Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 28837, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27354027

ABSTRACT

Invariant NKT (iNKT) cells are critical to the maintenance of tolerance toward alloantigens encountered during postnatal life pointing to the existence of a process for self-education. However, the impact of developmentally encountered alloantigens in shaping the phenotype and function of iNKT cells has not been described. To better understand this process, the current report examined naïve iNKT cells as they matured in an allogeneic environment. Following the prenatal transfer of fetal hematopoietic cells between age-matched allogeneic murine fetuses, cell-extrinsic signals appeared to dictate allospecific patterns of Ly49 receptor expression and lineage diversity in developing iNKT cells. Regulation for this process arose from cells of hematopoietic origin requiring only rare exposure to facilitate broad changes in developing iNKT cells. These findings highlight surprisingly asymmetric allospecific alterations in iNKT cells as they develop and mature in an allogeneic environment and establish a new paradigm for study of the self-education of iNKT cells.


Subject(s)
Natural Killer T-Cells/physiology , Animals , Cell Lineage , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Natural Killer T-Cells/transplantation , Transplantation Tolerance
2.
Chimerism ; 6(1-2): 8-20, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-26517600

ABSTRACT

Compulsory exposure to genetically foreign maternal tissue imprints in offspring sustained tolerance to noninherited maternal antigens (NIMA). Immunological tolerance to NIMA was first described by Dr. Ray D. Owen for women genetically negative for erythrocyte rhesus (Rh) antigen with reduced sensitization from developmental Rh exposure by their mothers. Extending this analysis to HLA haplotypes has uncovered the exciting potential for therapeutically exploiting NIMA-specific tolerance naturally engrained in mammalian reproduction for improved clinical outcomes after allogeneic transplantation. Herein, we summarize emerging scientific concepts stemming from tolerance to NIMA that includes postnatal maintenance of microchimeric maternal origin cells in offspring, expanded accumulation of immune suppressive regulatory T cells with NIMA-specificity, along with teleological benefits and immunological consequences of NIMA-specific tolerance conserved across mammalian species.


Subject(s)
HLA Antigens/immunology , Immune Tolerance , Immunologic Memory , Rh-Hr Blood-Group System/immunology , T-Lymphocytes, Regulatory/immunology , Animals , HLA Antigens/history , History, 20th Century , History, 21st Century , Humans , Rh-Hr Blood-Group System/history
3.
J Immunol ; 195(8): 4001-9, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26363051

ABSTRACT

Transplanting stem cells before birth offers an unparalleled opportunity to initiate corrective treatment for numerous childhood diseases with minimal or no host conditioning. Although long-term engraftment has been demonstrated following in utero hematopoietic cellular transplantation during immune quiescence, it is unclear if prenatal tolerance becomes unstable with immune activation such as during a viral syndrome. Using a murine model of in utero hematopoietic cellular transplantation, the impact of an infection with lymphocytic choriomeningitis virus on prenatal allospecific tolerance was examined. The findings in this report illustrate that established mechanisms of donor-specific tolerance are strained during potent immune activation. Specifically, a transient reversal in the anergy of alloreactive lymphocytes is seen in parallel with the global immune response toward the virus. However, these changes return to baseline following resolution of the infection. Importantly, prenatal engraftment remains stable during and after immune activation. Collectively, these findings illustrate the robust nature of allospecific tolerance in prenatal mixed chimerism compared with models of postnatal chimerism and provides additional support for the prenatal approach to the treatment of congenital benign cellular disease.


Subject(s)
Hematopoietic Stem Cell Transplantation , Transplantation Chimera/immunology , Transplantation Tolerance , Allografts , Animals , Female , Fetal Diseases/immunology , Fetal Diseases/therapy , Mice , Pregnancy
4.
Cell ; 162(3): 505-15, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26213383

ABSTRACT

Exposure to maternal tissue during in utero development imprints tolerance to immunologically foreign non-inherited maternal antigens (NIMA) that persists into adulthood. The biological advantage of this tolerance, conserved across mammalian species, remains unclear. Here, we show maternal cells that establish microchimerism in female offspring during development promote systemic accumulation of immune suppressive regulatory T cells (Tregs) with NIMA specificity. NIMA-specific Tregs expand during pregnancies sired by males expressing alloantigens with overlapping NIMA specificity, thereby averting fetal wastage triggered by prenatal infection and non-infectious disruptions of fetal tolerance. Therefore, exposure to NIMA selectively enhances reproductive success in second-generation females carrying embryos with overlapping paternally inherited antigens. These findings demonstrate that genetic fitness, canonically thought to be restricted to Mendelian inheritance, is enhanced in female placental mammals through vertically transferred maternal cells that promote conservation of NIMA and enforce cross-generational reproductive benefits.


