinges and the presence of SC appear to displace the four Fab regions out of the Fc plane in SIgA2. This may explain the noncovalent binding of SC in some SIgA2 molecules. This nonplanar structure is predicted to result in specific immune properties for SIgA2 and SIgA1. It may explain differences observed between the SIgA1 and SIgA2 subclasses in terms of their interactions with antigens, susceptibility to proteases, effects on receptors, and distribution in different tissues. The different structures account for the prevalence of both forms in mucosal secretions.
Collapse
Affiliation(s)
- Alexandra Bonner
- Institute of Structural and Molecular Biology, Division of Biosciences, Darwin Building, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | | | | | | | | |
Collapse
|
113
|
Regulatory T cells modulate staphylococcal enterotoxin B-induced effector T-cell activation and acceleration of colitis. Infect Immun 2008; 77:707-13. [PMID: 19064639 DOI: 10.1128/iai.00822-08] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Oral administration of bacterial superantigen Staphylococcus aureus enterotoxin B (SEB) activates mucosal T cells but does not cause mucosal inflammation. We examined the effect of oral SEB on the development of mucosal inflammation in mice in the absence of regulatory T (Treg) cells. SCID mice were fed SEB 3 and 7 days after reconstitution with CD4(+) CD45RB(high) or CD4(+) CD45RB(high) plus CD4(+) CD45RB(low) T cells. Mice were sacrificed at different time points to examine changes in tissue damage and in T-cell phenotypes. Feeding SEB failed to produce any clinical effect on SCID mice reconstituted with CD4(+) CD45RB(high) and CD4(+) CD45RB(low) T cells, but feeding SEB accelerated the development of colitis in SCID mice reconstituted with CD4(+) CD45RB(high) T cells alone. The latter was associated with an increase in the number of CD4(+) Vbeta8(+) T cells expressing CD69 and a significantly lower number of CD4(+) CD25(+) Foxp3(+) T cells. These changes were not observed in SCID mice reconstituted with both CD45RB(high) and CD45RB(low) T cells. In addition, SEB impaired the development of Treg cells in the SCID mice reconstituted with CD4(+) CD45RB(high) T cells alone but had no direct effect on Treg cells. In the absence of Treg cells, feeding SEB induced activation of mucosal T cells and accelerated the development of colitis. This suggests that Treg cells prevent SEB-induced mucosal inflammation through modulation of SEB-induced T-cell activation.
Collapse
|
114
|
|
115
|
|
116
|
Burkey TE, Skjolaas KA, Minton JE. Board-invited review: porcine mucosal immunity of the gastrointestinal tract. J Anim Sci 2008; 87:1493-501. [PMID: 19028849 DOI: 10.2527/jas.2008-1330] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The gastrointestinal tract (GIT) constitutes one of the largest immunological organs of the body. The GIT must permit absorption of nutrients while also maintaining the ability to respond appropriately to a diverse milieu of dietary and microbial antigenic components. Because of the diverse population of antigenic components within the GIT, a sophisticated mucosal immune system has evolved that relies on collaboration between the innate and adaptive arms of immunity. The collaborative, mucosal immune effort offers protection from harmful pathogens while also being tolerant of dietary antigens and normal microbial flora. Knowledge with respect to porcine mucosal immunity is important as we strive to understand the interrelationships among GIT physiology, immunology, and the resident microbiota. The aim of this review is to provide a descriptive overview of GIT immunity and components of the mucosal immune system and to highlight differences that exist between the porcine species and other mammals.
Collapse
Affiliation(s)
- T E Burkey
- Department of Animal Science, University of Nebraska, Lincoln 68583, USA.
| | | | | |
Collapse
|
117
|
Man AL, Lodi F, Bertelli E, Regoli M, Pin C, Mulholland F, Satoskar AR, Taussig MJ, Nicoletti C. Macrophage migration inhibitory factor plays a role in the regulation of microfold (M) cell-mediated transport in the gut. THE JOURNAL OF IMMUNOLOGY 2008; 181:5673-80. [PMID: 18832726 DOI: 10.4049/jimmunol.181.8.5673] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been shown previously that certain bacteria rapidly (3 h) up-regulated in vivo microfold cell (M cell)-mediated transport of Ag across the follicle-associated epithelium of intestinal Peyer's patch. Our aim was to determine whether soluble mediators secreted following host-bacteria interaction were involved in this event. A combination of proteomics and immunohistochemical analyses was used to identify molecules produced in the gut in response to bacterial challenge in vivo; their effects were then tested on human intestinal epithelial cells in vitro. Macrophage migration inhibitory factor (MIF) was the only cytokine produced rapidly after in vivo bacterial challenge by CD11c(+) cells located beneath the M cell-rich area of the follicle-associated epithelium of the Peyer's patch. Subsequently, in vitro experiments conducted using human Caco-2 cells showed that, within hours, MIF induced the appearance of cells that showed temperature-dependent transport of microparticles and M cell-specific bacterium Vibrio cholerae, and acquired biochemical features of M cells. Furthermore, using an established in vitro human M cell model, we showed that anti-MIF Ab blocked Raji B cell-mediated conversion of Caco-2 cells into Ag-sampling cells. Finally, we report that MIF(-/-) mice, in contrast to wild-type mice, failed to show increased M cell-mediated transport following in vivo bacterial challenge. These data show that MIF plays a role in M cell-mediated transport, and cross-talk between bacteria, gut epithelium, and immune system is instrumental in regulating key functions of the gut, including M cell-mediated Ag sampling.
Collapse
Affiliation(s)
- Angela L Man
- Programme of Gastrointestinal Tract Biology and Health, Institute of Food Research, Norwich, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Abstract
The gastrointestinal innate and adaptive immune system continuously faces the challenge of potent stimuli from the commensal microflora and food constituents. These local immune responses require a tight control, the outcome of which is in most cases the induction of tolerance. Local T cell immunity is an important compartment of the specific intestinal immune system. T cell reactivity is programmed during the initial stage of its activation by professional presenting cells. Mucosal dendritic cells (DCs) are assumed to play key roles in regulating immune responses in the antigen-rich gastrointestinal environment. Mucosal DCs are a heterogeneous population that can either initiate (innate and adaptive) immune responses, or control intestinal inflammation and maintain tolerance. Defects in this regulation are supposed to lead to the two major forms of inflammatory bowel disease (IBD), Crohn’s disease (CD) and ulcerative colitis (UC). This review will discuss the emerging role of mucosal DCs in regulating intestinal inflammation and immune responses.
