Advances in Animal and Veterinary Sciences

Download PDF Download ePUB
 Review Article

Review Article

Advances in Animal and Veterinary Sciences 1 (5): 129 – 133

Regulatory T Cells in Avian Species

Ramesh Kumar Selvaraj*, Revathi Shanmugasundaram

    Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, OH 44691. USA

*Corresponding author:selvaraj.7@osu.edu

ARTICLE CITATION: Selvaraj RK (2013). Regulatory T cells in avian species. Adv. Anim. Vet. Sci. 1 (5): 129 – 133.
Received: 2013–07–16, Revised: 2013–08–02, Accepted: 2013–08–03
The electronic version of this article is the complete one and can be found online at ( http://nexusacademicpublishers.com/table_contents_detail/4/85/html ) which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited

ABSTRACT

The immune system protects the host from foreign pathogens while avoiding damage towards self–antigens. T regulatory cells (Tregs), a subset of T cells, specialize in immune suppression. A host immune response is a result of interplay between different components of the immune system. Interplay between Tregs and other components of the immune system will determine whether the outcome will be a persistent infection or successful pathogen clearance. Avian Tregs are characterized by the presence of both CD4 and CD25. Avian CD4+ and CD25+ cells produce high amounts of IL–10 and lack IL–2 mRNA; and suppress T cell proliferation in vitro through both contact–dependent and independent pathways. Avian Treg properties and numbers are influenced by infections and inflammatory status of the bird. Compared to mammals, avian Treg research is still in early stages of reseearch and, thus, extensive characterization of avian Tregs is required. In mammals, Treg–targeted therapy is applied for numerous situations, e.g. infections, tumors, autoimmune diseases, sepsis, shock, and vaccine. Similar to mammals, avian diseases will benefit from Treg–targeted therapy.

INTRODUCTION

The immune system protects the host from foreign pathogens while avoiding damage towards self–antigens. T regulatory cells (Tregs), a subset of T cells, specialize in immune suppression. An adaptive immune response involves recruitment of effector (T and B) cells and Tregs. Activated immune cells, although essential for pathogen elimination, produce inflammatory cytokines and reactive oxygen species and can cause undesirable host damage (Belkaid and Rouse, 2005). Tregs protect the host from an excessive immune response and maintain self tolerance and mucosal tolerance (Workman et al., 2009). The balance between the effector cells and Tregs is important for optimal immune responses, the proper control of immune responses, and for establishing tolerance to self–antigens. Disruption in function of Tregs is a primary cause of autoimmune and inflammatory diseases. On the other hand, hyperactive Tregs can impair T cell, B cell, and other immune cell functions and, therefore, are implicated in impaired microbial defenses, pathogen persistence (Li et al., 2008), and impaired vaccine responses (Stober et al., 2005).

LACK OF UNIQUE MARKERS FOR TREGS

Among the different species in which Tregs have been characterized, human and mice Tregs have been extensively studied. Tregs constitutively express surface proteins like CD25, CD45, CTLA–4, HLA–DR, or GITR (Jonuleit and Schmitt, 2003), but these markers are not present in Tregs of all species or exclusive to Tregs in any particular species. Among the markers that can be defined to be unique to Tregs, the most commonly used marker is FoxP3 (Hori et al., 2003). FoxP3, a transcription factor, is essential for development and function of mammalian Tregs (Belkaid and Rouse, 2005). FoxP3 transcriptionally represses IL–2 and maintains suppressor functions of Tregs (Raimondi et al., 2007). Mutations in the FoxP3 gene cause autoimmune disease in scurfy mice (Brunkow et al., 2001) and such mice succumb to autoimmune pathology (Huter et al., 2008). Though absence or mutations in FoxP3 gene results in impaired Treg functions, presence of FoxP3 genes does not necessarily confer suppressive properties. Human T cells transiently express FoxP3, without expressing the suppressive properties or cytokines characteristics of Tregs (Gavin et al., 2006; Wang et al., 2007), though such T cells with transient FoxP3 expression with no suppressive properties are yet to be reported in other species. Our group reported that in chickens, CD4+CD25+ cells express suppressive properties even though in silico analysis failed to identify FoxP3 gene in the chicken genome (Selvaraj, 2013).

PROPERTIES OF TREGS

There are several different categories of suppressive cells, namely T regulatory–1 cells (Tr1) (Roncarolo et al., 2006), T helper 3 cells (Th3) (Carrier et al., 2007), CD8+FoxP3+ (Lu and Cantor, 2008), γδ T cells (Wildner et al., 1997; Wildner et al., 2004; Hoffmann et al., 2008), natural killer T cells (Smyth and Godfrey, 2000), and CD4–8–TCRαβ+ (Zhang et al., 2000). The hall mark characteristics of CD4+25+ Tregs that differentiate CD4+25+ Tregs from the above mentioned suppressive populations are:

  1. CD4+25+ Tregs originate as a separate lineage of cells in the thymus (Apostolou et al., 2002).

  2. CD4+25+ Tregs are anergic (meaning they don’t proliferate) in vitro (Thornton and Shevach, 1998).

  3. CD4+25+ Tregs in vitro anergy and suppressive properties are reversed by exogenous IL–2 (Thornton and Shevach, 1998).

  4. CD4+25+ Tregs express CTLA–4, LAG–3 and PD–1 (Yi et al., 2006).

  5. CD4+25+ Tregs produce high amounts of IL–10 (Dieckmann et al., 2001) and low amounts of IL–2 (Jonuleit et al., 2001; Takahashi et al., 1998)

