Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 8054, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33850164

ABSTRACT

Cromolyn is a known mast cell stabilizer and is approved for treatment of asthma and for other allergic indications. Cromolyn, in a new redesigned dry powder formulation, is being tested in a pivotal clinical trial in combination with low dose ibuprofen to treat early Alzheimer's Disease (AD) subjects. To better understand the mechanistic effect cromolyn has in slowing down or halting the neuroinflammatory response associated with AD progression, we tested the effect of cromolyn to dampen the inflammatory response in the human HMC3 microglia cell line. The direct effect of cromolyn on HMC3 microglia is on cytokines and chemokines production following their activation by the inflammatory cytokine TNF-α. Cromolyn and a new fluorinated analog dramatically reduced the secretion of a wide spectrum of inflammatory mediators, which included cytokines such as IL-1ß, IL-6, IL-8 and IFN-γ, and chemokines such as CXCL10, CCL2, CCL3 and CCL4. These results bolster our understanding of how our cromolyn platform modulates toxic microglia behavior as a dynamic future treatment option for neurodegenerative disorders.


Subject(s)
Cromolyn Sodium , Microglia , Alzheimer Disease/metabolism , Cytokines/metabolism , Inflammation/metabolism
2.
J Allergy Clin Immunol ; 143(3): 1131-1142.e4, 2019 03.
Article in English | MEDLINE | ID: mdl-30053528

ABSTRACT

BACKGROUND: Grass pollen-specific immunotherapy involves immunomodulation of allergen-specific TH2 responses and induction of IL-10+ and/or TGF-ß+CD4+CD25+ regulatory T cells (induced Treg cells). IL-35+CD4+CD25+ forkhead box protein 3-negative T (IL-35-inducible regulatory T [iTR35]) cells have been reported as a novel subset of induced Treg cells with modulatory characteristics. OBJECTIVE: We sought to investigate mechanisms underlying the induction and maintenance of immunologic tolerance induced by IL-35 and iTR35 cells. METHODS: The biological effects of IL-35 were assessed on group 2 innate lymphoid cells (ILC2s); dendritic cells primed with thymic stromal lymphopoietin, IL-25, and IL-33; and B and TH2 cells by using flow cytometry and quantitative RT-PCR. Grass pollen-driven TH2 cell proliferation and cytokine production were measured by using tritiated thymidine and Luminex MagPix, respectively. iTR35 cells were quantified in patients with grass pollen allergy (seasonal allergic rhinitis [SAR] group, n = 16), sublingual immunotherapy (SLIT)-treated patients (SLIT group, n = 16), and nonatopic control subjects (NACs; NAC group, n = 16). RESULTS: The SAR group had increased proportions of ILC2s (P = .002) and IL-5+ cells (P = .042), IL-13+ cells (P = .042), and IL-5+IL-13+ ILC2s (P = .003) compared with NACs. IL-35 inhibited IL-5 and IL-13 production by ILC2s in the presence of IL-25 or IL-33 (P = .031) and allergen-driven TH2 cytokines by effector T cells. IL-35 inhibited CD40 ligand-, IL-4-, and IL-21-mediated IgE production by B cells (P = .015), allergen-driven T-cell proliferation (P = .001), and TH2 cytokine production mediated by primed dendritic cells. iTR35 cells suppressed TH2 cell proliferation and cytokine production. In addition, allergen-driven IL-35 levels and iTR35 cell counts were increased in patients receiving SLIT (all, P < .001) and NACs (all, P < .001) compared with patients with SAR. CONCLUSION: IL-35 and iTR35 cells are potential novel immune regulators induced by SLIT. The clinical relevance of SLIT can be underscored by restoration of protective iTR35 cells.