Collapse
|
119
|
Eksteen B, Liaskou E, Adams DH. Lymphocyte homing and its role in the pathogenesis of IBD. Inflamm Bowel Dis 2008; 14:1298-312. [PMID: 18393377 DOI: 10.1002/ibd.20453] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic disorder of chronic inflammation of the gastrointestinal tract. Experimental models of IBD and results from human genomewide linkage studies suggest that the primary defect that leads to IBD is an inappropriate mucosal immune response to normal intestinal microbes. Genetic alterations not only confer increased susceptibility to IBD but also appear to determine the nature and location of the intestinal inflammation, as is evident in patients with genetic alterations of NOD2 and their susceptibility for ileal Crohn's disease. IBD has traditionally been classified into 2 subtypes, namely, ulcerative colitis (UC) and Crohn's disease (CD), based on histological appearance and anatomical distribution. However, an increasing body of data supports the concept that IBD is an umbrella diagnosis encompassing a variety of disorders with distinct genetic, microbial, and environmental determinants that cluster either into a UC or CD phenotype. The shared common pathway is uncontrolled intestinal inflammation. A key element in the pathogenesis of intestinal inflammation in both UC and CD is increased leukocyte recruitment from the circulation, and this provides a potential target for pharmaceutical inhibition. In this article we review the current understanding of the molecules that determine leukocyte trafficking to the gut and highlight opportunities where their inhibition could be exploited to treat IBD.(Inflamm Bowel Dis 2008).
Collapse
Affiliation(s)
- Bertus Eksteen
- Liver Research Laboratories, MRC Centre for Immune Regulation, Institute for Biomedical Research, Medical School, University of Birmingham, Birmingham, United Kingdom
| | | | | |
Collapse
|
120
|
Eskan MA, Rose BG, Benakanakere MR, Zeng Q, Fujioka D, Martin MH, Lee MJ, Kinane DF. TLR4 and S1P receptors cooperate to enhance inflammatory cytokine production in human gingival epithelial cells. Eur J Immunol 2008; 38:1138-47. [PMID: 18395849 DOI: 10.1002/eji.200737898] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Toll-like receptors (TLR) are pattern recognition receptors for highly conserved microbial molecular patterns. Activation of TLR is a pivotal step in the initiation of innate, inflammatory, and immune defense mechanisms. Recent findings indicate that G protein-coupled receptors (GPCR) may modulate TLR signaling, but it is unclear which GPCR are involved in this process. One such cooperation between GPCR and TLR can be attributed to the sphingosine 1-phosphate (S1P) receptor family. The S1P receptors (S1P1-5) are a family of GPCR with a high affinity for S1P, a serum-borne bioactive lipid associated with diverse biological activities such as inflammation and healing. In this study, we show that pro-inflammatory cytokine production, including IL-6 and IL-8, was increased with LPS and concomitant S1P stimulation. Furthermore, elevated cytokine production following LPS and S1P challenge in human gingival epithelial cells (HGEC) was significantly reduced when TLR4, S1P1 or S1P3 signaling was blocked. Our study also shows that S1P1 and S1P3 expression was induced by LPS in HGEC, and this elevated expression enhanced the influence of S1P in its cooperation with TLR4 to increase cytokine production. This cooperation between TLR4 and S1P1 or S1P3 demonstrates that TLR4 and GPCR can interact to enhance cytokine production in epithelial cells.
Collapse
Affiliation(s)
- Mehmet A Eskan
- Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, University of Louisville, Louisville,KY 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
121
|
tRNA modification by GidA/MnmE is necessary for Streptococcus pyogenes virulence: a new strategy to make live attenuated strains. Infect Immun 2008; 76:3176-86. [PMID: 18426891 DOI: 10.1128/iai.01721-07] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studies directed at vaccine development and mucosal immunity against Streptococcus pyogenes would benefit from the availability of live attenuated strains. Our approach for production of candidate live attenuated strains was to identify mutations that did not alter growth in vitro and did not alter the overall complement of virulence factors produced but did result in reduced levels of expression of multiple secreted virulence factors. A global reduction but not elimination of expression would likely lead to attenuation while maximizing the number of antigenic targets available for stimulation of immunity. Adaptation of Tn5-based transposome mutagenesis to S. pyogenes with initial screening for reduced expression of the SpeB protease resulted in identification of mutations in gidA, which encodes an enzyme involved in tRNA modification. Reduced SpeB expression was due to delayed onset of speB transcription resulting from reduced translation efficiency of the message for RopB, a transcriptional activator. Overall, GidA(-) mutants had a nearly normal global transcription profile but expressed significantly reduced levels of multiple virulence factors due to impaired translation efficiencies. A translation defect was supported by the observation that mutants lacking MnmE, which functions in the same tRNA modification pathway as GidA, phenocopied GidA deficiency. The mutants stimulated a cytokine response in cultured macrophages identical to that in the wild type, with the exception of reduced levels of tumor necrosis factor alpha and interleukin-23. Significantly, GidA(-) mutants were highly attenuated in the murine ulcer model of soft tissue infection. These characteristics suggest that GidA pathway tRNA modification mutants are attractive candidates for further evaluation as live attenuated strains.
Collapse
|
122
|
Niess JH, Leithäuser F, Adler G, Reimann J. Commensal gut flora drives the expansion of proinflammatory CD4 T cells in the colonic lamina propria under normal and inflammatory conditions. THE JOURNAL OF IMMUNOLOGY 2008; 180:559-68. [PMID: 18097058 DOI: 10.4049/jimmunol.180.1.559] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We tested in B6 mice whether the local expansion of CD4 T cells producing proinflammatory cytokines including IL-17 (Th17 cells) in the colonic lamina propria (cLP) depends on the commensal microflora. High numbers of CD4 Th17 cells were found in the lamina propria of the ileum and colon but not the duodenum, jejunum, mesenteric lymph nodes, spleen, or liver of specific pathogen-free (SPF) mice. The microflora is required for the accumulation of cytokine (IL-17, IFN-gamma, TNF-alpha, IL-10)-producing CD4 T cells in the cLP because only low numbers of cytokine-producing cLP CD4 T cells were found in syngeneic (age- and sex-matched) germfree mice. The fraction of cLP Th17 cells was higher in (type I and type II) IFN- but not IL-4- or IL-12p40-deficient SPF congenics. cLP CD4 Th17 cells produce IL-17 but not IFN-gamma, TNF-alpha, IL-4, or IL-10. cLP CD4 Th17 cells accumulate locally in colitis induced by adoptive transfer of IFN-gamma+/+ or IFN-gamma-/- CD4 T cells into congenic SPF (but not germfree) RAG-/- hosts. In this colitis model, cLP CD4 T cells that "spontaneously" produce IL-17 progressively increase in number in the inflamed cLP, and increasing serum IL-17 levels appear as the disease progresses. Commensal bacteria-driven, local expansion of cLP CD4 Th17 cells may contribute to the pathogenesis of this inflammatory bowel disease.