TREGS DYSREGULATION DURING INFECTIONS

Tregs are a central player in immune suppression. Tregs are the only cells that can directly suppress every other component of the immune system. Tregs can suppress T effector cells, dendritic cells (Sakaguchi et al., 2008), B cells (Lim et al., 2005), macrophages ( Tiemessen et al., 2007), NK cell (Frimpong–Boateng et al., 2010), mast cells (Gri et al., 2008), neutrophils and eosinophils (Thorburn and Hansbro, 2010). Given this “universal” action of Tregs, it is reasonable to assume that though Treg activity could be beneficial to the host, Tregs simultaneously inhibit host immunity and cause persistent infections. Treg dysregulation during persistent viral infections has been reviewed elegantly (Belkaid, 2007; Li et al., 2008). During viral infections, T cell response leads to viral clearance. However, many viruses induce persistent infections despite continuous measurable T cell responses (Rehermann et al., 1996), a situation in which Tregs may be involved (Ward et al., 2007).

Tregs suppress the functions of CD4+ and CD8+ cells in the host and cause persistent infections of Friends virus (Robertson et al., 2006; Zelinskyy et al., 2006), Vaccinia virus (Haeryfar et al., 2005), Human Immunodeficiency virus (Epple et al., 2006; Nilsson et al., 2006), Hepatitis C virus (Boettler et al., 2005; Bolacchi et al., 2006), Hepatitis B virus (Stoop et al., 2007; Xu et al., 2006), Human T Lymphotropic virus (Yamano et al., 2005; Oh et al., 2006), Cytomegalovirus (Aandahl et al., 2004), and Feline Immunodeficiency virus (Mikkelsen et al., 2010). The influenza virus might have evolved to induce Tregs. Influenza–specific–Tregs suppress cytotoxic T lymphocytes by blocking CD8+ cell expansion. Tregs, stimulated with hemagglutinin antigen, expand more rapidly than CD8+ T cells and are highly suppressive in mice (Chappert et al., 2010). In humans, Treg numbers increase while CD4+ cell numbers, B–lymphocytes numbers, and macrophage IFNγ and TNFα production decrease post H1N1 infection (Giamarellos–Bourboulis et al., 2009). Tregs inhibit proliferation and IFNγ production of influenza–specific CD8 in the local environment (Lund et al., 2008; Khatri et al., 2010). Treg dysregulation is present during influenza A infection in mice (Haeryfar et al., 2005). In chickens, CD4+CD25+ (Tregs) numbers increased following H9N2 avian influenza virus infection, but the authors could not explain the upregulated roles of Tregs during viral infection (Teng et al., 2006). The above studies strongly suggest the involvement of Tregs in augmenting the pathogenesis of avian influenza infections. Treg research with mammals suggests that the increase in Tregs percentage post–influenza infection might be a local effect rather than a systemic effect (Lund et al., 2008; Khatri et al., 2010). Tregs reduce accumulation of macrophages in the lungs of influenza A virus–infected mice (Antunes and Kassiotis, 2010).

Tregs express several toll–like receptors (TLR), which recognize pathogen–associated molecular patterns (Caramalho et al., 2003). Tregs express TLR3, which recognizes double stranded RNA present during viral infections (Qian et al., 2007). TLR3–mediated activation amplifies the suppressive properties of Tregs (Qian et al., 2007). In addition, the host damage that occurs during infection and inflammation activates Tregs (Belkaid and Rouse, 2005). Some pathogens have evolved to selectively induce Tregs (Wilson et al., 2005; Lysaght et al., 2007) and thereby impede host immune responses. Treg–mediated suppression of host immune cells prevents an effective immune response against pathogens. IL–10 and TGFβ produced by Tregs suppress CD8+ effector cells against viral pathogens (Kinter et al., 2004). Tregs suppress IFNγ production by the host during an anti–viral response and thereby effectively impair the host defense against viral infections (Bolacchi et al., 2006). The role of Tregs in depressing a host immune response during viral infections has been confirmed by experiments that selectively target or deplete Tregs during viral infections.

ENHANCED ANTI–VIRAL IMMUNE RESPONSE FOLLOWING TREG DEPLETION/ABLATION

Depletion of Tregs using anti–CD25 neutralizing antibody relieves the in vivo suppression of an antiviral immune response and contributes to faster oncolytic viral clearance (Kottke et al., 2008). Depletion of Tregs enhances the activity of natural killer cells, activity of lymphokine–activated killer cells, and production of IFN (Kottke et al., 2008). Treg ablation enhances the virus–specific CD8+ T cell numbers and production of IFN in the spleen of infected animal (Zelinskyy et al., 2009). Treg depletion results in reactivation of virus–specific T cells in chronically infected mice (Dietze et al., 2011). In vitro, anti–IL–10 antibodies, which are expected to abrogate Treg functions (Sun et al., 2010), increase viral antigen–specific T cell proliferation (Landay et al., 1996).

