Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 12(4)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684370

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR)-T cell quality and stemness are associated with responsiveness, durability, and memory formation, which benefit clinical responses. Autologous T cell starting material across patients with cancer is variable and CAR-T expansion or potency can fail during manufacture. Thus, strategies to develop allogeneic CAR-T platforms including the identification and expansion of T cell subpopulations that correspond with CAR-T potency are an active area of investigation. Here, we compared CAR-T cells generated from healthy adult peripheral blood T cells versus placental circulating T (P-T) cells. METHODS: CAR-T cells from healthy adult peripheral blood mononuclear cells (PBMCs) and P-T cells were generated using the same protocol. CAR-T cells were characterized in detail by a combination of multiparameter flow cytometry, functional assays, and RNA sequencing. In vivo antitumor efficacy and persistence of CAR-T cells were evaluated in a Daudi lymphoma xenograft model. RESULTS: P-T cells possess stemness advantages compared with T cells from adult PBMCs. P-T cells are uniformly naïve prior to culture initiation, maintain longer telomeres, resist immune checkpoint upregulation, and resist further differentiation compared with PBMC T cells during CD19 CAR-T manufacture. P-T CD19 CAR-T cells are equally cytotoxic as PBMC-CD19 CAR-T cells but produce less interferon gamma in response to lymphoma. Transcriptome analysis shows P-T CD19 CAR-T cells retain a stem-like gene signature, strongly associate with naïve T cells, an early memory phenotype, and a unique CD4 T cell signature compared with PBMC-CD19 CAR-T cells, which enrich for exhaustion and stimulated memory T cell signatures. Consistent with functional data, P-T CD19 CAR-T cells exhibit attenuated inflammatory cytokine and chemokine gene signatures. In a murine in vivo model, P-T CD19 CAR-T cells eliminate lymphoma beyond 90 days. PBMC-CD19 CAR-T cells provide a non-durable benefit, which only delays disease onset. CONCLUSION: We identified characteristics of T cell stemness enriched in P-T CD19 CAR-T which are deficient in PBMC-derived products and translate into response durability in vivo. Our findings demonstrate that placental circulating T cells are a valuable cell source for allogeneic CAR-T products. Stemness advantages inherent to P-T cells translate to in vivo persistence advantages and long-term durable activity.


Subject(s)
Cytokines , Immunotherapy, Adoptive , Leukocytes, Mononuclear , Placenta , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Female , Animals , Mice , Pregnancy , Placenta/immunology , Placenta/metabolism , Cytokines/metabolism , Immunotherapy, Adoptive/methods , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adult , Xenograft Model Antitumor Assays
2.
Front Immunol ; 13: 900624, 2022.
Article in English | MEDLINE | ID: mdl-36341337

ABSTRACT

Influenza A virus (IAV) infections are a significant recurrent threat to public health and a significant burden on global economy, highlighting the need for developing more effective therapies. Natural killer (NK) cells play a pivotal role in the control of pulmonary IAV infection, however, little is known about the therapeutic potential of adoptively transferred NK cells for viral infections. Here, we investigated the antiviral activity of CYNK, human placental hematopoietic stem cell-derived NK cells, against IAV infection in vitro. Virus infection induced the expression of NK cell activating ligands on respiratory epithelial cells, resulting in enhanced recognition by CYNK cells. Upon co-culture with IAV-infected epithelial cells, CYNK exhibited elevated degranulation and increased production of IFN-γ, TNF-α and GM-CSF in a virus dose-dependent manner. Furthermore, CYNK showed virus dose-dependent cytotoxicity against IAV-infected cells. The antiviral activity of CYNK was mediated by NKp46 and NKG2D. Together, these data demonstrate that CYNK possesses potent antiviral function against IAV and warrant clinical investigations for adoptive NK cell therapies against viral infections.