Collapse
|
123
|
Intestinal alkaline phosphatase is a gut mucosal defense factor maintained by enteral nutrition. Proc Natl Acad Sci U S A 2008; 105:3551-6. [PMID: 18292227 DOI: 10.1073/pnas.0712140105] [Citation(s) in RCA: 274] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Under conditions of starvation and disease, the gut barrier becomes impaired, and trophic feeding to prevent gut mucosal atrophy has become a standard treatment of critically ill patients. However, the mechanisms responsible for the beneficial effects of enteral nutrition have remained a mystery. Using in vitro and in vivo models, we demonstrate that the brush-border enzyme, intestinal alkaline phosphatase (IAP), has the ability to detoxify lipopolysaccharide and prevent bacterial invasion across the gut mucosal barrier. IAP expression and function are lost with starvation and maintained by enteral feeding. It is likely that the IAP silencing that occurs during starvation is a key component of the gut mucosal barrier dysfunction seen in critically ill patients.
Collapse
|
124
|
Kadaoui KA, Corthésy B. Secretory IgA mediates bacterial translocation to dendritic cells in mouse Peyer's patches with restriction to mucosal compartment. THE JOURNAL OF IMMUNOLOGY 2008; 179:7751-7. [PMID: 18025221 DOI: 10.4049/jimmunol.179.11.7751] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In addition to fulfilling its function of immune exclusion at mucosal surfaces, secretory IgA (SIgA) Ab exhibits the striking feature to adhere selectively to M cells in the mouse and human intestinal Peyer's patches (PPs). Subsequent uptake drives the SIgA Ab to dendritic cells (DCs), which become partially activated. Using freshly isolated mouse DCs, we found that the interaction with SIgA was tissue and DC subtype dependent. Only DCs isolated from PPs and mesenteric lymph nodes interacted with the Ab. CD11c(+)CD11b(+) DCs internalized SIgA, while CD11c(+)CD19(+) DCs only bound SIgA on their surface, and no interaction occurred with CD11c(+)CD8alpha(+) DCs. We next examined whether SIgA could deliver a sizeable cargo to PP DCs in vivo by administering SIgA-Shigella flexneri immune complexes into a mouse ligated intestinal loop containing a PP. We found that such immune complexes entered the PPs and were internalized by subepithelial dome PP DCs, in contrast to S. flexneri alone that did not penetrate the intestinal epithelium in mice. Dissemination of intraepithelial S. flexneri delivered as immune complexes was limited to PPs and mesenteric lymph nodes. We propose that preexisting SIgA Abs associated with microbes contribute to mucosal defense by eliciting responses that prevent overreaction while maintaining productive immunity.
Collapse
Affiliation(s)
- Khalil A Kadaoui
- R&D Laboratory of the Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon, Switzerland
| | | |
Collapse
|
125
|
Immune response by nasal delivery of hepatitis B surface antigen and codelivery of a CpG ODN in alginate coated chitosan nanoparticles. Eur J Pharm Biopharm 2008; 69:405-16. [PMID: 18364251 DOI: 10.1016/j.ejpb.2008.01.019] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2007] [Revised: 12/28/2007] [Accepted: 01/15/2008] [Indexed: 11/24/2022]
Abstract
Alginate coated chitosan nanoparticles were previously developed with the aim of protecting the antigen, adsorbed on the surface of those chitosan nanoparticles, from enzymatic degradation at mucosal surfaces. In this work, this new delivery system was loaded with the recombinant hepatitis B surface antigen (HBsAg) and applied to mice by the intranasal route. Adjuvant effect of the delivery system was studied by measuring anti-HBsAg IgG in serum, anti-HBsAg sIgA in faeces extracts or nasal and vaginal secretions and interferon-gamma production in supernatants of the spleen cells. The mice were primed with 10 microg of the vaccine associated or not with nanoparticles and associated or not with 10 microg CpG oligodeoxynucleotide (ODN) followed by two sequential boosts at three week intervals. The association of HBsAg with the alginate coated chitosan nanoparticles, administered intranasally to the mice, gave rise to the humoral mucosal immune response. Humoral systemic immune response was not induced by the HBsAg loaded nanoparticles alone. The generation of Th1-biased antigen-specific systemic antibodies, however, was observed when HBsAg loaded nanoparticles were applied together with a second adjuvant, the immunopotentiator, CpG ODN. Moreover, all intranasally vaccinated groups showed higher interferon-gamma production when compared to naïve mice.
Collapse
|
126
|
Rescigno M, Mileti E. Interactions between Epithelial Cells and Dendritic Cells in Bacterial Handling. Biosci Microflora 2008. [DOI: 10.12938/bifidus.27.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
127
|
Kogan AN, von Andrian UH. Lymphocyte Trafficking. Microcirculation 2008. [DOI: 10.1016/b978-0-12-374530-9.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
128
|
Hirata Y, Broquet AH, Menchén L, Kagnoff MF. Activation of innate immune defense mechanisms by signaling through RIG-I/IPS-1 in intestinal epithelial cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:5425-32. [PMID: 17911629 DOI: 10.4049/jimmunol.179.8.5425] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal epithelial cells (IECs) are a first line of defense against microbial pathogens that enter the host through the intestinal tract. Moreover, viral pathogens that infect the host via the intestinal epithelium are an important cause of morbidity and mortality. However, the mechanisms by which viral pathogens activate antiviral defense mechanisms in IECs are largely unknown. The synthetic dsRNA analog polyinosinic-polycytidylic acid and infection with live virus were used to probe the molecules that are activated and the mechanisms of signaling in virus-infected human IECs. Polyinosinic-polycytidylic acid activated IFN regulatory factor 3 dimerization and phosphorylation, increased activity of the IFN-stimulated response element, induced a significant increase in IFN-beta mRNA transcripts and IFN-beta secretion, and up-regulated the expression of IFN-regulated genes in IECs. Those responses were dependent upon activation of the dsRNA binding protein retinoic acid inducible gene I (RIG-I) and the RIG-I interacting protein IFN promoter stimulator-1, but not on dsRNA-activated protein kinase or TLR3, which also were expressed by IECs. Virus replication and virus-induced cell death increased in IECs in which RIG-I was silenced, consistent with the importance of the RIG-I signaling pathway in IEC antiviral innate immune defense mechanisms.