TREGS OF AVIANS

Tregs have been extensively characterized in several animals like baboons (Porter et al., 2007), cows (Seo et al., 2007; de Almeida et al., 2008), pigs (Kaser et al., 2008), cats (Lankford et al., 2008), and rabbits (Nesburn et al., 2007). We earlier identified and characterized chicken CD4+CD25+ cells as Tregs in chickens (Shanmugasundaram and Selvaraj, 2011b). Similar to the CD25 expression in mammals (Baecher–Allan et al., 2001), CD25 expression in chicken thymic CD4+ cells was continuous in that cells express high, intermediate, or low levels of CD25, and the boundary between CD25high, CD25intermediate, and CD25low population is not clear (Shanmugasundaram and Selvaraj, 2011b). Chicken Tregs produce 29–fold higher IL–10 mRNA than non–Tregs. IL–10, an immunosuppressive cytokine, inhibits macrophages and dendritic cell functions (Fujio et al., 2010) and is a critical cytokine responsible for the suppressive properties of Tregs (Gangi et al., 2005). Similar to chicken Tregs, ducks (Shanmugasundaram and Selvaraj, 2012d) and turkey (Shanmugasundaram and Selvaraj, 2012e) CD4+CD25+ cells had higher IL–10, TGF–β, CTLA–4, and LAG–3 mRNA amounts than CD4+CD25– cells from the respective species.

In chickens, Tregs initially appear at 16 d of embryonic development, and the first wave of Tregs preferentially migrates to the intestine (Shanmugasundaram and Selvaraj, 2012a). We identified that a single peritoneal injection of anti–chicken CD25 mAb decreases IL–10–producing Tregs in the intestine of chickens by approximately 80% (Shanmugasundaram and Selvaraj, 2012b). The depletion is temporary as Tregs return to their baseline levels at approximately 20 d post–CD25 injection. In ovo injection of 0.5 mg/egg of anti–chicken CD25 mAb at 16 d of embryonic development almost completely depleted circulating Tregs at hatch and that the birds remained depleted of Tregs until 25 d post–hatch. Chicks hatched from anti–chicken CD25–mAb–injected eggs had ~75% decrease in Tregs in the cecal tonsils at 16 d post–hatch (Shanmugasundaram and Selvaraj, 2013). Chicks hatched from anti–chicken CD25 mAb injected eggs also had no detectable amount of Tregs in cecal tonsils at 0, 3, and 5 d post–hatch (unpublished observations).

We have also characterized chicken Tregs during Salmonella (a gut pathogen like coccidia) lipopolysaccharide–induced inflammation (Shanmugasundaram and Selvaraj, 2011a; 2012c). The LPS injection increases CD4+CD25+ cell percentage approximately 2.5–fold in the spleen at 2 d post–LPS injection compared to the no–LPS–injected group, though the Treg numbers came back to normal levels at 5 d post–LPS injection (Shanmugasundaram and Selvaraj, 2012c). We evaluated the suppressive properties of chicken CD25+ cells from LPS injected or control groups. At a Treg: T responder cell ratio of 1:1, CD25+ cells only from 5 and 12d post–LPS injection were suppressive while CD25+ cells from 2 d post–LPs injection were not suppressive. Chicken non–Tregs appear, therefore, to up-regulate CD25 transiently, with no suppressive properties, post–LPS treatment. The other possibility is Tregs undergoes extensive proliferation and lose suppressive properties post–LPS treatment. Because chicken Treg specific markers are not available, we cannot exclude either of the above possibility.

Compared to mammalian Tregs, research in avian Tregs are in earlier stages. Further characterization of avian Tregs will benefit poultry production. For example, avian Tregs, with anti–inflammatory potential, can be targeted to decrease inflammation and mortality during an immune response in commercial settings. In mammals, Treg–targeted therapy [anti–CTLA–4 (Gabriel and Lattime, 2007), anti–IL–2 (Kottke et al., 2008), anti–CD25 (Bielekova et al., 2004)] is applied for numerous situations, e.g. infections, tumors, autoimmune diseases, sepsis, shock, and vaccine. Similar to mammals, avian diseases will benefit from Treg–targeted therapy. A host immune response is a result of interplay between different components of the immune system. Interplay between Tregs and other components of the immune system will determine whether the outcome will be a persistent infection or successful pathogen clearance.

REFERENCES

Aandahl EM, Michaelsson J, Moretto WJ, Hecht FM and Nixon DF (2004). Human CD4+ CD25+ regulatory T cells control T–cell responses to human immunodeficiency virus and cytomegalovirus antigens. J. Virol. 78: 2454–2459.
http://dx.doi.org/10.1128/JVI.78.5.2454-2459.2004
PMid:14963140 PMCid:PMC369239

Antunes I and Kassiotis G (2010). Suppression of innate immune pathology by regulatory T cells during Influenza A virus infection of immunodeficient mice. J. Virol. 84: 12564–12575.
http://dx.doi.org/10.1128/JVI.01559-10
PMid:20943986 PMCid:PMC3004299

Apostolou I, Sarukhan A, Klein L and von Boehmer H (2002). Origin of regulatory T cells with known specificity for antigen. Nat. Immunol. 3: 756–763.
PMid:12089509

Baecher–Allan C, Brown JA, Freeman GJ and Hafler DA (2001). CD4+CD25 high regulatory cells in human peripheral blood. J. Immunol. 167: 1245–1253.
http://dx.doi.org/10.4049/jimmunol.167.3.1245
PMid:11466340

Belkaid Y (2007). Regulatory T cells and infection: a dangerous necessity. Nat. Rev. Immunol. 7: 875–888.
http://dx.doi.org/10.1038/nri2189
PMid:17948021

Belkaid Y and Rouse BT (2005). Natural regulatory T cells in infectious disease. Nat. Immunol. 6: 353–360.
http://dx.doi.org/10.1038/ni1181
PMid:15785761