Subject(s)
Fetus/immunology , Genetic Fitness , Immune Tolerance , Mammals/physiology , Pregnancy/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Chimerism , Female , Humans , Male , Mammals/immunology , Mice , Placenta/immunology
5.
J Immunol ; 195(4): 1506-16, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26136432

ABSTRACT

Little is known about how the prenatal interaction between NK cells and alloantigens shapes the developing NK cell repertoire toward tolerance or immunity. Specifically, the effect on NK cell education arising from developmental corecognition of alloantigens by activating and inhibitory receptors with shared specificity is uncharacterized. Using a murine prenatal transplantation model, we examined the manner in which this seemingly conflicting input affects NK cell licensing and repertoire formation in mixed hematopoietic chimeras. We found that prenatal NK cell tolerance arose from the elimination of phenotypically hostile NK cells that express an allospecific activating receptor without coexpressing any allospecific inhibitory receptors. Importantly, the checkpoint for the system appeared to occur centrally within the bone marrow during the final stage of NK cell maturation and hinged on the instructive recognition of allogeneic ligand by the activating receptor rather than through the inhibitory receptor as classically proposed. Residual nondeleted hostile NK cells expressing only the activating receptor exhibited an immature, anergic phenotype, but retained the capacity to upregulate inhibitory receptor expression in peripheral sites. However, the potential for this adaptive change to occur was lost in developmentally mature chimeras. Collectively, these findings illuminate the intrinsic process in which developmental allorecognition through the activating receptor regulates the emergence of durable NK cell tolerance and establishes a new paradigm to fundamentally guide future investigations of prenatal NK cell-allospecific education.


Subject(s)
Immune Tolerance , Isoantigens/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation , Receptors, Immunologic/metabolism , Adoptive Transfer , Animals , Bone Marrow Transplantation , Clonal Anergy/genetics , Clonal Anergy/immunology , Graft Rejection/immunology , H-2 Antigens/immunology , Homeostasis , Immunophenotyping , Killer Cells, Natural/cytology , Mice , Models, Animal , Phenotype , Transplantation Chimera
6.
Front Pharmacol ; 6: 51, 2015.
Article in English | MEDLINE | ID: mdl-25852555

ABSTRACT

The primary benefits of in utero hematopoietic cellular transplantation (IUHCT) arise from transplanting curative cells prior to the immunologic maturation of the fetus. However, this approach has been routinely successful only in the treatment of congenital immunodeficiency diseases that include an inherent NK cell deficiency despite the existence of normal maternal immunity in either setting. These observations raise the possibility that fetal NK cells function as an early barrier to allogeneic IUHCT. Herein, we summarize the findings of previous studies of prenatal NK cell allospecific tolerance in mice and in humans. Cumulatively, this new information reveals the complexity of the fetal immune response in the setting of rejection or tolerance and illustrates the role for fetal NK cells in the final endorsement of allospecific prenatal tolerance.

7.
Nature ; 504(7478): 158-62, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24196717

ABSTRACT

Newborn infants are highly susceptible to infection. This defect in host defence has generally been ascribed to the immaturity of neonatal immune cells; however, the degree of hyporesponsiveness is highly variable and depends on the stimulation conditions. These discordant responses illustrate the need for a more unified explanation for why immunity is compromised in neonates. Here we show that physiologically enriched CD71(+) erythroid cells in neonatal mice and human cord blood have distinctive immunosuppressive properties. The production of innate immune protective cytokines by adult cells is diminished after transfer to neonatal mice or after co-culture with neonatal splenocytes. Neonatal CD71(+) cells express the enzyme arginase-2, and arginase activity is essential for the immunosuppressive properties of these cells because molecular inhibition of this enzyme or supplementation with L-arginine overrides immunosuppression. In addition, the ablation of CD71(+) cells in neonatal mice, or the decline in number of these cells as postnatal development progresses parallels the loss of suppression, and restored resistance to the perinatal pathogens Listeria monocytogenes and Escherichia coli. However, CD71(+) cell-mediated susceptibility to infection is counterbalanced by CD71(+) cell-mediated protection against aberrant immune cell activation in the intestine, where colonization with commensal microorganisms occurs swiftly after parturition. Conversely, circumventing such colonization by using antimicrobials or gnotobiotic germ-free mice overrides these protective benefits. Thus, CD71(+) cells quench the excessive inflammation induced by abrupt colonization with commensal microorganisms after parturition. This finding challenges the idea that the susceptibility of neonates to infection reflects immune-cell-intrinsic defects and instead highlights processes that are developmentally more essential and inadvertently mitigate innate immune protection. We anticipate that these results will spark renewed investigation into the need for immunosuppression in neonates, as well as improved strategies for augmenting host defence in this vulnerable population.