Collapse
Affiliation(s)
- Yoshihiro Hirata
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
129
|
Yamazaki K, Murray JA, Kita H. Innate immunomodulatory effects of cereal grains through induction of IL-10. J Allergy Clin Immunol 2007; 121:172-178.e3. [PMID: 17919702 DOI: 10.1016/j.jaci.2007.08.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 08/08/2007] [Accepted: 08/08/2007] [Indexed: 02/02/2023]
Abstract
BACKGROUND Interactions between the gastrointestinal immune system and the luminal environment play critical roles in maintaining immune homeostasis and in diseases such as inflammatory bowel disease. Although immunomodulation by microbial factors has been studied extensively, little attention has been given to the potential immunomodulatory effects of ingested foods. OBJECTIVE We characterized the effects of cereal grains on the immune response in human subjects and investigated the mechanism. METHODS PBMCs from healthy individuals were incubated with cereal grain extracts, and cytokine levels in cell-free supernatants were measured. The cellular source of IL-10 and the role of monocytes were investigated by means of flow cytometry and cell-depletion/reconstitution experiments. RESULTS Extracts of cereal grains, including rice and wheat, induced marked IL-10 production from PBMCs. Intracellular cytokine staining and cell-depletion experiments showed that CD14+ monocytes produced IL-10. Importantly, when PBMCs were stimulated with concanavalin A, cereal grains concentration-dependently inhibited their production of IL-5, IL-13, and IFN-gamma; neutralizing IL-10 or removing the monocytes abrogated this inhibitory effect. This cereal grain-induced IL-10 response was polymyxin B sensitive, heat resistant, and inhibited by blocking the Toll-like receptor 4. CONCLUSION Cereal grains have strong innate immunomodulatory effects by inducing marked production of IL-10 from CD14+ monocytes in vitro. LPS or LPS-mimicking activity in cereal grains might be responsible. The potential immunomodulatory effects of cereal grains need further study in vivo.
Collapse
Affiliation(s)
- Kiyoshi Yamazaki
- Division of Allergic Diseases, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | |
Collapse
|
130
|
Ebensen T, Link C, Riese P, Schulze K, Morr M, Guzmán CA. A pegylated derivative of alpha-galactosylceramide exhibits improved biological properties. THE JOURNAL OF IMMUNOLOGY 2007; 179:2065-73. [PMID: 17675464 DOI: 10.4049/jimmunol.179.4.2065] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The glycolipid alpha-galactosylceramide (alphaGalCer) has immunomodulatory properties, which have been exploited to combat cancer, chronic inflammatory diseases, and infections. However, its poor solubility makes alphaGalCer a suboptimal compound for in vivo applications. In this study, a pegylated derivative of alphaGalCer is characterized, which exhibits improved physical and biological properties. The new compound, alphaGalCerMPEG, is water-soluble and retains the specificity for the CD1d receptor of alphaGalCer. The in vitro stimulatory properties on immune cells (e.g., dendritic cells and splenocytes) are maintained intact, even when tested at a 33-fold lower concentration of the active moiety than alphaGalCer. NK cells isolated from mice treated with alphaGalCerMPEG also had stronger cytotoxic activity on YAC-1 cells than those obtained from animals receiving either alphaGalCer or CpG. Intranasal immunization studies performed in mice showed that alphaGalCerMPEG exerts stronger adjuvant activities than the parental compound alphaGalCer when tested at 0.35 vs 11.7 nM/dose. Coadministration of beta-galactosidase with alphaGalCerMPEG resulted not only in high titers of Ag-specific Abs in serum (i.e., 1:512,000), but also in the stimulation of stronger Th2 and secretory IgA responses, both at local and remote mucosal effector sites (i.e., nose, lung, and vagina). The new synthetic derivative alphaGalCerMPEG represents a promising tool for the development of immune interventions against infectious and noninfectious diseases.
Collapse
Affiliation(s)
- Thomas Ebensen
- Department of Vaccinology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | | | | | | |
Collapse
|
131
|
Vélez MP, De Keersmaecker SCJ, Vanderleyden J. Adherence factors of Lactobacillus in the human gastrointestinal tract. FEMS Microbiol Lett 2007; 276:140-8. [PMID: 17888009 DOI: 10.1111/j.1574-6968.2007.00908.x] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Despite the increasing number of scientific reports describing adhesion of Lactobacillus to components of the human intestinal mucosa, information on the surface molecules mediating this adhesion and their corresponding receptors is fragmentary. This MiniReview compiles present knowledge of the genetically and functionally characterized Lactobacillus factors responsible for mediating adhesion to different components of the human gastrointestinal tract. In addition, for the proteins among these factors, the domain structure is discussed, and where appropriate the results of in silico analyses are reported.
Collapse
|
132
|
He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J, Shan M, Chadburn A, Villanacci V, Plebani A, Knowles DM, Rescigno M, Cerutti A. Intestinal bacteria trigger T cell-independent immunoglobulin A(2) class switching by inducing epithelial-cell secretion of the cytokine APRIL. Immunity 2007; 26:812-26. [PMID: 17570691 DOI: 10.1016/j.immuni.2007.04.014] [Citation(s) in RCA: 571] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2006] [Revised: 04/02/2007] [Accepted: 04/25/2007] [Indexed: 12/27/2022]
Abstract
Bacteria colonize the intestine shortly after birth and thereafter exert several beneficial functions, including induction of protective immunoglobulin A (IgA) antibodies. The distal intestine contains IgA(2), which is more resistant to bacterial proteases than is IgA(1). The mechanism by which B cells switch from IgM to IgA(2) remains unknown. We found that human intestinal epithelial cells (IECs) triggered IgA(2) class switching in B cells, including IgA(1)-expressing B cells arriving from mucosal follicles, through a CD4(+) T cell-independent pathway involving a proliferation-inducing ligand (APRIL). IECs released APRIL after sensing bacteria through Toll-like receptors (TLRs) and further increased APRIL production by activating dendritic cells via thymic stromal lymphopoietin. Our data indicate that bacteria elicit IgA(2) class switching by linking lamina propria B cells with IECs through a TLR-inducible signaling program requiring APRIL. Thus, mucosal vaccines should activate IECs to induce more effective IgA(2) responses.
Collapse
Affiliation(s)
- Bing He
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Borges O, Tavares J, de Sousa A, Borchard G, Junginger HE, Cordeiro-da-Silva A. Evaluation of the immune response following a short oral vaccination schedule with hepatitis B antigen encapsulated into alginate-coated chitosan nanoparticles. Eur J Pharm Sci 2007; 32:278-90. [PMID: 17884394 DOI: 10.1016/j.ejps.2007.08.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 07/09/2007] [Accepted: 08/06/2007] [Indexed: 11/19/2022]
Abstract
The purpose of this work was to assess the ability of recombinant hepatitis B vaccine, encapsulated in alginate-coated chitosan nanoparticles, to induce local and systemic immune responses following oral vaccination. The antigen was administered either alone or in combination with the immunopotentiator, synthetic oligodeoxynucleotide containing immunostimulatory CpG motif (CpG ODN) as adjuvant, and associated or not with the alginate-coated chitosan nanoparticles. After two immunizations the group I (HBsAg associated with nanoparticles) and the group VI (HBsAg and CpG, both associated with nanoparticles) showed enhanced immune responses. Both groups showed significant higher values of the CD69 expression in CD4+ and CD8+ T-lymphocytes and lower values of this marker in B lymphocytes. Moreover, a strongest proliferative response of the splenocytes, ex vivo stimulated with concanavalin A, was observed in the same groups. Although with a presence of non-responder mice within the groups, only mice of the groups I and VI elicited the generation of anti-HBsAg antibodies detected in serum (IgG) and in the intestinal washings (sIgA). The results demonstrated that coated chitosan nanoparticles might have potential for being used as a deliver system for oral vaccination with the recombinant hepatitis B surface antigen.