Bielekova B, Richert N, Howard T, Blevins G, Markovic–Plese S, McCartin J, Frank JA, Wurfel J, Ohayon J, Waldmann TA, McFarland HF and Martin R (2004). Humanized anti–CD25 (daclizumab) inhibits disease activity in multiple sclerosis patients failing to respond to interferon beta. Proc. Natl. Acad. Sci. U S A. 101: 8705–8708.
http://dx.doi.org/10.1073/pnas.0402653101
PMid:15161974 PMCid:PMC423259

Boettler T, Spangenberg HC, Neumann–Haefelin C, Panther E, Urbani S, Ferrari C, Blum HE, von Weizsacker F and Thimme R (2005). T cells with a CD4+CD25+ regulatory phenotype suppress in vitro proliferation of virus–specific CD8+ T cells during chronic hepatitis C virus infection. J. Virol. 79: 7860–7867.
http://dx.doi.org/10.1128/JVI.79.12.7860-7867.2005
PMid:15919940 PMCid:PMC1143651

Bolacchi F, Sinistro A, Ciaprini C, Demin F, Capozzi M, Carducci FC, Drapeau CM, Rocchi G and Bergamini A (2006). Increased hepatitis C virus (HCV)–specific CD4+CD25+ regulatory T lymphocytes and reduced HCV–specific CD4+ T cell response in HCV–infected patients with normal versus abnormal alanine aminotransferase levels. Clin. Exp. Immunol. 144: 188–196.
http://dx.doi.org/10.1111/j.1365-2249.2006.03048.x
PMid:16634790 PMCid:PMC1809656

Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, Wilkinson JE, Galas D, Ziegler SF and Ramsdell F (2001). Disruption of a new forkhead/winged–helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat. Genet. 27: 68–73.
http://dx.doi.org/10.1038/83784
PMid:11138001

Caramalho I, Lopes–Carvalho T, Ostler D, Zelenay S, Haury M and Demengeot J (2003). Regulatory T cells selectively express toll–like receptors and are activated by lipopolysaccharide. J. Exp. Med. 197: 403–411.
http://dx.doi.org/10.1084/jem.20021633
PMid:12591899 PMCid:PMC2193858

Carrier Y, Yuan J, Kuchroo VK and Weiner HL (2007). Th3 cells in peripheral tolerance. I. Induction of Foxp3–positive regulatory T cells by Th3 cells derived from TGF–beta T cell–transgenic mice. J. Immunol. 178: 179–185.
http://dx.doi.org/10.4049/jimmunol.178.1.172
http://dx.doi.org/10.4049/jimmunol.178.1.179
PMid:17182553

Chappert P, Leboeuf M, Rameau P, Lalfer M, Desbois S, Liblau RS, Danos O, Davoust JM and Gross DA (2010). Antigen–specific Treg impair CD8(+) T–cell priming by blocking early T–cell expansion. Eur. J. Immunol. 40: 339–350.
http://dx.doi.org/10.1002/eji.200839107
PMid:19877007

de Almeida DE, Colvin CJ and Coussens PM (2008). Antigen–specific regulatory T cells in bovine paratuberculosis. Vet. Immunol. Immunopathol. 125: 234–245.
http://dx.doi.org/10.1016/j.vetimm.2008.05.019
PMid:18602164

Dieckmann D, Plottner H, Berchtold S, Berger T and Schuler G (2001). Ex vivo isolation and characterization of CD4(+)CD25(+) T cells with regulatory properties from human blood. J. Exp. Med. 193: 1303–1310.
http://dx.doi.org/10.1084/jem.193.11.1303
PMid:11390437 PMCid:PMC2193384

Dietze KK, Zelinskyy G, Gibbert K, Schimmer S, Francois S, Myers L, Sparwasser T, Hasenkrug KJ and Dittmer U (2011). Transient depletion of regulatory T cells in transgenic mice reactivates virus–specific CD8+ T cells and reduces chronic retroviral set points. Proc. Natl. Acad. Sci. U S A. 108: 2420–2425.
http://dx.doi.org/10.1073/pnas.1015148108
PMid:21262821 PMCid:PMC3038736

Epple HJ, Loddenkemper C, Kunkel D, Troger H, Maul J, Moos V, Berg E, Ullrich R, Schulzke JD, Stein H, Duchmann R, Zeitz M and Schneider T (2006). Mucosal but not peripheral FOXP3+ regulatory T cells are highly increased in untreated HIV infection and normalize after suppressive HAART. Blood. 108: 3072–3078.
http://dx.doi.org/10.1182/blood-2006-04-016923
PMid:16728694

Frimpong–Boateng K, van Rooijen N and Geiben–Lynn R (2010). Regulatory T cells suppress natural killer cells during plasmid DNA vaccination in mice, blunting the CD8+ T Cell Immune Response by the Cytokine TGFbeta. PLoS One. 5: e12281.
http://dx.doi.org/10.1371/journal.pone.0012281
PMid:20808850 PMCid:PMC2924372

Fujio K, Okamura T and Yamamoto K (2010). The family of IL–10–secreting CD4+ T cells. Adv. Immunol. 105: 99–130.
http://dx.doi.org/10.1016/S0065-2776(10)05004-2

Gabriel EM and Lattime EC (2007). Anti–CTL–associated antigen 4: are regulatory T cells a target? Clin. Cancer. Res. 13: 785–788.
http://dx.doi.org/10.1158/1078-0432.CCR-06-2820
PMid:17289867