Subject(s)
Allergens/immunology , Interleukins/immunology , Lymphocytes/immunology , Poaceae/immunology , Pollen/immunology , Rhinitis, Allergic, Seasonal/therapy , Sublingual Immunotherapy , Adult , Female , Humans , Immune Tolerance , Male , Middle Aged , Rhinitis, Allergic, Seasonal/immunology , Young Adult
3.
Science ; 360(6388): 558-563, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29724957

ABSTRACT

Deficiency of C1q, the initiator of the complement classical pathway, is associated with the development of systemic lupus erythematosus (SLE). Explaining this association in terms of abnormalities in the classical pathway alone remains problematic because C3 deficiency does not predispose to SLE. Here, using a mouse model of SLE, we demonstrate that C1q, but not C3, restrains the response to self-antigens by modulating the mitochondrial metabolism of CD8+ T cells, which can themselves propagate autoimmunity. C1q deficiency also triggers an exuberant effector CD8+ T cell response to chronic viral infection leading to lethal immunopathology. These data establish a link between C1q and CD8+ T cell metabolism and may explain how C1q protects against lupus, with implications for the role of viral infections in the perpetuation of autoimmunity.


Subject(s)
Autoimmunity/immunology , CD8-Positive T-Lymphocytes/metabolism , Complement C1q/physiology , Lupus Erythematosus, Systemic/immunology , Lymphocytic Choriomeningitis/immunology , Animals , Autoantibodies/immunology , Autoimmunity/genetics , Complement C1q/genetics , Complement C3/genetics , Complement C3/physiology , Complement Pathway, Classical/genetics , Complement Pathway, Classical/immunology , Disease Models, Animal , Immunoglobulins/immunology , Immunologic Memory/immunology , Lupus Erythematosus, Systemic/genetics , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Mutant Strains
4.
J Allergy Clin Immunol ; 142(3): 904-913.e3, 2018 09.
Article in English | MEDLINE | ID: mdl-29355678

ABSTRACT

BACKGROUND: Mutations in the perforin 1 (PRF1) gene account for up to 58% of familial hemophagocytic lymphohistiocytosis syndromes. The resulting defects in effector cell cytotoxicity lead to hypercytokinemia and hyperactivation with inflammation in various organs. OBJECTIVE: We sought to determine whether autologous gene-corrected T cells can restore cytotoxic function, reduce disease activity, and prevent hemophagocytic lymphohistiocytosis (HLH) symptoms in in vivo models. METHODS: We developed a gammaretroviral vector to transduce murine CD8 T cells in the Prf-/- mouse model. To verify functional correction of Prf-/- CD8 T cells in vivo, we used a lymphocytic choriomeningitis virus (LCMV) epitope-transfected murine lung carcinoma cell tumor model. Furthermore, we challenged gene-corrected and uncorrected mice with LCMV. One patient sample was transduced with a PRF1-encoding lentiviral vector to study restoration of cytotoxicity in human cells. RESULTS: We demonstrated efficient engraftment and functional reconstitution of cytotoxicity after intravenous administration of gene-corrected Prf-/- CD8 T cells into Prf-/- mice. In the tumor model infusion of Prf-/- gene-corrected CD8 T cells eliminated the tumor as efficiently as transplantation of wild-type CD8 T cells. Similarly, mice reconstituted with gene-corrected Prf-/- CD8 T cells displayed complete protection from the HLH phenotype after infection with LCMV. Patients' cells showed correction of cytotoxicity in human CD8 T cells after transduction. CONCLUSION: These data demonstrate the potential application of T-cell gene therapy in reconstituting cytotoxic function and protection against HLH in the setting of perforin deficiency.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Lymphocytic Choriomeningitis/therapy , Lymphohistiocytosis, Hemophagocytic/therapy , Perforin/genetics , Animals , Cell Line, Tumor , Child, Preschool , Genetic Therapy , Humans , Lymphocytic choriomeningitis virus , Male , Mice, Inbred C57BL , Mice, Knockout
5.
J Allergy Clin Immunol ; 142(2): 569-581.e5, 2018 08.
Article in English | MEDLINE | ID: mdl-29106998