Collapse
Affiliation(s)
- Olga Borges
- Center for Pharmaceutical Studies, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Rua do Norte, 3000-295 Coimbra, Portugal.
| | | | | | | | | | | |
Collapse
|
134
|
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
|
135
|
Sato M, Adachi T, Tsubata T. Augmentation of signaling through BCR containing IgE but not that containing IgA due to lack of CD22-mediated signal regulation. THE JOURNAL OF IMMUNOLOGY 2007; 178:2901-7. [PMID: 17312134 DOI: 10.4049/jimmunol.178.5.2901] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The B cell membrane molecules CD22 and CD72 contain ITIMs in their cytoplasmic portion, and negatively regulate signaling through BCR. Various lines of evidence suggest that ligation of BCR containing IgG (IgG-BCR) transmits augmented signaling due to lack of CD22-mediated signal regulation. However, the signaling capacities of BCR containing IgA and IgE remain largely undefined. In this study, we demonstrate that both IgE-BCR and IgG-BCR, but not IgA-BCR, transmit augmented signaling compared with IgM-BCR. Ligation of IgE-BCR does not induce signaling events required for CD22-mediated signal inhibition, and restoration of these signaling events by coligation of CD22 with BCR abrogates signal augmentation. Furthermore, the cytoplasmic portion of IgE but not that of IgA is sufficient for suppressing CD22-mediated signal inhibition. These findings strongly suggest that the cytoplasmic portion of IgE but not that of IgA reverses CD22-mediated signal inhibition, leading to augmentation of signaling through IgE-BCR but not IgA-BCR. Augmented IgE-BCR signaling appears to play a role in production of large amounts of IgE during helminth infection, whereas regulated signaling through IgA-BCR may be crucial for constitutive production of IgA for mucosal immunity.
Collapse
Affiliation(s)
- Motohiko Sato
- Laboratory of Immunology, School of Biomedical Science, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | | |
Collapse
|
136
|
Roy N, Barnett M, Knoch B, Dommels Y, McNabb W. Nutrigenomics applied to an animal model of Inflammatory Bowel Diseases: transcriptomic analysis of the effects of eicosapentaenoic acid- and arachidonic acid-enriched diets. Mutat Res 2007; 622:103-16. [PMID: 17574631 DOI: 10.1016/j.mrfmmm.2007.04.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 03/30/2007] [Accepted: 04/12/2007] [Indexed: 01/07/2023]
Abstract
In vivo models of Inflammatory Bowel Diseases (IBD) elucidate important mechanisms of chronic inflammation. Complex intestinal responses to food components create a unique "fingerprint" discriminating health from disease. Five-week-old IL10(-/-) and C57BL/6J (C57; control) mice were inoculated orally with complex intestinal microflora (CIF) and/or pure cultures of Enterococcus faecalis and E. faecalis (EF) aiming for more consistent inflammation of the intestinal mucosa. Inoculation treatments were compared to non-inoculated IL10(-/-) and C57 mice, either kept in specific pathogen free (SPF) or conventional conditions (2x5 factorial design). At 12 weeks of age, mice were sacrificed for intestinal histological (HIS) and transcriptomic analysis using limma and Ingenuity Pathway Analysis Software. Colonic HIS was significantly affected (P<0.05) in inoculated IL10(-/-) mice and accounted for approximately 60% of total intestinal HIS. Inoculation showed a strong effect on colonic gene expression, with more than 2000 genes differentially expressed in EF.CIF-inoculated IL10(-/-) mice. Immune response gene expression was altered (P<0.05) in these mice. The second study investigated the effect of arachidonic (AA) and eicosapentaenoic acid (EPA) on colonic HIS and gene expression to test whether EPA, contrary to AA, diminished intestinal inflammation in EF.CIF IL10(-/-) mice (2 x 4 factorial design). AIN-76A (5% corn oil) and AIN-76A (fat-free) +1% corn oil supplemented with either 3.7% oleic acid (OA), AA or EPA were used. IL10(-/-) mice fed EPA- and AA-enriched diets had at least 40% lower colonic HIS (P<0.05) than those fed control diets (AIN-76A and OA diets). The expression of immune response and 'inflammatory disease' genes (down-regulated: TNFalpha, IL6, S100A8, FGF7, PTGS2; up-regulated: PPARalpha, MGLL, MYLK, PPSS23, ABCB4 with EPA and/or AA) was affected in IL10(-/-) mice fed EPA- and AA-enriched diets, compared to those fed AIN-76A diet.
Collapse
Affiliation(s)
- Nicole Roy
- Food, Metabolism & Microbiology Section, Food & Health Group, AgResearch Grasslands, Palmerston North, New Zealand.
| | | | | | | | | |
Collapse
|
137
|
Bonner A, Perrier C, Corthésy B, Perkins SJ. Solution structure of human secretory component and implications for biological function. J Biol Chem 2007; 282:16969-80. [PMID: 17428798 DOI: 10.1074/jbc.m701281200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Secretory component (SC) in association with polymeric IgA (pIgA) forms secretory IgA, the major antibody active at mucosal surfaces. SC also exists in the free form, with innate-like neutralizing properties against pathogens. Free SC consists of five glycosylated variable (V)-type Ig domains (D1-D5), whose structure was determined by x-ray and neutron scattering, ultracentrifugation, and modeling. With a radius of gyration of 3.53-3.63 nm, a length of 12.5 nm, and a sedimentation coefficient of 4.0 S, SC possesses an unexpected compact structure. Constrained scattering modeling based on up to 13,000 trial models shows that SC adopts a J-shaped structure in which D4 and D5 are folded back against D2 and D3. The seven glycosylation sites are located on one side of SC, leaving known IgA-binding motifs free to interact with pIgA. This work represents the first analysis of the three-dimensional structure of full-length free SC and paves the way to a better understanding of the association between SC and its potential ligands, i.e. pIgA and pathogenic-associated motifs.
