Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Nat Immunol ; 17(9): 1093-101, 2016 09.
Article in English | MEDLINE | ID: mdl-27478940

ABSTRACT

The manner in which regulatory T cells (Treg cells) control lymphocyte homeostasis is not fully understood. We identified two Treg cell populations with differing degrees of self-reactivity and distinct regulatory functions. We found that GITR(hi)PD-1(hi)CD25(hi) (Triple(hi)) Treg cells were highly self-reactive and controlled lympho-proliferation in peripheral lymph nodes. GITR(lo)PD-1(lo)CD25(lo) (Triple(lo)) Treg cells were less self-reactive and limited the development of colitis by promoting the conversion of CD4(+) Tconv cells into induced Treg cells (iTreg cells). Although Foxp3-deficient (Scurfy) mice lacked Treg cells, they contained Triple(hi)-like and Triple(lo)-like CD4(+) T cells zsuper> T cells infiltrated the skin, whereas Scurfy Triple(lo)CD4(+) T cells induced colitis and wasting disease. These findings indicate that the affinity of the T cell antigen receptor for self antigen drives the differentiation of Treg cells into distinct subsets with non-overlapping regulatory activities.


Subject(s)
Colitis/immunology , Lymph Nodes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Wasting Syndrome/immunology , Animals , Autoantigens/immunology , Autoimmunity , Cell Differentiation , Cell Proliferation , Cells, Cultured , Clonal Selection, Antigen-Mediated , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/transplantation
2.
Immunity ; 47(1): 171-182.e4, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723549

ABSTRACT

Interleukin-7 (IL-7) availability determines the size and proliferative state of the resting T cell pool. However, the mechanisms that regulate steady-state IL-7 amounts are unclear. Using experimental lymphopenic mouse models and IL-7-induced homeostatic proliferation to measure IL-7 availability in vivo, we found that radioresistant cells were the source of IL-7 for both CD4+ and CD8+ T cells. Hematopoietic lineage cells, although irrelevant as a source of IL-7, were primarily responsible for limiting IL-7 availability via their expression of IL-7R. Unexpectedly, innate lymphoid cells were found to have a potent influence on IL-7 amounts in the primary and secondary lymphoid tissues. These results demonstrate that IL-7 homeostasis is achieved through consumption by multiple subsets of innate and adaptive immune cells.


Subject(s)
Hematopoietic Stem Cells/physiology , Interleukin-7/metabolism , Lymphocytes/immunology , Lymphopenia/immunology , T-Lymphocytes/physiology , Adaptive Immunity , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Homeostasis , Humans , Immunity, Innate , Interleukin-7/genetics , Interleukin-7/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Tolerance , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism
3.
Nat Immunol ; 12(6): 478-84, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21739670

ABSTRACT

Weak T cell antigen receptor (TCR) signals from contact with self ligands act in synergy with antiapoptotic signals induced by interleukin 7 (IL-7) to promote the survival of naive T cells in a resting state. The amount of background TCR signaling in naive T cells is set by post-thymic TCR tuning and operates at an intensity just below that required to induce entry into the cell cycle. Costimulation from higher concentrations of IL-7 and other common γ-chain cytokines can induce T cells to undergo homeostatic proliferation and conversion into cells with a memory phenotype; many of these memory phenotype cells may be the progeny of cells responding to self antigens. The molecular mechanisms that control the conversion of naive resting T cells into memory-phenotype cells TCR-dependent in normal animals are beginning to be understood.


Subject(s)
Homeostasis/immunology , Immunologic Memory/immunology , Interleukin-7/immunology , T-Lymphocytes/immunology , Animals , Cell Survival/immunology , Humans , Interleukin-7/metabolism , Models, Immunological , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/metabolism
4.
Proc Natl Acad Sci U S A ; 116(3): 1007-1016, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30598454

ABSTRACT

T cells proliferate vigorously following acute depletion of CD4+ Foxp3+ T regulatory cells [natural Tregs (nTregs)] and also when naive T cells are transferred to syngeneic, nTreg-deficient Rag1-/- hosts. Here, using mice raised in an antigen-free (AF) environment, we show that proliferation in these two situations is directed to self ligands rather than food or commensal antigens. In both situations, the absence of nTregs elevates B7 expression on host dendritic cells (DCs) and enables a small subset of naive CD4 T cells with high self affinity to respond overtly to host DCs: bidirectional T/DC interaction ensues, leading to progressive DC activation and reciprocal strong proliferation of T cells accompanied by peripheral Treg (pTreg) formation. Likewise, high-affinity CD4 T cells proliferate vigorously and form pTregs when cultured with autologous DCs in vitro in the absence of nTregs: this anti-self response is MHCII/peptide dependent and elicited by the raised level of B7 on cultured DCs. The data support a model in which self tolerance is imposed via modulation of CD28 signaling and explains the pathological effects of superagonistic CD28 antibodies.