Subject(s)
Influenza A virus , Influenza, Human , Pregnancy , Humans , Female , Placenta , Killer Cells, Natural/metabolism , Influenza, Human/metabolism , Hematopoietic Stem Cells , Antiviral Agents/metabolism
3.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33741730

ABSTRACT

BACKGROUND: Tumors often develop resistance to surveillance by endogenous immune cells, which include natural killer (NK) cells. Ex vivo activated and/or expanded NK cells demonstrate cytotoxicity against various tumor cells and are promising therapeutics for adoptive cancer immunotherapy. Genetic modification can further enhance NK effector cell activity or activation sensitization. Here, we evaluated the effect of the genetic deletion of ubiquitin ligase Casitas B-lineage lymphoma pro-oncogene-b (CBLB), a negative regulator of lymphocyte activity, on placental CD34+ cell-derived NK (PNK) cell cytotoxicity against tumor cells. METHODS: Using CRISPR/Cas9 technology, CBLB was knocked out in placenta-derived CD34+ hematopoietic stem cells, followed by differentiation into PNK cells. Cell expansion, phenotype and cytotoxicity against tumor cells were characterized in vitro. The antitumor efficacy of CBLB knockout (KO) PNK cells was tested in an acute myeloid leukemia (HL-60) tumor model in NOD-scid IL2R gammanull (NSG) mice. PNK cell persistence, biodistribution, proliferation, phenotype and antitumor activity were evaluated. RESULTS: 94% of CBLB KO efficacy was achieved using CRISPR/Cas9 gene editing technology. CBLB KO placental CD34+ cells differentiated into PNK cells with high cell yield and >90% purity determined by CD56+ CD3- cell identity. Ablation of CBLB did not impact cell proliferation, NK cell differentiation or phenotypical characteristics of PNK cells. When compared with the unmodified PNK control, CBLB KO PNK cells exhibited higher cytotoxicity against a range of liquid and solid tumor cell lines in vitro. On infusion into busulfan-conditioned NSG mice, CBLB KO PNK cells showed in vivo proliferation and maturation as evidenced by increased expression of CD16, killer Ig-like receptors and NKG2A over 3 weeks. Additionally, CBLB KO PNK cells showed greater antitumor activity in a disseminated HL60-luciferase mouse model compared with unmodified PNK cells. CONCLUSION: CBLB ablation increased PNK cell effector function and proliferative capacity compared with non-modified PNK cells. These data suggest that targeting CBLB may offer therapeutic advantages via enhancing antitumor activities of NK cell therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Cytotoxicity, Immunologic , Immunotherapy, Adoptive , Killer Cells, Natural/transplantation , Neoplasms/therapy , Proto-Oncogene Proteins c-cbl/deficiency , Stem Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD34/metabolism , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Coculture Techniques , Female , GPI-Linked Proteins/metabolism , Gene Knockout Techniques , HL-60 Cells , Humans , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, Inbred NOD , Mice, SCID , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Phenotype , Placenta/cytology , Pregnancy , Proto-Oncogene Proteins c-cbl/genetics , Receptors, IgG/metabolism , Stem Cells/immunology , Stem Cells/metabolism , Xenograft Model Antitumor Assays
4.
J Clin Invest ; 128(12): 5647-5662, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30352428

ABSTRACT

Tumor-associated myeloid cells maintain immunosuppressive microenvironments within tumors. Identification of myeloid-specific receptors to modulate tumor-associated macrophage and myeloid-derived suppressor cell (MDSC) functions remains challenging. The leukocyte immunoglobulin-like receptor B (LILRB) family members are negative regulators of myeloid cell activation. We investigated how LILRB targeting could modulate tumor-associated myeloid cell function. LILRB2 antagonism inhibited receptor-mediated activation of SHP1/2 and enhanced proinflammatory responses. LILRB2 antagonism also inhibited AKT and STAT6 activation in the presence of M-CSF and IL-4. Transcriptome analysis revealed that LILRB2 antagonism altered genes involved in cell cytoskeleton remodeling, lipid/cholesterol metabolism, and endosomal sorting pathways, as well as changed differentiation gene networks associated with inflammatory myeloid cells as opposed to their alternatively activated phenotype. LILRB2 blockade effectively suppressed granulocytic MDSC and Treg infiltration and significantly promoted in vivo antitumor effects of T cell immune checkpoint inhibitors. Furthermore, LILRB2 blockade polarized tumor-infiltrating myeloid cells from non-small cell lung carcinoma tumor tissues toward an inflammatory phenotype. Our studies suggest that LILRB2 can potentially act as a myeloid immune checkpoint by reprogramming tumor-associated myeloid cells and provoking antitumor immunity.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasm Proteins/immunology , Receptors, Immunologic/immunology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , Cytoskeleton/genetics , Cytoskeleton/immunology , Cytoskeleton/pathology , Gene Regulatory Networks/immunology , Lipid Metabolism/genetics , Lipid Metabolism/immunology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Proteins/genetics , Receptors, Immunologic/genetics
5.
J Immunol ; 201(6): 1727-1734, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30068593