Collapse
Affiliation(s)
- Alexandra Bonner
- Department of Biochemistry and Molecular Biology, University College London, Darwin Building, Gower Street, London WC1E 6BT, United Kingdom
| | | | | | | |
Collapse
|
138
|
Corthésy B. Roundtrip ticket for secretory IgA: role in mucosal homeostasis? THE JOURNAL OF IMMUNOLOGY 2007; 178:27-32. [PMID: 17182536 DOI: 10.4049/jimmunol.178.1.27] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
An important activity of mucosal surfaces is the production of Ab referred to as secretory IgA (SIgA). SIgA serves as the first line of defense against microorganisms through a mechanism called immune exclusion. In addition, SIgA adheres selectively to M cells in intestinal Peyer's patches, thus mediating the transepithelial transport of the Ab molecule from the intestinal lumen to underlying gut-associated organized lymphoid tissue. In Peyer's patches, SIgA binds and is internalized by dendritic cells in the subepithelial dome region. When used as carrier for Ags in oral immunization, SIgA induces mucosal and systemic responses associated with production of anti-inflammatory cytokines and limits activation of dendritic cells. In terms of humoral immunity at mucosal surfaces, SIgA appears thus to combine properties of a neutralizing agent (immune exclusion) and of a mucosal immunopotentiator inducing effector immune responses in a noninflammatory context favorable to preserve local homeostasis of the gastrointestinal tract.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory of the Division of Immunology and Allergy, State University Hospital (Centré Hospitalìer Universitaire Vandois), Rue du Bugnon, BH 19-650, 1011 Lausanne, Switzerland.
| |
Collapse
|
139
|
Zaph C, Troy AE, Taylor BC, Berman-Booty LD, Guild KJ, Du Y, Yost EA, Gruber AD, May MJ, Greten FR, Eckmann L, Karin M, Artis D. Epithelial-cell-intrinsic IKK-beta expression regulates intestinal immune homeostasis. Nature 2007; 446:552-6. [PMID: 17322906 DOI: 10.1038/nature05590] [Citation(s) in RCA: 419] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 01/11/2007] [Indexed: 12/30/2022]
Abstract
Intestinal epithelial cells (IECs) provide a primary physical barrier against commensal and pathogenic microorganisms in the gastrointestinal (GI) tract, but the influence of IECs on the development and regulation of immunity to infection is unknown. Here we show that IEC-intrinsic IkappaB kinase (IKK)-beta-dependent gene expression is a critical regulator of responses of dendritic cells and CD4+ T cells in the GI tract. Mice with an IEC-specific deletion of IKK-beta show a reduced expression of the epithelial-cell-restricted cytokine thymic stromal lymphopoietin in the intestine and, after infection with the gut-dwelling parasite Trichuris, fail to develop a pathogen-specific CD4+ T helper type 2 (T(H)2) response and are unable to eradicate infection. Further, these animals show exacerbated production of dendritic-cell-derived interleukin-12/23p40 and tumour necrosis factor-alpha, increased levels of CD4+ T-cell-derived interferon-gamma and interleukin-17, and develop severe intestinal inflammation. Blockade of proinflammatory cytokines during Trichuris infection ablates the requirement for IKK-beta in IECs to promote CD4+ T(H)2 cell-dependent immunity, identifying an essential function for IECs in tissue-specific conditioning of dendritic cells and limiting type 1 cytokine production in the GI tract. These results indicate that the balance of IKK-beta-dependent gene expression in the intestinal epithelium is crucial in intestinal immune homeostasis by promoting mucosal immunity and limiting chronic inflammation.
Collapse
Affiliation(s)
- Colby Zaph
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Oliveira FM, Dos Santos EM, Alves AC, Campana-Pereira MA, Ramaldes GA, Cardoso VN, Ruiz-de-Souza V, Gontijo CM. Digestion, Absorption and Tissue Distribution of Ovalbumin and Palmitoyl-ovalbumin: Impact on Immune Responses Triggered by Orally Administered Antigens. Scand J Immunol 2007; 65:139-47. [PMID: 17257218 DOI: 10.1111/j.1365-3083.2006.01884.x] [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] [Indexed: 11/26/2022]
Abstract
Previous work in this laboratory has demonstrated that ovalbumin coupled to palmitoyl residues (palmitoyl-Ova) does not induce oral tolerance. The present study sought to determine whether this coupling affects digestion, absorption and transfer of antigen. Ova and palmitoyl-Ova were shown to be digested differently in vitro by proteolytic enzymes and presented different tissue distribution kinetics after being labelled with (99m)technetium and orally administered to animals. Palmitoyl-Ova remained longer in the stomach, while native Ova was quickly transferred to the gut and other organs. After 3 h, higher levels of palmitoyl-Ova were found in the blood, Peyer's patches, mesenteric lymph nodes, liver and, especially, the spleen, which appears to be essential for immunization with palmitoyl-Ova. In fact, splenectomized mice treated orally with palmitoyl-Ova became tolerant, while tolerance to Ova was not affected. Thus, palmitoyl coupling was demonstrated to affect antigen digestion, absorption and transport. This is the first time that the spleen has been shown to be required for oral immunization with palmitoyl-Ova.
Collapse
Affiliation(s)
- F M Oliveira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, UFMG, Belo Horizonte, MG, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
141
|
Sjögren YM, Duchén K, Lindh F, Björkstén B, Sverremark-Ekström E. Neutral oligosaccharides in colostrum in relation to maternal allergy and allergy development in children up to 18 months of age. Pediatr Allergy Immunol 2007; 18:20-6. [PMID: 17295795 DOI: 10.1111/j.1399-3038.2006.00486.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Several recent studies have demonstrated a relationship between the composition of the gut microbiota in infancy and subsequent development of allergic disease. Human milk is the major food in infancy and may thus profoundly influence the composition of the gut flora. Oligosaccharides in breast milk survive the passage through the stomach and are utilized by the gut microbiota. As the relationship between breast feeding and childhood allergy is controversial we hypothesized that the composition of oligosaccharides in breast milk might explain the controversy. Nine of the most abundant neutral oligosaccharides in human milk were analysed in colostrum samples from allergic and non-allergic women and related to subsequent development of allergy in their children. The carbohydrate fraction of the colostrum was separated by gel permeation chromatography and neutral oligosaccharides, tri- to hexasaccharides were collected. Neutral oligosaccharides were analysed with high-performance liquid chromatography. There was a large variation in the concentration of neutral oligosaccharides in colostrum, which could not be explained by the allergic status of the women. Allergic children consumed higher amounts of neutral oligosaccharides in total, although not significantly (p = 0.12). When different oligosaccharides were analysed separately, there was no significant difference in consumption between the infants who developed atopic allergy later (n = 9) and infants who did not (n = 11). Thus, the amount of neutral oligosaccharides in colostrum does not directly correlate with maternal allergy, nor with allergy development in children up to 18 months of age.