Subject(s)
Cell Proliferation , Dendritic Cells/immunology , Immune Tolerance , Models, Immunological , T-Lymphocytes, Regulatory/immunology , Animals , B7 Antigens/genetics , B7 Antigens/immunology , CD28 Antigens/genetics , CD28 Antigens/immunology , Dendritic Cells/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/cytology
6.
Blood ; 134(16): 1312-1322, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31387916

ABSTRACT

The microbiota regulate hematopoiesis in the bone marrow (BM); however, the detailed mechanisms remain largely unknown. In this study, we explored how microbiota-derived molecules (MDMs) were transferred to the BM and sensed by the local immune cells to control hematopoiesis under steady-state conditions. We reveal that MDMs, including bacterial DNA (bDNA), reach the BM via systemic blood circulation and are captured by CX3CR1+ mononuclear cells (MNCs). CX3CR1+ MNCs sense MDMs via endolysosomal Toll-like receptors (TLRs) to produce inflammatory cytokines, which control the basal expansion of hematopoietic progenitors, but not hematopoietic stem cells, and their differentiation potential toward myeloid lineages. CX3CR1+ MNCs colocate with hematopoietic progenitors at the perivascular region, and the depletion of CX3CR1+ MNCs impedes bDNA influx into the BM. Moreover, the abrogation of TLR pathways in CX3CR1+ MNCs abolished the microbiota effect on hematopoiesis. These studies demonstrate that systemic MDMs control BM hematopoiesis by producing CX3CR1+ MNC-mediated cytokines in the steady-state.


Subject(s)
Bone Marrow Cells/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Microbiota/physiology , Animals , CX3C Chemokine Receptor 1/metabolism , Cytokines/metabolism , Mice , Mice, Inbred C57BL
7.
EMBO Rep ; 20(4)2019 04.
Article in English | MEDLINE | ID: mdl-30783017

ABSTRACT

A lacteal is a blunt-ended, long, tube-like lymphatic vessel located in the center of each intestinal villus that provides a unique route for drainage of absorbed lipids from the small intestine. However, key regulators for maintaining lacteal integrity are poorly understood. Here, we explore whether and how the gut microbiota regulates lacteal integrity. Germ depletion by antibiotic treatment triggers lacteal regression during adulthood and delays lacteal maturation during the postnatal period. In accordance with compromised lipid absorption, the button-like junction between lymphatic endothelial cells, which is ultrastructurally open to permit free entry of dietary lipids into lacteals, is significantly reduced in lacteals of germ-depleted mice. Lacteal defects are also found in germ-free mice, but conventionalization of germ-free mice leads to normalization of lacteals. Mechanistically, VEGF-C secreted from villus macrophages upon MyD88-dependent recognition of microbes and their products is a main factor in lacteal integrity. Collectively, we conclude that the gut microbiota is a crucial regulator for lacteal integrity by endowing its unique microenvironment and regulating villus macrophages in small intestine.


Subject(s)
Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Macrophages/metabolism , Vascular Endothelial Growth Factor C/biosynthesis , Age Factors , Animals , Biological Transport , Biomarkers , CX3C Chemokine Receptor 1/metabolism , Fluorescent Antibody Technique , Intestinal Absorption , Intestinal Mucosa/cytology , Intestinal Mucosa/ultrastructure , Lipid Metabolism , Mice , Microvessels/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction
9.
Immunity ; 32(2): 214-26, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20137986