Gangi E, Vasu C, Cheatem D and Prabhakar BS (2005). IL–10–producing CD4+CD25+ regulatory T cells play a critical role in granulocyte–macrophage colony–stimulating factor–induced suppression of experimental autoimmune thyroiditis. J. Immunol. 174: 7006–7013.
http://dx.doi.org/10.4049/jimmunol.174.11.7006
PMid:15905543

Gavin MA, Torgerson TR, Houston E, DeRoos P, Ho WY, Stray–Pedersen A, Ocheltree EL, Greenberg PD, Ochs HD and Rudensky AY (2006). Single–cell analysis of normal and FOXP3–mutant human T cells: FOXP3 expression without regulatory T cell development. Proc. Natl. Acad. Sci. U S A. 103: 6659–6664.
http://dx.doi.org/10.1073/pnas.0509484103
PMid:16617117 PMCid:PMC1458937

Giamarellos–Bourboulis EJ, Raftogiannis M, Antonopoulou A, Baziaka F, Koutoukas P, Savva A, Kanni T, Georgitsi M, Pistiki A, Tsaganos T, Pelekanos N, Athanassia S, Galani L, Giannitsioti E, Kavatha D, Kontopidou F, Mouktaroudi M, Poulakou G, Sakka V, Panagopoulos P, Papadopoulos A, Kanellakopoulou K and Giamarellou H (2009). Effect of the novel influenza A (H1N1) virus in the human immune system. PLoS One. 4: e8393.
http://dx.doi.org/10.1371/journal.pone.0008393
PMid:20037642 PMCid:PMC2792719

Gri G, Piconese S, Frossi B, Manfroi V, Merluzzi S, Tripodo C, Viola A, Odom S, Rivera J, Colombo MP and Pucillo CE (2008). CD4+CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40–OX40L interaction. Immunity. 29: 771–781.
http://dx.doi.org/10.1016/j.immuni.2008.08.018
PMid:18993084 PMCid:PMC2590499

Haeryfar SM, DiPaolo RJ, Tscharke DC, Bennink JR and Yewdell JW (2005). Regulatory T cells suppress CD8+ T cell responses induced by direct priming and cross–priming and moderate immunodominance disparities. J. Immunol. 174: 3344–3351.
http://dx.doi.org/10.4049/jimmunol.174.6.3344
PMid:15749866

Hoffmann JC, Pawlowski NN, Grollich K, Loddenkemper C, Zeitz M and Kuhl AA (2008). Gammadelta T lymphocytes: a new type of regulatory T cells suppressing murine 2,4,6–trinitrobenzene sulphonic acid (TNBS)–induced colitis. Int. J. Colorectal. Dis. 23: 909–920.
http://dx.doi.org/10.1007/s00384-008-0535-8
PMid:18649083

Hori S, Nomura T and Sakaguchi S (2003). Control of regulatory T cell development by the transcription factor Foxp3. Science. 299: 1057–1061.
http://dx.doi.org/10.1126/science.1079490
PMid:12522256

Huter EN, Punkosdy GA, Glass DD, Cheng LI, Ward JM and Shevach EM (2008). TGF–beta–induced Foxp3+ regulatory T cells rescue scurfy mice. Eur. J. Immunol. 38: 1814–1821.
http://dx.doi.org/10.1002/eji.200838346
PMid:18546144 PMCid:PMC2574868

Jonuleit H and Schmitt E (2003). The regulatory T cell family: distinct subsets and their interrelations. J. Immunol. 171: 6323–6327.
http://dx.doi.org/10.4049/jimmunol.171.12.6323
PMid:14662827

Jonuleit H, Schmitt E, Stassen M, Tuettenberg A, Knop J and Enk AH (2001). Identification and functional characterization of human CD4(+)CD25(+) T cells with regulatory properties isolated from peripheral blood. J. Exp. Med. 193: 1285–1294.
http://dx.doi.org/10.1084/jem.193.11.1285
PMid:11390435 PMCid:PMC2193380

Kaser T, Gerner W, Hammer SE, Patzl M and Saalmuller A (2008). Phenotypic and functional characterisation of porcine CD4(+)CD25(high) regulatory T cells. Vet. Immunol. Immunopathol. 122: 153–158.
http://dx.doi.org/10.1016/j.vetimm.2007.08.002
PMid:17868905

Khatri M, Dwivedi V, Krakowka S, Manickam C, Ali A, Wang L, Qin Z, Renukaradhya GJ and Lee CW (2010). Swine influenza H1N1 virus induces acute inflammatory immune responses in pig lungs: a potential animal model for human H1N1 influenza virus. J. Virol. 84: 11210–11218.
http://dx.doi.org/10.1128/JVI.01211-10
PMid:20719941 PMCid:PMC2953174

Kinter AL, Hennessey M, Bell A, Kern S, Lin Y, Daucher M, Planta M, McGlaughlin M, Jackson R, Ziegler SF and Fauci AS (2004). CD25(+)CD4(+) regulatory T cells from the peripheral blood of asymptomatic HIV–infected individuals regulate CD4(+) and CD8(+) HIV–specific T cell immune responses in vitro and are associated with favorable clinical markers of disease status. J. Exp. Med. 200: 331–343.
http://dx.doi.org/10.1084/jem.20032069
PMid:15280419 PMCid:PMC2211981