Collapse
Affiliation(s)
- Ylva Margareta Sjögren
- Department of Immunology, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
142
|
Xu W, He B, Chiu A, Chadburn A, Shan M, Buldys M, Ding A, Knowles DM, Santini PA, Cerutti A. Epithelial cells trigger frontline immunoglobulin class switching through a pathway regulated by the inhibitor SLPI. Nat Immunol 2007; 8:294-303. [PMID: 17259987 DOI: 10.1038/ni1434] [Citation(s) in RCA: 232] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Accepted: 12/21/2006] [Indexed: 12/24/2022]
Abstract
Epithelial cells (ECs) transport class-switched immunoglobulin G (IgG) and IgA antibodies across mucous membranes. Whether ECs initiate class switching remains unknown. Here we found that ECs lining tonsillar crypts formed pockets populated by B cells expressing activation-induced cytidine deaminase (AID), an enzyme associated with ongoing class switching. ECs released B cell-activating AID-inducing factors after sensing microbial products through Toll-like receptors. The resulting class switching was amplified by thymic stromal lymphopoietin, an epithelial interleukin 7-like cytokine that enhanced the B cell 'licensing' function of dendritic cells, and was restrained by secretory leukocyte protease inhibitor, an epithelial homeostatic protein that inhibited AID induction in B cells. Thus, ECs may function as mucosal 'guardians' orchestrating frontline IgG and IgA class switching through a Toll-like receptor-inducible signaling program regulated by secretory leukocyte protease inhibitor.NOTE: In the version of this article initially published online, the middle label above Figure 6c is incorrect. The correct label should be 'BAFF'. The error has been corrected for all versions of the article.
Collapse
Affiliation(s)
- Weifeng Xu
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Matsumura T, Fujinaga Y, Jin Y, Kabumoto Y, Oguma K. Human milk SIgA binds to botulinum type B 16S toxin and limits toxin adherence on T84 cells. Biochem Biophys Res Commun 2007; 352:867-72. [PMID: 17156748 DOI: 10.1016/j.bbrc.2006.11.095] [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: 11/15/2006] [Accepted: 11/17/2006] [Indexed: 12/21/2022]
Abstract
Botulinum neurotoxin produced by Clostridium botulinum type B is in the form of a complex of 12S and 16S toxins. Food-borne botulism is caused by these complex toxins which are ingested orally and absorbed from the digestive tract. Here, we show that the human milk SIgA binds to the type B16S toxin. The binding of SIgA to 16S toxin and HA was inhibited by carbohydrates such as galactose, suggesting that the interaction of carbohydrate side chain of the SIgA with the HA of the 16S toxin is important for SIgA-16S complex formation. We also demonstrate that SIgA inhibits the attachment of 16S toxin to intestinal epithelial cells. These data suggest that the interaction of antigen nonspecific SIgA with 16S toxin has a large influence on the absorption of 16S toxin from the intestinal epithelium, and that SIgA may provide insight into developing a therapeutic agent for type B food-borne botulism.
Collapse
Affiliation(s)
- Takuhiro Matsumura
- Laboratory for Infection Cell Biology, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Yamada-oka 3-1, Suita, Osaka 565-0871, Japan
| | | | | | | | | |
Collapse
|
144
|
Abstract
The neonatal adaptive immune system, relatively naïve to foreign antigens, requires synergy with the innate immune system to protect the intestine. Goblet cells provide mucins, Paneth cells produce antimicrobial peptides, and dendritic cells (DCs) present luminal antigens. Intracellular signaling by Toll-like receptors (TLRs) elicits chemokines and cytokines that modulate inflammation. Enteric neurons and lymphocytes provide paracrine and endocrine signaling. However, full protection requires human milk. Breast-feeding reduces enteric infection and may reduce chronic disease in later life. Although human milk contains significant secretory immunoglobulin A (sIgA), most of its protective factors are constitutively expressed. Multifunctional milk components are nutrients whose partial digestion products inhibit pathogens. Cytokines, cytokine receptors, TLR agonists and antagonists, hormones, anti-inflammatory agents, and nucleotides in milk modulate inflammation. Human milk is rich in glycans (complex carbohydrates): As prebiotics, indigestible glycans stimulate colonization by probiotic organisms, modulating mucosal immunity and protecting against pathogens. Through structural homology to intestinal cell surface receptors, glycans inhibit pathogen binding, the essential first step of pathogenesis. Bioactive milk components comprise an innate immune system of human milk whereby the mother protects her nursing infant. Interactions between human milk glycans, intestinal microflora, and intestinal mucosa surface glycans underlie ontogeny of innate mucosal immunity, pathobiology of enteric infection, and inflammatory bowel diseases.
Collapse
Affiliation(s)
- David S Newburg
- Pediatric Gastroenterology and Nutrition Unit, MassGeneral Hospital for Children and Harvard Medical School, Boston, Massachusetts, USA.
| | | |
Collapse
|
145
|
Iweala OI, Nagler CR. Immune privilege in the gut: the establishment and maintenance of non-responsiveness to dietary antigens and commensal flora. Immunol Rev 2006; 213:82-100. [PMID: 16972898 DOI: 10.1111/j.1600-065x.2006.00431.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Immune privilege in the gut is the result of a complex interplay between the gut microbiome, gut luminal antigens, and the intestinal epithelial barrier. Composed of both physical and immunochemical components, the intestinal barrier secretes immunoregulatory mediators that promote the generation of tolerogenic antigen-presenting cells, phagocytic innate immune cells characterized by 'inflammatory anergy', and regulatory cells of the adaptive immune system. Innate immune cells mediate controlled transepithelial transport of luminal antigens as far as the mesenteric lymph nodes, where the intestinal and peripheral immune systems intersect. This promotes the generation of adaptive regulatory lymphocytes that actively suppress effector cell responses against gut luminal antigens and flora. The net result is the generation of tolerance to dietary antigens and the maintenance of gut homeostasis. Dysregulation of this complex immunoregulatory network leads to diseases such as food allergy and inflammatory bowel disease. Future therapies for these diseases will likely involve the functional restoration of the barrier and regulatory cell functions at the epithelial/luminal interface.
Collapse
Affiliation(s)
- Onyinye I Iweala
- Mucosal Immunology Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-4404, USA
| | | |
Collapse
|
146
|
Mazmanian SK, Kasper DL. The love-hate relationship between bacterial polysaccharides and the host immune system. Nat Rev Immunol 2006; 6:849-58. [PMID: 17024229 DOI: 10.1038/nri1956] [Citation(s) in RCA: 250] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This article explores the fascinating relationship between the mammalian immune system and the bacteria that are present in the mammalian gut. Every human is an ecosystem that hosts 10(13)-10(14) bacteria. We review the evidence that immunomodulatory molecules produced by commensal bacteria in the gut have a beneficial influence on the development of certain immune responses, through eliciting the clonal expansion of CD4(+) T-cell populations. This process seems to contribute to the overall health of the host by offering protection against various diseases and might provide supporting evidence at a molecular level for the 'hygiene hypothesis' of allergic immune disorders.
Collapse
Affiliation(s)
- Sarkis K Mazmanian
- Division of Biology, California Institute of Technology, Mail code 156-29, 1200 East California Boulevard, Pasadena, California 91125, USA
| | | |
Collapse
|
147
|
Zaph C, Rook KA, Goldschmidt M, Mohrs M, Scott P, Artis D. Persistence and function of central and effector memory CD4+ T cells following infection with a gastrointestinal helminth. THE JOURNAL OF IMMUNOLOGY 2006; 177:511-8. [PMID: 16785548 PMCID: PMC1805702 DOI: 10.4049/jimmunol.177.1.511] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunity in the gastrointestinal tract is important for resistance to many pathogens, but the memory T cells that mediate such immunity are poorly characterized. In this study, we show that following sterile cure of a primary infection with the gastrointestinal parasite Trichuris muris, memory CD4+ T cells persist in the draining mesenteric lymph node and protect mice against reinfection. The memory CD4+ T cells that developed were a heterogeneous population, consisting of both CD62L(high) central memory T cells (T(CM)) and CD62L(low) effector memory T cells (T(EM)) that were competent to produce the Th type 2 effector cytokine, IL-4. Unlike memory T cells that develop following exposure to several other pathogens, both CD4+ T(CM) and T(EM) populations persisted in the absence of chronic infection, and, critically, both populations were able to transfer protective immunity to naive recipients. CD62L(high)CD4+ T(CM) were not apparent early after infection, but emerged following clearance of primary infection, suggesting that they may be derived from CD4+ T(EM). Consistent with this theory, transfer of CD62L(low)CD4+ T(EM) into naive recipients resulted in the development of a population of protective CD62L(high)CD4+ T(CM). Taken together, these studies show that distinct subsets of memory CD4+ T cells develop after infection with Trichuris, persist in the GALT, and mediate protective immunity to rechallenge.
Collapse
Affiliation(s)
- Colby Zaph
- Department of Pathobiology, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
148
|
Salazar-Gonzalez RM, Niess JH, Zammit DJ, Ravindran R, Srinivasan A, Maxwell JR, Stoklasek T, Yadav R, Williams IR, Gu X, McCormick BA, Pazos MA, Vella AT, Lefrancois L, Reinecker HC, McSorley SJ. CCR6-mediated dendritic cell activation of pathogen-specific T cells in Peyer's patches. Immunity 2006; 24:623-32. [PMID: 16713979 PMCID: PMC2855652 DOI: 10.1016/j.immuni.2006.02.015] [Citation(s) in RCA: 196] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 12/05/2005] [Accepted: 02/24/2006] [Indexed: 12/30/2022]
Abstract
T cell activation by dendritic cells (DCs) is critical to the initiation of adaptive immune responses and protection against pathogens. Here, we demonstrate that a specialized DC subset in Peyer's patches (PPs) mediates the rapid activation of pathogen specific T cells. This DC subset is characterized by the expression of the chemokine receptor CCR6 and is found only in PPs. CCR6(+) DCs were recruited into the dome regions of PPs upon invasion of the follicle associated epithelium (FAE) by an enteric pathogen and were responsible for the rapid local activation of pathogen-specific T cells. CCR6-deficient DCs were unable to respond to bacterial invasion of PPs and failed to initiate T cell activation, resulting in reduced defense against oral infection. Thus, CCR6-dependent regulation of DCs is responsible for localized T cell dependent defense against entero-invasive pathogens.
Collapse
Affiliation(s)
- Rosa Maria Salazar-Gonzalez
- Department of Medicine, GI Division and Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, McGuire Translational Research Facility, TRF DC 2873, 2001 6th Street S.E., Minneapolis, Minnesota 55455
| | - Jan H. Niess
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114
| | - David J. Zammit
- Division of Immunology, Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030
| | - Rajesh Ravindran
- Department of Medicine, GI Division and Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, McGuire Translational Research Facility, TRF DC 2873, 2001 6th Street S.E., Minneapolis, Minnesota 55455
| | - Aparna Srinivasan
- Department of Medicine, GI Division and Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, McGuire Translational Research Facility, TRF DC 2873, 2001 6th Street S.E., Minneapolis, Minnesota 55455
| | - Joseph R. Maxwell
- Division of Immunology, Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030
| | - Thomas Stoklasek
- Division of Immunology, Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030
| | - Rajwardhan Yadav
- Division of Immunology, Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030
| | - Ifor R. Williams
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Whitehead 105D, 615 Michael Street, Atlanta, Georgia 30322
| | - Xiubin Gu
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114
| | - Beth A. McCormick
- Department of Pediatric Gastroenterology, Center for the Study of Inflammatory Bowel Diseases, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114
| | - Michael A. Pazos
- Department of Pediatric Gastroenterology, Center for the Study of Inflammatory Bowel Diseases, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114
| | - Anthony T. Vella
- Division of Immunology, Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030
| | - Leo Lefrancois
- Division of Immunology, Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030
| | - Hans-Christian Reinecker
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114
| | - Stephen J. McSorley
- Department of Medicine, GI Division and Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, McGuire Translational Research Facility, TRF DC 2873, 2001 6th Street S.E., Minneapolis, Minnesota 55455
- Correspondence:
| |
Collapse
|
149
|
Abstract
PURPOSE OF REVIEW Intestinal dendritic cells have emerged as key regulators of immunity to pathogens, oral tolerance and intestinal inflammation. Studies have begun to elucidate the regulatory mechanisms responsible for defining region- and compartment-specific phenotypes and functions of dendritic cells in mucosal tissues. RECENT FINDINGS Specific subsets of dendritic cells appear to be associated with the various routes for antigen acquisition in the intestine. The constant sampling of intestinal antigenic content ensures establishment of tolerance to commensal bacteria and food antigens. Tolerance development to oral antigens is restricted to the mucosal immune system. Other advances have provided insight into the molecular basis of microbial recognition and innate immune responses by intestinal dendritic cells. Differences in the involvement of dendritic cells have begun to emerge in Crohn's disease and ulcerative colitis and link gene regulation in dendritic cells to therapeutic responses. SUMMARY A major focus of mucosal immunology will be to understand how diverse dendritic cell subsets cooperate in regulating homeostasis and host defense in the different intestinal immune compartments. This will be pivotal to understanding how the mucosal immune system makes the distinction between commensal microbiota, pathogens and self antigens.
Collapse
Affiliation(s)
- Jan H Niess
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
150
|
Abstract
Mucosal dendritic cells (DCs) constantly survey the luminal microenvironment which contains commensal microbiota and potentially harmful organisms regulating pathogen recognition and adaptive as well as innate defense activation. Distinct mechanisms are beginning to emerge by which intestinal antigen sampling and handling is achieved ensuring specificity and contributing to redundancy in pathogen detection. Distinct DC subsets are associated with these mechanisms and regulate specific innate or adaptive immune responses to help distinguish between commensal microbiota, pathogens and self antigens. Understanding DC biology in the mucosal immune system may contribute to the unraveling of infection routes of intestinal pathogens and may aid in developing novel vaccines and therapeutic strategies for the treatment of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Jan Hendrik Niess
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|