ABSTRACT

T cell receptor (TCR) contact with self ligands keeps T cells alive and is shown here to cause naive CD8(+), but not CD4(+), T cells to be hypersensitive to certain gamma(c) cytokines, notably interleukin (IL)-2, IL-15, and IL-7. Hypersensitivity of CD8(+) T cells to IL-2 was dependent on a low-level TCR signal, associated with high expression of CD5 and GM1, a marker for lipid rafts, and was abolished by disruption of lipid rafts. By contrast, CD4(+) T cells expressed low amounts of GM1 and were unresponsive to IL-2. Physiologically, sensitivity to IL-7 and IL-15 maintains survival of resting CD8(+) T cells, whereas sensitivity to IL-2 may be irrelevant for normal homeostasis but crucial for the immune response. Thus, TCR contact with antigen upregulates GM1 and amplifies responsiveness of naive CD8(+) T cells to IL-2, thereby making the cells highly sensitive to exogenous IL-2 from CD4(+) T helper cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Glycosphingolipids/biosynthesis , Membrane Microdomains/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Autoantigens/immunology , Autoantigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Survival/immunology , Cells, Cultured , Cytokines/immunology , Glycosphingolipids/genetics , Glycosphingolipids/immunology , Histocompatibility Antigens/metabolism , Homeostasis , Interleukin Receptor Common gamma Subunit/metabolism , Mice , Mice, Inbred C57BL , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology
10.
Gastroenterology ; 152(8): 1998-2010, 2017 06.
Article in English | MEDLINE | ID: mdl-28246016

ABSTRACT

BACKGROUND & AIMS: Obesity and metabolic syndrome have been associated with alterations to the intestinal microbiota. However, few studies examined the effects of obesity on the intestinal immune system. We investigated changes in subsets of intestinal CD4+ T-helper (TH) cells with obesity and the effects of gut-tropic TH17 cells in mice on a high-fat diet (HFD). METHODS: We isolated immune cells from small intestine and adipose tissue of C57BL/6 mice fed a normal chow diet or a HFD for 10 weeks and analyzed the cells by flow cytometry. Mice fed a vitamin A-deficient HFD were compared with mice fed a vitamin A-sufficient HFD. Obese RAG1-deficient mice were given injections of only regulatory T cells or a combination of regulatory T cells and TH17 cells (wild type or deficient in integrin ß7 subunit or interleukin 17 [IL17]). Mice were examined for weight gain, fat mass, fatty liver, glucose tolerance, and insulin resistance. Fecal samples were collected before and after T cell transfer and analyzed for microbiota composition by metagenomic DNA sequencing and quantitative polymerase chain reaction. RESULTS: Mice placed on a HFD became obese, which affected the distribution of small intestinal CD4+ TH cells. Intestinal tissues from obese mice had significant reductions in the proportion of TH17 cells but increased proportion of TH1 cells, compared with intestinal tissues from nonobese mice. Depletion of vitamin A in obese mice further reduced the proportion of TH17 cells in small intestine; this reduction correlated with more weight gain and worsening of glucose intolerance and insulin resistance. Adoptive transfer of in vitro-differentiated gut-tropic TH17 cells to obese mice reduced these metabolic defects, which required the integrin ß7 subunit and IL17. Delivery of TH17 cells to intestines of mice led to expansion of commensal microbes associated with leanness. CONCLUSIONS: In mice, intestinal TH17 cells contribute to development of a microbiota that maintains metabolic homeostasis, via IL17. Gut-homing TH17 cells might be used to reduce metabolic disorders in obese individuals.


Subject(s)
Adoptive Transfer , Immunity, Mucosal , Insulin Resistance , Intestine, Small/immunology , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Th17 Cells/transplantation , Animals , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Feces/microbiology , Gastrointestinal Microbiome/immunology , Genotype , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Host-Pathogen Interactions , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Interleukin-17/deficiency , Interleukin-17/genetics , Intestine, Small/metabolism , Intestine, Small/microbiology , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/microbiology , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/immunology , Obesity/microbiology , Phenotype , Th17 Cells/immunology , Th17 Cells/microbiology , Time Factors , Vitamin A Deficiency/complications
11.
Int Immunol ; 29(2): 71-78, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28338920