Kottke T, Galivo F, Wongthida P, Diaz RM, Thompson J, Jevremovic D, Barber GN, Hall G, Chester J, Selby P, Harrington K, Melcher A and Vile RG (2008). Treg depletion–enhanced IL–2 treatment facilitates therapy of established tumors using systemically delivered oncolytic virus. Mol. Ther. 16: 1217–1226.
http://dx.doi.org/10.1038/mt.2008.83
PMid:18431359 PMCid:PMC2729455

Landay AL, Clerici M, Hashemi F, Kessler H, Berzofsky JA and Shearer GM (1996). In vitro restoration of T cell immune function in human immunodeficiency virus–positive persons: effects of interleukin (IL)–12 and anti–IL–10. J. Infect. Dis. 173: 1085–1091.
http://dx.doi.org/10.1093/infdis/173.5.1085
PMid:8627058

Lankford S, Petty C, LaVoy A, Reckling S, Tompkins W and Dean GA (2008). Cloning of feline FOXP3 and detection of expression in CD4+CD25+ regulatory T cells. Vet. Immunol. Immunopathol. 122: 159–166.
http://dx.doi.org/10.1016/j.vetimm.2007.11.007
PMid:18180044 PMCid:PMC2277518

Li S, Gowans EJ, Chougnet C, Plebanski M and Dittmer U (2008). Natural regulatory T cells and persistent viral infection. J. Virol. 82: 21–30.
http://dx.doi.org/10.1128/JVI.01768-07
PMid:17855537 PMCid:PMC2224364

Lim HW, Hillsamer P, Banham AH and Kim CH (2005). Cutting edge: direct suppression of B cells by CD4+ CD25+ regulatory T cells. J. Immunol. 175: 4180–4183.
http://dx.doi.org/10.5772/4668

Lu L and Cantor H (2008). Generation and regulation of CD8(+) regulatory T cells. Cell. Mol. Immunol. 5: 401–406.
http://dx.doi.org/10.1038/cmi.2008.50
PMid:19118505 PMCid:PMC4072402

Lund JM, Hsing L, Pham TT and Rudensky AY (2008). Coordination of early protective immunity to viral infection by regulatory T cells. Science. 320: 1220–1224.
http://dx.doi.org/10.1126/science.1155209
PMid:18436744 PMCid:PMC2519146

Lysaght J, Jarnicki AG and Mills KH (2007). Reciprocal effects of Th1 and Treg cell inducing pathogen–associated immunomodulatory molecules on anti–tumor immunity. Cancer. Immunol. Immunother. 56: 1367–1379.
http://dx.doi.org/10.1007/s00262-007-0288-1
PMid:17279412

Mikkelsen SR, Reckling SK, Egan EA and Dean GA (2010). In vivo depletion of CD4(+)CD25(hi) regulatory T cells is associated with improved antiviral responses in cats chronically infected with feline immunodeficiency virus. Virology. 403: 163–172.
http://dx.doi.org/10.1016/j.virol.2010.04.016
PMid:20471053 PMCid:PMC2879469

Nesburn AB, Bettahi I, Dasgupta G, Chentoufi AA, Zhang X, You S, Morishige N, Wahlert AJ, Brown DJ, Jester JV, Wechsler SL and BenMohamed L (2007). Functional Foxp3+ CD4+ CD25(Bright+) "natural" regulatory T cells are abundant in rabbit conjunctiva and suppress virus–specific CD4+ and CD8+ effector T cells during ocular herpes infection. J. Virol. 81: 7647–7661.
http://dx.doi.org/10.1128/JVI.00294-07
PMid:17475646 PMCid:PMC1933381

Nilsson J, Boasso A, Velilla PA, Zhang R, Vaccari M, Franchini G, Shearer GM, Andersson J and Chougnet C (2006). HIV–1–driven regulatory T–cell accumulation in lymphoid tissues is associated with disease progression in HIV/AIDS. Blood. 108: 3808–3817.
http://dx.doi.org/10.1182/blood-2006-05-021576
PMid:16902147 PMCid:PMC1895475

Oh U, Grant C, Griffith C, Fugo K, Takenouchi N and Jacobson S (2006). Reduced Foxp3 protein expression is associated with inflammatory disease during human T lymphotropic virus type 1 Infection. J. Infect. Dis. 193: 1557–1566.
http://dx.doi.org/10.1086/503874
PMid:16652285

Porter CM, Horvath–Arcidiacono JA, Singh AK, Horvath KA, Bloom ET and Mohiuddin MM (2007). Characterization and expansion of baboon CD4+CD25+ Treg cells for potential use in a non–human primate xenotransplantation model. Xenotransplantation. 14: 298–308.
http://dx.doi.org/10.1111/j.1399-3089.2007.00416.x
PMid:17669171

Qian C, An H, Yu Y, Liu S and Cao X (2007). TLR agonists induce regulatory dendritic cells to recruit Th1 cells via preferential IP–10 secretion and inhibit Th1 proliferation. Blood. 109: 3308–3315.
http://dx.doi.org/10.1182/blood-2006-08-040337
PMid:17170125

Raimondi G, Turner MS, Thomson AW and Morel PA (2007). Naturally occurring regulatory T cells: recent insights in health and disease. Crit. Rev. Immunol. 27: 61–95.
http://dx.doi.org/10.1615/CritRevImmunol.v27.i1.50
PMid:17430097