ABSTRACT

Glatiramer acetate (GA; Copaxone) is a copolymer therapeutic that is approved by the Food and Drug Administration for the relapsing-remitting form of multiple sclerosis. Despite an unclear mechanism of action, studies have shown that GA promotes protective Th2 immunity and stimulates release of cytokines that suppress autoimmunity. In this study, we demonstrate that GA interacts with murine paired Ig-like receptor B (PIR-B) on myeloid-derived suppressor cells and suppresses the STAT1/NF-κB pathways while promoting IL-10/TGF-ß cytokine release. In inflammatory bowel disease models, GA enhanced myeloid-derived suppressor cell-dependent CD4+ regulatory T cell generation while reducing proinflammatory cytokine secretion. Human monocyte-derived macrophages responded to GA by reducing TNF-α production and promoting CD163 expression typical of alternative maturation despite the presence of GM-CSF. Furthermore, GA competitively interacts with leukocyte Ig-like receptors B (LILRBs), the human orthologs of PIR-B. Because GA limited proinflammatory activation of myeloid cells, therapeutics that target LILRBs represent novel treatment modalities for autoimmune indications.


Subject(s)
Antigens, CD/immunology , Glatiramer Acetate/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Receptors, Immunologic/immunology , Animals , Antigens, CD/genetics , Autoimmune Diseases/drug therapy , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cytokines/genetics , Cytokines/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid-Derived Suppressor Cells/pathology , NF-kappa B/genetics , NF-kappa B/immunology , Receptors, Immunologic/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th2 Cells/immunology , Th2 Cells/pathology
6.
Cancer Immunol Immunother ; 66(8): 1079-1087, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28638976

ABSTRACT

The leukocyte immunoglobulin-like receptor (LILR) family comprises a set of paired immunomodulatory receptors expressed among human myeloid and lymphocyte cell populations. While six members of LILR subfamily A (LILRA) associate with membrane adaptors to signal via immunoreceptor tyrosine-based activating motifs (ITAM), LILR subfamily B (LILRB) members signal via multiple cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIM). Ligand specificity of some LILR family members has been studied in detail, but new perspective into the immunoregulatory aspects of this receptor family in human myeloid cells has been limited. LILRB receptors and the murine ortholog, paired immunoglobulin-like receptor B (PIRB), have been shown to negatively regulate maturation pathways in myeloid cells including mast cells, neutrophils, dendritic cells, as well as B cells. Our laboratory further demonstrated in mouse models that PIRB regulated functional development of myeloid-derived suppressor cell and the formation of a tumor-permissive microenvironment. Based on observations from the literature and our own studies, our laboratory is focusing on how LILRs modulate immune homeostasis of human myeloid cells and how these pathways may be targeted in disease states. Integrity of this pathway in tumor microenvironments, for example, permits a myeloid phenotype that suppresses antitumor adaptive immunity. This review presents the evidence supporting a role of LILRs as myeloid cell regulators and ongoing efforts to understand the functional immunology surrounding this family.