ABSTRACT

Immune tolerance in the lung is important for preventing hypersensitivity, such as allergic asthma. Maintenance of tolerance in the lung is established by coordinated activities of poorly understood cellular and molecular mechanisms, including participation of dendritic cells (DCs). We have previously identified DC expression of the signaling molecule TRAF6 as a non-redundant requirement for the maintenance of immune tolerance in the small intestine of mice. Because mucosal tissues share similarities in how they interact with exogenous antigens, we examined the role of DC-expressed TRAF6 in the lung. As with the intestine, we found that the absence TRAF6 expression by DCs led to spontaneous generation of Th2-associated immune responses and increased susceptibility to model antigen-induced asthma. To examine the role of commensal microbiota, mice deficient in TRAF6 in DCs were treated with broad-spectrum antibiotics and/or re-derived on a germ-free (GF) background. Interestingly, we found that antibiotics-treated specific pathogen-free, but not GF, mice showed restored immune tolerance in the absence of DC-expressed TRAF6. We further found that antibiotics mediate microbiota-independent effects on lung T cells to promote immune tolerance in the lung. This work provides both a novel tool for studying immune tolerance in the lung and an advance in our conceptual understanding of potentially common molecular mechanisms of immune tolerance in both the intestine and the lung.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Lung/immunology , TNF Receptor-Associated Factor 6/metabolism , Th2 Cells/immunology , Animals , Anti-Bacterial Agents/administration & dosage , Asthma/genetics , Cells, Cultured , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Immune Tolerance/genetics , Immunity, Mucosal , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , TNF Receptor-Associated Factor 6/genetics
12.
Proc Natl Acad Sci U S A ; 112(2): 476-81, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25548153

ABSTRACT

Lymphodepleting regimens are used before adoptive immunotherapy to augment the antitumor efficacy of transferred T cells by removing endogenous homeostatic "cytokine sinks." These conditioning modalities, however, are often associated with severe toxicities. We found that microRNA-155 (miR-155) enabled tumor-specific CD8(+) T cells to mediate profound antitumor responses in lymphoreplete hosts that were not potentiated by immune-ablation. miR-155 enhanced T-cell responsiveness to limited amounts of homeostatic γc cytokines, resulting in delayed cellular contraction and sustained cytokine production. miR-155 restrained the expression of the inositol 5-phosphatase Ship1, an inhibitor of the serine-threonine protein kinase Akt, and multiple negative regulators of signal transducer and activator of transcription 5 (Stat5), including suppressor of cytokine signaling 1 (Socs1) and the protein tyrosine phosphatase Ptpn2. Expression of constitutively active Stat5a recapitulated the survival advantages conferred by miR-155, whereas constitutive Akt activation promoted sustained effector functions. Our results indicate that overexpression of miR-155 in tumor-specific T cells can be used to increase the effectiveness of adoptive immunotherapies in a cell-intrinsic manner without the need for life-threatening, lymphodepleting maneuvers.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , MicroRNAs/genetics , MicroRNAs/immunology , Animals , Base Sequence , Cell Line, Tumor , Cytokines/biosynthesis , HEK293 Cells , Humans , Immunotherapy, Adoptive , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/immunology
15.
Immunity ; 29(6): 848-62, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19100699

ABSTRACT

The peripheral mature T cell pool is regulated by complex homeostatic mechanisms. Naive T cells are maintained by interleukin-7 (IL-7) and T cell receptor (TCR) signaling from contact with major histocompatibility complex (MHC), which sustain expression of antiapoptotic molecules and allow the cells to survive in interphase. Competition for these ligands declines when T cell numbers are reduced and causes residual naive T cells to proliferate and differentiate into memory-like cells. This memory cell population is thus heterogeneous and comprised of cells derived from responses to both foreign and self-antigens. Typical memory cells are kept alive and induced to divide intermittently by a mixture of IL-7 and IL-15. This review highlights recent advances in how naive and memory T cell homeostasis is regulated.


Subject(s)
Homeostasis/immunology , Immunologic Memory , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Humans , Interleukin-15/immunology , Interleukin-15/metabolism , Interleukin-7/immunology , Interleukin-7/metabolism , Interleukin-7 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Mice , Receptors, Antigen, T-Cell/metabolism , STAT Transcription Factors/immunology , STAT Transcription Factors/metabolism , Signal Transduction , T-Lymphocyte Subsets/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism
16.
J Virol ; 90(5): 2273-84, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26656713