Rehermann B, Ferrari C, Pasquinelli C and Chisari FV (1996). The hepatitis B virus persists for decades after patients' recovery from acute viral hepatitis despite active maintenance of a cytotoxic T–lymphocyte response. Nat. Med. 2: 1104–1108.
http://dx.doi.org/10.1038/nm1096-1104
PMid:8837608

Robertson SJ, Messer RJ, Carmody AB and Hasenkrug KJ (2006). In vitro suppression of CD8+ T cell function by Friend virus–induced regulatory T cells. J. Immunol. 176: 3342–3349.
http://dx.doi.org/10.4049/jimmunol.176.6.3342
PMid:16517701

Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K and Levings MK (2006). Interleukin–10–secreting type 1 regulatory T cells in rodents and humans. Immunol. Rev. 212: 28–50.
http://dx.doi.org/10.1111/j.0105-2896.2006.00420.x
PMid:16903904

Sakaguchi S, Yamaguchi T, Nomura T and Ono M (2008). Regulatory T cells and immune tolerance. Cell. 133: 775–787.
http://dx.doi.org/10.1016/j.cell.2008.05.009
PMid:18510923

Selvaraj RK (2013). Avian CD4 CD25 regulatory T cells: Properties and therapeutic applications. Dev. Comp. Immunol. (in press).
http://dx.doi.org/10.1016/j.dci.2013.04.018
PMid:23665004

Seo KS, Lee SU, Park YH, Davis WC, Fox LK and Bohach GA (2007). Long–term staphylococcal enterotoxin C1 exposure induces soluble factor–mediated immunosuppression by bovine CD4+ and CD8+ T cells. Infect. Immun. 75: 260–269.
http://dx.doi.org/10.1128/IAI.01358-06
PMid:17030573 PMCid:PMC1828382

Shanmugasundaram R and Selvaraj RK (2011a). In vitro lipopolysaccharide treatment alters regulatory T cell properties in chickens. Vet. Immunol. Immunopathol. 144: 476–481.
http://dx.doi.org/10.1016/j.vetimm.2011.08.016
PMid:21944051

Shanmugasundaram R and Selvaraj RK (2011b). Regulatory T cell properties of chicken CD4+CD25+ cells. J. Immunol. 186: 1997–2002.
http://dx.doi.org/10.4049/jimmunol.1002040
PMid:21242520

Shanmugasundaram R and Selvaraj RK (2012a). CD4+CD25+ Regulatory T cell ontogeny and preferential migration to the cecal tonsils in chickens. PLoS One. 7: e33970.
http://dx.doi.org/10.1371/journal.pone.0033970
PMid:22479489 PMCid:PMC3313958

Shanmugasundaram R and Selvaraj RK (2012b). Effects of in vivo injection of anti–chicken CD25 monoclonal antibody on regulatory T cell depletion and CD4+CD25– T cell properties in chickens. Develop. Comp. Immunol. 36: 578–583.
http://dx.doi.org/10.1016/j.dci.2011.09.015
PMid:22004798

Shanmugasundaram R and Selvaraj RK (2012c). In vivo lipopolysaccharide injection alters CD4+CD25+ cell properties in chickens. J. Anim. Sci. 90: 2498–2504.
http://dx.doi.org/10.2527/jas.2011-4652
PMid:22287676

Shanmugasundaram R and Selvaraj RK (2012d). Regulatory T cell properties of thymic CD4+CD25+ cells in ducks. Vet. Immunol. Immunopathol. 149: 20–27.
http://dx.doi.org/10.1016/j.vetimm.2012.05.019
PMid:22717168

Shanmugasundaram R and Selvaraj RK (2012e). Regulatory T cell properties of thymic CD4+CD25+ cells in turkeys. Poult. Sci. 91: 1833–1837.
http://dx.doi.org/10.3382/ps.2012-02202
PMid:22802175

Shanmugasundaram R and Selvaraj RK (2013). In ovo injection of anti–chicken CD25 monoclonal antibodies depletes CD4+CD25+ T cells in chickens. Poult. Sci. 92: 138–142.
http://dx.doi.org/10.3382/ps.2012-02593
PMid:23243240

Smyth MJ and Godfrey DI (2000). NKT cells and tumor immunity––a double–edged sword. Nat. Immunol. 1: 459–460.
http://dx.doi.org/10.1038/82698
PMid:11101862

Stober CB, Lange UG, Roberts MT, Alcami A and Blackwell JM (2005). IL–10 from regulatory T cells determines vaccine efficacy in murine Leishmania major infection. J. Immunol. 175: 2517–2524.
http://dx.doi.org/10.4049/jimmunol.175.4.2517
PMid:16081824

Stoop JN, van der Molen RG, Kuipers EJ, Kusters JG and Janssen HL (2007). Inhibition of viral replication reduces regulatory T cells and enhances the antiviral immune response in chronic hepatitis B. Virology. 361: 141–148.
http://dx.doi.org/10.1016/j.virol.2006.11.018
PMid:17184802

Sun L, Yi S and O'Connell PJ (2010). IL–10 is required for human CD4(+)CD25(+) regulatory T cell–mediated suppression of xenogeneic proliferation. Immunol. Cell. Biol. 88: 477–485.
http://dx.doi.org/10.1038/icb.2009.117
http://dx.doi.org/10.1038/icb.2010.41
PMid:20177412

Takahashi T, Kuniyasu Y, Toda M, Sakaguchi N, Itoh M, Iwata M, Shimizu J and Sakaguchi S (1998). Immunologic self–tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int. Immunol. 10: 1969–1980.
http://dx.doi.org/10.1093/intimm/10.12.1969
PMid:9885918