Subject(s)
Antigens, CD/metabolism , Leukocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , Receptors, Immunologic/metabolism , Animals , Gene Expression Regulation, Neoplastic , Humans , Immunomodulation , Leukocyte Immunoglobulin-like Receptor B1 , Mice , Receptors, Immunologic/genetics , Signal Transduction/immunology , Tumor Microenvironment
7.
Immunity ; 42(6): 1143-58, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26070485

ABSTRACT

Tissue effector cells of the monocyte lineage can differentiate into different cell types with specific cell function depending on their environment. The phenotype, developmental requirements, and functional mechanisms of immune protective macrophages that mediate the induction of transplantation tolerance remain elusive. Here, we demonstrate that costimulatory blockade favored accumulation of DC-SIGN-expressing macrophages that inhibited CD8(+) T cell immunity and promoted CD4(+)Foxp3(+) Treg cell expansion in numbers. Mechanistically, that simultaneous DC-SIGN engagement by fucosylated ligands and TLR4 signaling was required for production of immunoregulatory IL-10 associated with prolonged allograft survival. Deletion of DC-SIGN-expressing macrophages in vivo, interfering with their CSF1-dependent development, or preventing the DC-SIGN signaling pathway abrogated tolerance. Together, the results provide new insights into the tolerogenic effects of costimulatory blockade and identify DC-SIGN(+) suppressive macrophages as crucial mediators of immunological tolerance with the concomitant therapeutic implications in the clinic.


Subject(s)
Cell Adhesion Molecules/metabolism , Graft Rejection/prevention & control , Heart Transplantation , Lectins, C-Type/metabolism , Macrophages/immunology , Receptors, Cell Surface/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/genetics , Cells, Cultured , Forkhead Transcription Factors/metabolism , Graft Rejection/etiology , Immune Tolerance , Interleukin-10/metabolism , Lectins, C-Type/genetics , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Receptors, Cell Surface/genetics , Signal Transduction , Toll-Like Receptor 4/metabolism , Transplantation Tolerance , Up-Regulation
8.
Semin Nephrol ; 33(6): 565-74, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24161041

ABSTRACT

Complement proteins are generated both by the liver (systemic compartment) and by peripheral tissue-resident cells and migratory immune cells (local compartment). The immune cell-derived, alternative pathway complement components activate spontaneously, yielding local, but not systemic, production of C3a and C5a. These anaphylatoxins bind to their respective G-protein-coupled receptors, the C3a receptor and the C5a receptor, expressed on T cells and antigen-presenting cells, leading to their reciprocal activation and driving T-cell differentiation, expansion, and survival. Complement deficiency or blockade attenuates T-cell-mediated autoimmunity and delays allograft rejection in mice. Increasing complement activation, achieved by genetic removal of the complement regulatory protein decay accelerating factor, enhances murine T-cell immunity and accelerates allograft rejection. Signaling through the C3a receptor and the C5a receptor reduces suppressive activity of natural regulatory T cells and the generation and stability of induced regulatory T cells. The concepts, initially generated in mice, recently were confirmed in human immune cells, supporting the need for testing of complement targeting therapies in organ transplants patients.


Subject(s)
Antigen-Presenting Cells/immunology , Autoimmunity/physiology , Complement System Proteins/metabolism , Graft Rejection/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Complement Activation/physiology , Complement Inactivating Agents/pharmacology , Graft Rejection/drug therapy , Graft Rejection/metabolism , Humans , Mice , T-Lymphocytes/metabolism
9.
J Immunol ; 190(12): 5921-5, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23690475

ABSTRACT

CD4(+)Foxp3(+) regulatory T cells (Treg) are critical regulators of immune homeostasis and self-tolerance. Whereas thymic-derived or natural Treg stably express Foxp3, adaptive or induced Treg (iTreg) generated from peripheral CD4 T cells are susceptible to inflammation-induced reversion to pathogenic effector T cells. Building upon our previous observations that T cell-expressed receptors for C3a (C3aR) and C5a (C5aR) drive Th1 maturation, we tested the impact of C3aR/C5aR signaling on induction and stability of alloreactive iTreg. We observed that genetic deficiency or pharmacological blockade of C3aR/C5aR signaling augments murine and human iTreg generation, stabilizes Foxp3 expression, resists iTreg conversion to IFN-γ/TNF-α-producing efffector T cells, and, as a consequence, limits the clinical expression of graft-versus-host disease. Taken together, the findings highlight the expansive role of complement as a crucial modulator of T cell alloimmunity and demonstrate proof-of-concept that targeting C3a/C3aR and C5a/C5aR interactions could facilitate iTreg-mediated tolerance to alloantigens in humans.