ABSTRACT

UNLABELLED: Influenza A virus (IAV) infection frequently causes hospitalization and mortality due to severe immunopathology. Annual vaccination and antiviral drugs are the current countermeasures against IAV infection, but they have a limited efficacy against new IAV variants. Here, we show that intranasal pretreatment with Fc-fused interleukin-7 (IL-7-mFc) protects mice from lethal IAV infections. The protective activity of IL-7-mFc relies on transcytosis via neonatal Fc receptor (FcRn) in the lung and lasts for several weeks. Introduction of IL-7-mFc alters pulmonary immune environments, leading to recruitment of T cells from circulation and their subsequent residency as tissue-resident memory-like T (TRM-like) cells. IL-7-mFc-primed pulmonary TRM-like cells contribute to protection upon IAV infection by dual modes. First, TRM-like cells, although not antigen specific but polyclonal, attenuate viral replication at the early phase of IAV infection. Second, TRM-like cells augment expansion of IAV-specific cytotoxic T lymphocytes (CTLs), in particular at the late phase of infection, which directly control viruses. Thus, accelerated viral clearance facilitated by pulmonary T cells, which are either antigen specific or not, alleviates immunopathology in the lung and mortality from IAV infection. Depleting a subset of pulmonary T cells indicates that both CD4 and CD8 T cells contribute to protection from IAV, although IL-7-primed CD4 T cells have a more prominent role. Collectively, we propose intranasal IL-7-mFc pretreatment as an effective means for generating protective immunity against IAV infections, which could be applied to a potential prophylaxis for influenza pandemics in the future. IMPORTANCE: The major consequence of a highly pathogenic IAV infection is severe pulmonary inflammation, which can result in organ failure and death at worst. Although vaccines for seasonal IAVs are effective, frequent variation of surface viral proteins hampers development of protective immunity. In this study, we demonstrated that intranasal IL-7-mFc pretreatment protected immunologically naive mice from lethal IAV infections. Intranasal pretreatment with IL-7-mFc induced an infiltration of T cells in the lung, which reside as effector/memory T cells with lung-retentive markers. Those IL-7-primed pulmonary T cells contributed to development of protective immunity upon IAV infection, reducing pulmonary immunopathology while increasing IAV-specific cytotoxic T lymphocytes. Since a single treatment with IL-7-mFc was effective in the protection against multiple strains of IAV for an extended period of time, our findings suggest a possibility that IL-7-mFc treatment, as a potential prophylaxis, can be developed for controlling highly pathogenic IAV infections.


Subject(s)
Immunoglobulin Fc Fragments/administration & dosage , Immunologic Factors/administration & dosage , Influenza A virus/immunology , Interleukin-7/administration & dosage , Lung/immunology , Orthomyxoviridae Infections/prevention & control , Administration, Intranasal , Animals , Female , Immunoglobulin Fc Fragments/genetics , Immunologic Factors/genetics , Interleukin-7/genetics , Lymphocyte Depletion , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , T-Lymphocytes/immunology
17.
Xenotransplantation ; 23(4): 279-92, 2016 07.
Article in English | MEDLINE | ID: mdl-27373998

ABSTRACT

BACKGROUND: Despite the development of α1,3-galactosyl transferase-knockout (GTKO) pigs, acute humoral xenograft rejection caused by antibodies against non-Gal antigens, along with complement activation, are hurdles that need to be overcome. Among non-Gal antigens, N-glycolylneuraminic acid (Neu5Gc) is considered to play an important role in xenograft rejection in human. METHODS: We generated human embryonic kidney 293 (HEK293) cells that expressed xenogeneic Neu5Gc (HEK293-pCMAH) or α1,3Gal (HEK293-pGT) antigen and investigated the degree of human antibody binding and complement-dependent cytotoxicity (CDC) against these antigens using 100 individual human sera. RESULTS: Both IgM and IgG bound to α1,3Gal, while only IgG bound to Neu5Gc. Of the ABO blood groups, the degree of IgG binding to α1,3Gal was highest for blood group A. The degree of CDC against HEK293-pCMAH cells was significantly lower than that against HEK293-pGT cells. However, CDC against HEK293-pCMAH cells was significantly higher than that against control HEK293 cells. In addition, the severity of CDC against HEK293-pCMAH cells positively correlated with that against GTKO pig aortic endothelial cells (PAECs), suggesting that Neu5Gc is the main antigen in GTKO PAECs. Similar to antibody-binding activity, only IgG binding correlated with CDC against HEK293-pCMAH cells. The most common subclass of IgGs against Neu5Gc was IgG1, which typically induces strong complement activation. CONCLUSIONS: We showed that IgG-mediated CDC was detected in Neu5Gc-overexpressed HEK293 cells incubated with human sera; however, this antibody reactivity to Neu5Gc was highly variable among individuals. Our results suggest that additional modifications to the CMAH gene should be considered for widespread use of pig organs for human transplants.