Subject(s)
Antigens, CD/metabolism , Erythroid Cells/immunology , Escherichia coli Infections/immunology , Immune Tolerance/immunology , Listeriosis/immunology , Receptors, Transferrin/metabolism , Animals , Animals, Newborn , Arginase/genetics , Arginase/metabolism , Disease Susceptibility/immunology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Erythroid Cells/enzymology , Escherichia coli/immunology , Female , Fetal Blood/cytology , Humans , Immune Tolerance/drug effects , Immune Tolerance/genetics , Listeria monocytogenes/immunology , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
8.
Chimerism ; 4(4): 126-31, 2013.
Article in English | MEDLINE | ID: mdl-24121538

ABSTRACT

In utero hematopoietic cellular transplantation (IUHCT) holds great promise for the treatment of congenital diseases of cellular dysfunction such as sickle cell disease, immunodeficiency disorders and inherited metabolic disorders. However, repeated failures in clinical cases of IUHCT that do not involve an immunodeficiency disease force a closer examination of the fetal immune system. While the mechanisms regulating T cell tolerance have been previously studied, the educational mechanisms leading to NK cell tolerance in prenatal chimeras remain unknown. As a low level of donor cells (1.8%) is required to induce and maintain this tolerance, it is likely that these mechanisms employ indirect host-donor interaction. This report examines donor-to-host MHC transfer (trogocytosis) as an intrinsic mechanism regulating the development and maintenance of NK cell tolerance in prenatal chimeras. The findings demonstrate that phenotypically tolerant host NK cells express low levels of transferred donor MHC antigens during development and later as mature cytotoxic lymphocytes. Further study is needed to understand how the cis-recognition of transferred donor MHC ligand influences the selection and maintenance of tolerant NK cells in prenatal chimeras.


Subject(s)
Chimerism/embryology , Embryo, Mammalian/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Immune Tolerance , Transplantation Chimera/immunology , Animals , Female , Fetus/immunology , Graft vs Host Reaction/immunology , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Pregnancy
9.
J Immunol ; 187(5): 2193-201, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21788443

ABSTRACT

In APCs, presentation by MHC II molecules of the chemically dominant peptide from the protein hen egg white lysozyme (HEL) generates different conformational isomers of the peptide-MHC II complexes (pMHC). Type B pMHCs are formed in early endosomes from exogenous peptides in the absence of H2-DM, whereas in contrast, type A pMHC complexes are formed from HEL protein in late vesicles after editing by H2-DM. Thus, H2-DM edits off the more unstable pMHC complexes, which are not presented from HEL. In this study, we show that type B pMHC complexes were presented from HEL protein only after stimulation of dendritic cells (DC) with TLR ligands or type I IFN. Type I IFN contributed to most TLR ligand-induced type B pMHC generation, as presentation decreased in DC lacking the receptor for type I IFNs (IFNAR1(-/-)). In contrast, presentation of type A pMHC from HEL and from peptide was minimally affected by TLR ligands. The relative effectiveness of CD8α(+) DC or CD8α(-) DC in presenting type B pMHC complexes varied depending on the TLR ligand used. The mechanisms of generation of type B pMHC from HEL protein with TLR stimulation did not involve H2-DM or release of peptides. DC from H2-DM-deficient mice in the presence of TLR ligands presented type B pMHC. Such DC showed a slight enhancement of HEL catabolism, but peptide release was not evident. Thus, TLR ligands and type I IFN alter the pathways of presentation by MHC II molecules of DC such that type B pMHCs are generated from protein Ag.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Interferon Type I/immunology , Muramidase/immunology , Animals , Cell Separation , Flow Cytometry , Ligands , Major Histocompatibility Complex/immunology , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/immunology
10.
J Immunol ; 185(5): 2927-34, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20679530

ABSTRACT

Neutrophils modulated Ag presentation following immunization with Ags in CFA or IFA or alum. The neutrophils had an important negative role in the CD4 T cell and B cell responses to three protein Ags: hen egg white lysozyme, OVA, and listeriolysin O. In their absence (by depleting with Abs for only the first 24 h, or using genetically neutropenic mice), the cellular responses increased several-fold. The CD8 response was not affected or slightly decreased. Competition for Ag between the presenting cells and the neutrophils, as well as an effect on the response to Ag-bearing dendritic cells (DCs), was documented. Neutrophils entered the draining lymph nodes rapidly and for a brief period of several hours, localizing mainly to the marginal sinus and superficial cortex. There they established brief contact with DCs and macrophages. Moreover, neutrophils imprinted on the quality of the subsequent DC-T cell interactions, despite no physical contact with them; by intravital microscopy, the clustering of Ag-specific T cells and DCs was improved in neutropenic mice. Thus, neutrophils are obligate cells that briefly enter sites of immunization and set the level of Ag presentation. A brief depletion may have a considerably positive impact on vaccination.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Antigen Presentation/immunology , Freund's Adjuvant/administration & dosage , Lipids/administration & dosage , Muramidase/immunology , Neutrophils/immunology , Neutrophils/metabolism , Adjuvants, Immunologic/physiology , Amino Acid Sequence , Animals , Antigen Presentation/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Toxins/administration & dosage , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Cell Movement/genetics , Cell Movement/immunology , Freund's Adjuvant/immunology , Heat-Shock Proteins/administration & dosage , Heat-Shock Proteins/immunology , Heat-Shock Proteins/metabolism , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/immunology , Hemolysin Proteins/metabolism , Lipids/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Muramidase/administration & dosage , Muramidase/metabolism , Neutrophils/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...