Subject(s)
Complement C3a/immunology , Isoantigens , Lymphocyte Activation/immunology , Receptor, Anaphylatoxin C5a/immunology , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Flow Cytometry , Gene Knockdown Techniques , Humans , Immunoblotting , Isoantigens/immunology , Mice , Mice, Knockout , T-Lymphocyte Subsets/immunology
10.
J Exp Med ; 210(2): 257-68, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23382542

ABSTRACT

Thymus-derived (natural) CD4(+) FoxP3(+) regulatory T cells (nT reg cells) are required for immune homeostasis and self-tolerance, but must be stringently controlled to permit expansion of protective immunity. Previous findings linking signals transmitted through T cell-expressed C5a receptor (C5aR) and C3a receptor (C3aR) to activation, differentiation, and expansion of conventional CD4(+)CD25(-) T cells (T conv cells), raised the possibility that C3aR/C5aR signaling on nT reg cells could physiologically modulate nT reg cell function and thereby further impact the induced strength of T cell immune responses. In this study, we demonstrate that nT reg cells express C3aR and C5aR, and that signaling through these receptors inhibits nT reg cell function. Genetic and pharmacological blockade of C3aR/C5aR signal transduction in nT reg cells augments in vitro and in vivo suppression, abrogates autoimmune colitis, and prolongs allogeneic skin graft survival. Mechanisms involve C3a/C5a-induced phosphorylation of AKT and, as a consequence, phosphorylation of the transcription factor Foxo1, which results in lowered nT reg cell Foxp3 expression. The documentation that C3a/C3aR and C5a/C5aR modulate nT reg cell function via controlling Foxp3 expression suggests targeting this pathway could be exploited to manipulate pathogenic or protective T cell responses.


Subject(s)
Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Colitis/immunology , Colitis/pathology , Colitis/prevention & control , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression , Graft Rejection/immunology , Graft Rejection/pathology , Graft Rejection/prevention & control , Immunity, Innate , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Receptors, Complement/deficiency , Receptors, Complement/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/immunology , Skin Transplantation/immunology , Skin Transplantation/pathology , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
11.
Transplantation ; 94(6): 575-84, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22914174

ABSTRACT

BACKGROUND: The role of natural killer (NK) cells in organ transplantation is poorly understood because studies link these cells to both regulatory and inflammatory functions. NK cells exacerbate inflammation and adaptive immunity under conditions of allograft rejection, but little is known regarding their roles in allograft tolerance. We test the hypothesis that NK cells have regulatory function and promote tolerance induction to murine cardiac allografts. METHODS: Murine hearts were transplanted as fully vascularized heterotopic grafts from BALB/c donors into C57BL/6 recipients. Allograft tolerance was achieved using donor splenocyte transfusion + anti-CD40L monoclonal antibody (mAb) before transplantation. The requirement for NK cells in tolerance induction was tested by administering anti-NK1.1-depleting mAb or anti-NKG2D-blocking mAb. Intragraft and peripheral immune cell populations were determined by flow cytometry and immunohistochemistry. CD4 T-cell alloantigen-specific responses and donor-specific alloantibody were also determined. RESULTS: NK cell-depleted recipients acutely reject allografts despite anti-CD40L blockade, but rejecting recipients lacked alloantibody and alloantigen-specific CD4 T-cell responses. NK cell depletion resulted in elevated numbers of graft-infiltrating macrophages. NKG2D blockade in tolerized recipients did not cause acute rejection but increased macrophage graft infiltration and increased the expression of NKG2D ligand Rae-1γ on these cells. CONCLUSIONS: Our data show that NK cells are required for tolerance induction in recipients given donor splenocyte transfusion + anti-CD40L mAb. Our data suggest NK cells regulate monocyte or macrophage activation and infiltration into allografts by a mechanism partially dependent on NKG2D receptor-ligand interactions between NK cells and monocytes/macrophages.


Subject(s)
Graft Rejection/prevention & control , Graft Survival , Heart Transplantation/immunology , Killer Cells, Natural/immunology , Transplantation Tolerance , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Antigens, Ly/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/immunology , Flow Cytometry , Genes, T-Cell Receptor , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Survival/drug effects , Immunohistochemistry , Immunosuppressive Agents/administration & dosage , Isoantibodies/blood , Isoantigens/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/transplantation , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , NK Cell Lectin-Like Receptor Subfamily B/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Spleen/immunology , Time Factors , Transplantation Tolerance/drug effects
12.
Immunol Res ; 54(1-3): 247-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22477527

ABSTRACT

Results of studies published since 2002 reveal that T cells and antigen-presenting cells (APCs) produce complement proteins. The immune cell-derived, alternative pathway complement components activate spontaneously, yielding local, but not systemic, production of C3a and C5a. These anaphylatoxins bind to their respective G-protein-coupled receptors, C3aR and C5aR, expressed on both partners. The resultant complement-induced T cell activation and APC activation drive T cell differentiation, expansion and survival. Complement deficiency or blockade attenuates T cell-mediated autoimmunity and delays allograft rejection in mice. Increasing complement activation, achieved by genetic removal of the complement regulatory protein decay-accelerating factor, enhances murine T cell immunity and accelerates allograft rejection. The findings support the need for design and testing of complement inhibitors in humans.


Subject(s)
Complement System Proteins/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Humans
13.
Methods Mol Biol ; 677: 127-47, 2011.
Article in English | MEDLINE | ID: mdl-20941607

ABSTRACT

Dendritic cells (DC) are professional antigen-presenting cells (APCs) that modulate the outcome of the immune response toward immunity or tolerance. There are a large variety of DC subsets according to surface phenotype, function, and tissue distribution. Murine plasmacytoid DC (pDC) represent a distinctive DC population and are characterized by the expression of CD11c, B220, Gr-1, CD45RA, Ly49Q, BST2, and siglec-H on the cell surface. PDC act as immunogenic cell sentinels by secreting large amounts of type I interferon (IFN) in the lymph nodes in response to viral stimulation. PDC also act as tolerogenic cells when expressing the inducible tolerogenic enzyme indoleamine 2,3-dioxygenase (IDO), the inducible costimulator ligand (ICOS-L), and/or the programmed death 1 ligand (PD-L1), which mediate regulatory T-cell (Treg) development and suppression of self- and alloreactive cells. The PDC ability to induce Treg development is associated with capture and presentation of antigenic peptides associated with major histocompatibility complex (MHC) class I and II. Here, we provide the tools to study PDC development from bone marrow cultures, their antigen presentation properties, and their interactions with Treg under a tolerogenic setting of sterile inflammation.


Subject(s)
Bone Marrow/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Cell Differentiation/physiology , Cells, Cultured , Flow Cytometry , Homeostasis/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Spleen/cytology
14.
J Immunol ; 182(1): 259-73, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109157

ABSTRACT

Foxp3, a winged-helix family transcription factor, serves as the master switch for CD4(+) regulatory T cells (Treg). We identified a unique and evolutionarily conserved CpG-rich island of the Foxp3 nonintronic upstream enhancer and discovered that a specific site within it was unmethylated in natural Treg (nTreg) but heavily methylated in naive CD4(+) T cells, activated CD4(+) T cells, and peripheral TGFbeta-induced Treg in which it was bound by DNMT1, DNMT3b, MeCP2, and MBD2. Demethylation of this CpG site using the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (Aza) induced acetylation of histone 3, interaction with TIEG1 and Sp1, and resulted in strong and stable induction of Foxp3. Conversely, IL-6 resulted in methylation of this site and repression of Foxp3 expression. Aza plus TGFbeta-induced Treg resembled nTreg, expressing similar receptors, cytokines, and stable suppressive activity. Strong Foxp3 expression and suppressor activity could be induced in a variety of T cells, including human CD4(+)CD25(-) T cells. Epigenetic regulation of Foxp3 can be predictably controlled with DNMT inhibitors to generate functional, stable, and specific Treg.


Subject(s)
DNA Methylation/immunology , Epigenesis, Genetic/immunology , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/prevention & control , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Azacitidine/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Cells, Cultured , Colitis/immunology , Colitis/pathology , Colitis/prevention & control , DNA Methylation/drug effects , Decitabine , Female , Forkhead Transcription Factors/physiology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...