Subject(s)
Galactose/immunology , Graft Rejection/immunology , Immunoglobulin G/immunology , Neuraminic Acids/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Complement Activation/immunology , HEK293 Cells , Humans , Immunoglobulin G/blood , Neuraminic Acids/metabolism , Swine , Transplantation, Heterologous/methods
18.
Semin Immunol ; 24(3): 209-17, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22551764

ABSTRACT

After their development in the thymus, mature T cells are maintained in the periphery by two sets of survival signals, namely TCR signals from contact with self-peptide/MHC ligands and the cytokine receptor signals from binding IL-7 and IL-15. These signals cooperate to maximize the utility of finite resources to support a diverse pool of mature T cells. It is becoming increasingly clear that multiple mechanisms exist to regulate expression of IL-7R at the transcriptional and post-translational levels. The interplay between TCR signals and IL-7R signals are also important in regulation of IL-7R expression. This review will focus on regulation of T cell homeostasis by IL-7R signaling, with an emphasis on the cross talk between signals from TCR and IL-7R.


Subject(s)
Homeostasis , Interleukin-7 Receptor alpha Subunit/immunology , Interleukin-7/immunology , Signal Transduction , T-Lymphocytes/immunology , Animals , Humans , Interleukin-7/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/metabolism
19.
J Virol ; 88(16): 8998-9009, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899182

ABSTRACT

UNLABELLED: T follicular helper (Tfh) cells are specialized providers of cognate B cell help, which is important in promoting the induction of high-affinity antibody production in germinal centers (GCs). Interleukin-6 (IL-6) and IL-21 have been known to play important roles in Tfh cell differentiation. Here, we demonstrate that IL-7 plays a pivotal role in Tfh generation and GC formation in vivo, as treatment with anti-IL-7 neutralizing antibody markedly impaired the development of Tfh cells and IgG responses. Moreover, codelivery of mouse Fc-fused IL-7 (IL-7-mFc) with a vaccine enhanced the generation of GC B cells as well as Tfh cells but not other lineages of T helper cells, including Th1, Th2, and Th17 cells. Interestingly, a 6-fold-lower dose of an influenza virus vaccine codelivered with Fc-fused IL-7 induced higher antigen-specific and cross-reactive IgG titers than the vaccine alone in both mice and monkeys and led to markedly enhanced protection against heterologous influenza virus challenge in mice. Enhanced generation of Tfh cells by IL-7-mFc treatment was not significantly affected by the neutralization of IL-6 and IL-21, indicating an independent role of IL-7 on Tfh differentiation. Thus, IL-7 holds promise as a critical cytokine for selectively inducing Tfh cell generation and enhancing protective IgG responses. IMPORTANCE: Here, we demonstrate for the first time that codelivery of Fc-fused IL-7 significantly increased influenza virus vaccine-induced antibody responses, accompanied by robust expansion of Tfh cells and GC B cells as well as enhanced GC formation. Furthermore, IL-7-mFc induced earlier and cross-reactive IgG responses, leading to striking protection against heterologous influenza virus challenge. These results suggest that Fc-fused IL-7 could be used for inducing strong and cross-protective humoral immunity against highly mutable viruses, such as HIV and hepatitis C virus, as well as influenza viruses.


Subject(s)
Immunity, Humoral/immunology , Interleukin-7/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Female , Germinal Center/immunology , Haplorhini/immunology , Immunoglobulin G/immunology , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae/immunology
20.
Blood ; 121(22): 4484-92, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23610371

ABSTRACT

Interleukin-7 (IL-7) is essential to T-cell survival as well as homeostatic proliferation, and clinical trials that exploit the mitogenic effects of IL-7 have achieved success in treating human diseases. In mice, the in vivo potency of IL-7 improves dramatically when it is administered as a complex with the anti-IL-7 neutralizing monoclonal antibody clone M25. However, the mechanism whereby M25 augments IL-7 potency is unknown. We have analyzed the discrete contributions of the antibody constant (Fc) and IL-7-binding (Fab) domains to the mechanism. By engaging the neonatal Fc receptor the Fc domain extends the in vivo lifespan of IL-7/M25 complexes and accounts for the majority of their activity. Unexpectedly, the IL-7-neutralizing Fab domain provides an additional, albeit smaller, contribution, possibly by serving as a cytokine depot. This study is the first to demonstrate that the neutralizing aspect of the monoclonal antibody is directly involved in enhancing the potency of a cytokine with a single form of receptor. Lessons from the mechanism of IL-7/M25 complexes inform the design of next-generation cytokine therapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Interleukin-7/immunology , Receptors, Interleukin-7/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Benzofurans , Binding, Competitive/immunology , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , Interleukin-7/metabolism , Male , Mice , Mice, Transgenic , Quinolines , Receptors, Fc/immunology , Receptors, Fc/metabolism , Receptors, Interleukin-7/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL