scholarly journals Ovalbumin-Protein σ1 M-Cell Targeting Facilitates Oral Tolerance With Reduction of Antigen-Specific CD4+ T Cells

2008 ◽  
Vol 135 (3) ◽  
pp. 917-925 ◽  
Author(s):  
Hideaki Suzuki ◽  
Shinichi Sekine ◽  
Kosuke Kataoka ◽  
David W. Pascual ◽  
Massimo Maddaloni ◽  
...  
2001 ◽  
Vol 120 (5) ◽  
pp. A321-A321
Author(s):  
A KHORUTS ◽  
K THORSTENSON
Keyword(s):  
T Cells ◽  

Blood ◽  
2015 ◽  
Vol 125 (15) ◽  
pp. 2418-2427 ◽  
Author(s):  
Xiaomei Wang ◽  
Jin Su ◽  
Alexandra Sherman ◽  
Geoffrey L. Rogers ◽  
Gongxian Liao ◽  
...  

Key Points Coadministering FIX orally and systemically induces tolerance via complex immune regulation, involving tolerogenic dendritic and T-cell subsets. Induced CD4+CD25−LAP+ regulatory T cells with increased IL-10 and TGF-β expression and CD4+CD25+ regulatory T cells suppress antibody formation against FIX.


2018 ◽  
Vol 16 (1) ◽  
pp. 41-51 ◽  
Author(s):  
Katherine S. Wetzel ◽  
Sarah T.C. Elliott ◽  
Ronald G. Collman

Pathogenic HIV-1 infection of humans and SIVmac infection of macaques are the result of zoonotic transfer of primate immunodeficiency viruses from their natural hosts into non-natural host species. Natural host infections do not result in pathogenesis despite high levels of virus replication, and evidence suggests that differences in anatomical location and specific subsets of CD4+ T cells infected may underlie distinct outcomes from infection. The coreceptor CCR5 has long been considered the sole pathway for SIV entry and the key determinant of CD4+ cell targeting, but it has also been known that natural hosts express exceedingly low levels of CCR5 despite maintaining high levels of virus replication. This review details emerging data indicating that in multiple natural host species, CCR5 is dispensable for SIV infection ex vivo and/or in vivo and, contrary to the established dogma, alternative coreceptors, particularly CXCR6, play a central role in infection and cell targeting. Infections of non-natural hosts, however, are characterized by CCR5-exclusive entry. These findings suggest that alternative coreceptor-mediated cell targeting in natural hosts, combined with low CCR5 expression, may direct the virus to distinct populations of cells that are dispensable for immune homeostasis, particularly extralymphoid and more differentiated CD4+ T cells. In contrast, CCR5-mediated entry in non-natural hosts results in targeting of CD4+ T cells that are located in lymphoid tissues, critical for immune homeostasis, or necessary for gut barrier integrity. Thus, fundamental differences in viral entry coreceptor use may be central determinants of infection outcome. These findings redefine the normal SIV/host relationship in natural host species, shed new light on key features linked to zoonotic immunodeficiency virus transfer, and highlight important questions regarding how and why this coreceptor bottleneck occurs and the coevolutionary equilibrium is lost following cross-species transfer that results in AIDS.


2001 ◽  
Vol 120 (5) ◽  
pp. A321
Author(s):  
Alexander Khoruts ◽  
Kristen M. Thorstenson
Keyword(s):  
T Cells ◽  

Blood ◽  
2003 ◽  
Vol 102 (9) ◽  
pp. 3295-3301 ◽  
Author(s):  
Bertrand Dubois ◽  
Ludivine Chapat ◽  
Anne Goubier ◽  
Martine Papiernik ◽  
Jean-François Nicolas ◽  
...  

AbstractTo elucidate the role of CD4+CD25+ regulatory T cells in oral tolerance, we used the model of contact hypersensitivity (CHS) to 2,4-dinitrofluorobenzene (DNFB), which is mediated by CD8+ Tc1 effector cells independently of CD4+ T-cell help. Conversely to normal mice, invariant chain knock-out (KO) (Ii°/°) mice, which are deficient in CD4+ T cells, cannot be orally tolerized and develop a chronic hapten-specific CHS response. Transfer of naive CD4+ T cells before hapten gavage into Ii°/° mice restores oral tolerance by a mechanism independent of interleukin-10 (IL-10) production by CD4+ T cells. That naturally occurring CD4+CD25+ T cells are critical for oral tolerance induction is demonstrated by the finding that (1) transfer of CD4+CD25+ but not CD4+CD25– T cells into Ii°/° recipients completely prevents the CHS response and skin infiltration by CD8+ T cells, by blocking development of hapten-specific CD8+ T cells; (2) in vivo depletion of CD4+CD25+ cells by antibody treatment in normal mice impairs oral tolerance; and (3) CD4+CD25+ T cells inhibit hapten-specific CD8+ T-cell proliferation and interferon γ (IFNγ) production, in vitro. These data show that naturally occurring CD4+CD25+ T cells are instrumental for orally induced tolerance and are key actors for the control of antigen-specific CD8+ T-cell effectors mediating skin inflammation.


2016 ◽  
Vol 198 (3) ◽  
pp. 1202-1209 ◽  
Author(s):  
Chantal Kuhn ◽  
Rafael Machado Rezende ◽  
Hanane M’Hamdi ◽  
Andre Pires da Cunha ◽  
Howard L. Weiner

1996 ◽  
Vol 183 (6) ◽  
pp. 2605-2616 ◽  
Author(s):  
M F Neurath ◽  
I Fuss ◽  
B L Kelsall ◽  
D H Presky ◽  
W Waegell ◽  
...  

In previous studies we showed that a chronic colitis associated with a Th1 T cell response can be induced by the rectal administration of the haptenizing reagent 2,4,6-trinitrobenzene sulfonic acid (TNBS). We report here that oral administration of haptenized colonic proteins (HCP) before rectal administration of TNBS effectively suppresses the ability of the latter to induce colitis. This suppression (oral tolerance) appears to be due to the generation of mucosal T cells producing TGF-beta and Th2-type cytokines after oral HCP administration. Peyer's patch and lamina propria CD4+ T cells from HCP-fed animals stimulated with anti-CD3/anti-CD28 had a 5-10-fold increase in their production of TGF-beta and secreted increased amounts of IL-4 and IL-10 but lower levels of IFN-gamma in comparison to T cells from ovalbumin-fed control animals. In addition, the colons of HCP-fed mice showed strikingly increased TGF-beta but decreased IL-12 expression by immunohistochemical studies and isolated mononuclear cells from HCP-fed animals secreted less IL-12 heterodimer. Finally, and most importantly, the suppressive effect of orally administered HCP was abrogated by the concomitant systemic administration of anti-TGF-beta or rIL-12 suggesting a reciprocal relationship between IL-12 and TGF-beta on tolerance induction in TNBS-induced colitis. In parallel studies we demonstrated that TNBS-induced colitis can be transferred to naive recipient animals with purified CD4+ T cells from the colon of TNBS-treated animals and that such animals develop lethal pancolitis when exposed to very low doses of TNBS. Feeding of HCP suppressed this sensitivity to TNBS, indicating that oral feeding can suppress the response of pre-committed T cells in vivo. These studies suggest for the first time that TGF-beta production can abrogate experimental granulomatous colitis even after such colitis is established, and thus, that regulation of TGF-beta levels may have relevance to the treatment of human inflammatory bowel disease.


2001 ◽  
Vol 120 (5) ◽  
pp. A192-A192
Author(s):  
H TAKAISHI ◽  
T DENNING ◽  
K ITO ◽  
R MIFFLIN ◽  
P ERNST

Sign in / Sign up

Export Citation Format

Share Document