Teng Q–Y, Zhou J–Y, Wu J–J, Guo J–Q and Shen H–G (2006). Characterization of chicken interleukin 2 receptor α chain, a homolog to mammalian CD25. FEBS Letters. 580: 4274–4281.
http://dx.doi.org/10.1016/j.febslet.2006.06.044
PMid:16831435

Thorburn AN and Hansbro PM (2010). Harnessing regulatory T cells to suppress asthma: from potential to therapy. Am. J. Respir. Cell. Mol. Biol. 43: 511–519.
http://dx.doi.org/10.1165/rcmb.2009-0342TR
PMid:20097830 PMCid:PMC2970851

Thornton AM and Shevach EM (1998). CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J. Exp. Med. 188: 287–296.
http://dx.doi.org/10.1084/jem.188.2.287
PMid:9670041 PMCid:PMC2212461

Tiemessen MM, Jagger AL, Evans HG, van Herwijnen MJ, John S and Taams LS (2007). CD4+CD25+Foxp3+ regulatory T cells induce alternative activation of human monocytes/macrophages. Proc. Natl. Acad. Sci. U S A. 104: 19446–19451.
http://dx.doi.org/10.1073/pnas.0706832104
PMid:18042719 PMCid:PMC2148309

Wang J, Ioan–Facsinay A, van der Voort EI, Huizinga TW and Toes RE (2007). Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur. J. Immunol. 37: 129–138.
http://dx.doi.org/10.1002/eji.200636435
PMid:17154262

Ward SM, Fox BC, Brown PJ, Worthington J, Fox SB, Chapman RW, Fleming KA, Banham AH and Klenerman P (2007). Quantification and localisation of FOXP3+ T lymphocytes and relation to hepatic inflammation during chronic HCV infection. J. Hepatol. 47: 316–324.
http://dx.doi.org/10.1016/j.jhep.2007.03.023
PMid:17475362

Wildner G, Hünig T and Thurau SR (1997). Orally induced, HLA–peptide specific gamma–delta T–cells suppress experimental autoimmune uveitis in the rat. Immunology. Letters. 56: 35.
http://dx.doi.org/10.1016/S0165-2478(97)85132-2
http://dx.doi.org/10.1016/S0165-2478(97)86970-2

Wildner G, Thurau SR and Diedrichs–Mohring M (2004). Gamma–delta T cells as orally induced suppressor cells in rats: in vitro characterization. Ann. N Y Acad. Sci. 1029: 416–421.
http://dx.doi.org/10.1196/annals.1309.050
PMid:15681796

Wilson MS, Taylor MD, Balic A, Finney CA, Lamb JR and Maizels RM (2005). Suppression of allergic airway inflammation by helminth–induced regulatory T cells. J. Exp. Med. 202: 1199–1212.
http://dx.doi.org/10.1084/jem.20042572
PMid:16275759 PMCid:PMC2213237

Workman CJ, Szymczak–Workman AL, Collison LW, Pillai MR and Vignali DA (2009). The development and function of regulatory T cells. Cell. Mol. Life. Sci. 66: 2603–2622.
http://dx.doi.org/10.1007/s00018-009-0026-2
PMid:19390784 PMCid:PMC2715449

Xu D, Fu J, Jin L, Zhang H, Zhou C, Zou Z, Zhao JM, Zhang B, Shi M, Ding X, Tang Z, Fu YX and Wang FS (2006). Circulating and liver resident CD4+CD25+ regulatory T cells actively influence the antiviral immune response and disease progression in patients with hepatitis B. J. Immunol. 177: 739–747.
http://dx.doi.org/10.4049/jimmunol.177.1.739
PMid:16785573

Yamano Y, Takenouchi N, Li HC, Tomaru U, Yao K, Grant CW, Maric DA and Jacobson S (2005). Virus–induced dysfunction of CD4+CD25+ T cells in patients with HTLV–I–associated neuroimmunological disease. J. Clin. Invest. 115: 1361–1368.
http://dx.doi.org/10.1172/JCI200523913
http://dx.doi.org/10.1172/JCI23913
PMid:15864353 PMCid:PMC1087174

Yi H, Zhen Y, Jiang L, Zheng J and Zhao Y (2006). The phenotypic characterization of naturally occurring regulatory CD4+CD25+ T cells. Cell. Mol. Immunol. 3: 189–195.
PMid:16893499

Zelinskyy G, Dietze K, Sparwasser T and Dittmer U (2009). Regulatory T cells suppress antiviral immune responses and increase viral loads during acute infection with a lymphotropic retrovirus. PLoS Pathog. 5: e1000406.
http://dx.doi.org/10.1371/journal.ppat.1000406
PMid:19714239 PMCid:PMC2727466

Zelinskyy G, Kraft AR, Schimmer S, Arndt T and Dittmer U (2006). Kinetics of CD8+ effector T cell responses and induced CD4+ regulatory T cell responses during Friend retrovirus infection. Eur. J. Immunol. 36: 2658–2670.
http://dx.doi.org/10.1002/eji.200636059
PMid:16981182

Zhang ZX, Yang L, Young KJ, DuTemple B and Zhang L (2000). Identification of a previously unknown antigen–specific regulatory T cell and its mechanism of suppression. Nat. Med. 6: 782–789.
http://dx.doi.org/10.1038/77513
http://dx.doi.org/10.1038/72303