ABSTRACT

BACKGROUND: The mechanisms that regulate maintenance of persistent TH2 cells and potentiate allergic inflammation are not well understood. OBJECTIVE: The function of serine protease inhibitor 2A (Spi2A) was studied in mouse TH2 cells, and the serine protease inhibitor B3 (SERPINB3) and SERPINB4 genes were studied in TH2 cells from patients with grass pollen allergy. METHODS: Spi2A-deficient TH2 cells were studied in in vitro culture or in vivo after challenge of Spi2A knockout mice with ovalbumin in alum. Expression of SERPINB3 and SERPINB4 mRNA was measured in in vitro-cultured TH2 cells and in ex vivo CD27-CD4+ cells and innate lymphoid cell (ILC) 2 from patients with grass pollen allergy by using quantitative PCR. SERPINB3 and SERPINB4 mRNA levels were knocked down in cultured CD27-CD4+ cells with small hairpin RNA. RESULTS: There were lower levels of in vitro-polarized TH2 cells from Spi2A knockout mice (P < .005) and in vivo after ovalbumin challenge (P < .05), higher levels of apoptosis (Annexin V positivity, P < .005), and less lung allergic inflammation (number of lung eosinophils, P < .005). In vitro-polarized TH2 cells from patients with grass pollen allergy expressed higher levels of both SERPINB3 and SERPINB4 mRNA (both P < .05) compared with unpolarized CD4 T cells. CD27-CD4+ from patients with grass pollen allergy expressed higher levels of both SERPINB3 and SERPINB4 mRNA (both P < .0005) compared with CD27+CD4+ cells. ILC2 expressed higher levels of both SERPINB3 and SERPINB4 mRNA (both P < .0005) compared with ILC1. Knockdown of either SERPINB3 or SERPINB4 mRNA (both P < .005) levels resulted in decreased viability of CD27-CD4+ compared with control transduced cells. CONCLUSION: The Serpins Spi2A in mice and SERPINB3 and SERPINB4 in allergic patients control the viability of TH2 cells. This provides proof of principle for a therapeutic approach for allergic disease through ablation of allergic memory TH2 cells through SERPINB3 and SERPINB4 mRNA downregulation.


Subject(s)
Antigens, Neoplasm/metabolism , Hypersensitivity/immunology , Inflammation Mediators/metabolism , Serpins/metabolism , Th2 Cells/immunology , Adult , Allergens/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Plant/immunology , Cell Survival , Cells, Cultured , Disease Models, Animal , Female , Humans , Immunologic Memory , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Poaceae/immunology , Pollen/immunology , RNA, Small Interfering/genetics , Serpins/genetics , Young Adult
6.
Cell Metab ; 23(6): 967-968, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27304497

ABSTRACT

The development and function of cytotoxic CD8 T cells (CTLs), which provide immunity to viral infections, are regulated by changes in mitochondrial respiration. Champagne et al. (2016) describe a new mechanism through which mitochondrial metabolism controls production of ATP required for the secretion of critical anti-viral molecules by CTLs.


Subject(s)
Immunity , Mitochondria/immunology , Virus Diseases/immunology , Adenosine Triphosphate/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Humans , Mice , Models, Biological
7.
Science ; 348(6238): 995-1001, 2015 05 29.
Article in English | MEDLINE | ID: mdl-25883318

ABSTRACT

Protective CD8(+) T cell-mediated immunity requires a massive expansion in cell number and the development of long-lived memory cells. Using forward genetics in mice, we identified an orphan protein named lymphocyte expansion molecule (LEM) that promoted antigen-dependent CD8(+) T cell proliferation, effector function, and memory cell generation in response to infection with lymphocytic choriomeningitis virus. Generation of LEM-deficient mice confirmed these results. Through interaction with CR6 interacting factor (CRIF1), LEM controlled the levels of oxidative phosphorylation (OXPHOS) complexes and respiration, resulting in the production of pro-proliferative mitochondrial reactive oxygen species (mROS). LEM provides a link between immune activation and the expansion of protective CD8(+) T cells driven by OXPHOS and represents a pathway for the restoration of long-term protective immunity based on metabolically modified cytotoxic CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Respiration , Immunity, Cellular , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Molecular Sequence Data , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism
8.
Org Biomol Chem ; 12(44): 8952-65, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25277547

ABSTRACT

A structure-based design campaign for non-covalent small molecule inhibitors of human granzyme B was carried out by means of a virtual screening strategy employing three constraints and probe site-mapping with FTMAP to identify ligand "hot spots". In addition, new scaffolds of diverse structures were subsequently explored with ROCS shape-based superposition methods, following by Glide SP docking, induced fit docking and analysis of QikProp molecular properties. Novel classes of moderately active small molecule blockers (≥25 µM IC50 values) from commercially available libraries were identified, and three novel scaffolds have been synthesized by multi-step procedures. Furthermore, we provide an example of a comprehensive structure-based drug discovery approach to non-covalent inhibitors that relies on the X-ray structure of a covalently bound ligand and suggest that the design path may be compromised by alternative and unknown binding poses.


Subject(s)
Drug Design , Granzymes/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Algorithms , Crystallography, X-Ray , Dose-Response Relationship, Drug , Granzymes/metabolism , Humans , Models, Molecular , Molecular Conformation , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
9.
Stem Cells ; 32(9): 2550-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24964278

ABSTRACT

Prime regulation over hematopoietic progenitor cell (HPC) production is exerted by hematopoietins (HPs) and their Janus kinase-coupled receptors (HP-Rs). For HP/HP-R studies, one central challenge in determining specific effects involves the delineation of nonredundant signal transduction factors and their lineage restricted actions. Via loss-of-function studies, we define roles for an HP-regulated Serpina3g/Spi2A intracellular serpin during granulomyelocytic, B-cell, and hematopoietic stem cell (HSC) formation. In granulomyelocytic progenitors, granulocyte macrophage colony stimulating factor (GMCSF) strongly induced Serpina3g expression with Stat5 dependency. Spi2A-knockout (KO) led to 20-fold decreased CFU-GM formation, limited GMCSF-dependent granulocyte formation, and compromised neutrophil survival upon tumor necrosis factor alpha (TNF-α) exposure. In B-cell progenitors, Serpina3g was an interleukin-7 (IL7) target. Spi2A-KO elevated CFU-preB greater than sixfold and altered B-cell formation in competitive bone marrow transplant (BMT), and CpG challenge experiments. In HSCs, Serpina3g/Spi2A expression was also elevated. Spi2A-KO compromised LT-HSC proliferation (as well as lineage(neg) Sca1(pos) Kit(pos) (LSK) cell lysosomal integrity), and skewed LSK recovery post 5-FU. Spi2A therefore functions to modulate HP-regulated immune cell and HSC formation post-5-FU challenge.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Serpins/metabolism , Animals , Mice
10.
Immunology ; 141(3): 446-56, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24383645

ABSTRACT

Natural regulatory T (nTreg) cells generated in the thymus are essential throughout life for the maintenance of T-cell homeostasis and the prevention of autoimmunity. T-cell receptor (TCR)/CD28-mediated activation of nuclear factor-κB and (J)un (N)-terminal kinase pathways is known to play a key role in nTreg cell development but many of the predicted molecular interactions are based on extrapolations from non-Treg cell TCR stimulation with non-physiological ligands. For the first time, we provide strong genetic evidence of a scaffold function for the Caspase Recruitment Domain (CARD) of the TCR signalling protein CARD-MAGUK1 (CARMA1) in nTreg cell development in vivo. We report two, new, N-ethyl-N-nitrosourea-derived mutant mice, Vulpo and Zerda, with a profound block in the development of nTreg cells in the thymus as well as impaired inducible Treg cell differentiation in the periphery. Despite independent heritage, both mutants harbour different point mutations in the CARD of the CARMA1 protein. Mutations in vulpo and zerda do not affect expression levels of CARMA1 but still impair signalling through the TCR due to defective downstream Bcl-10 recruitment by the mutated CARD of CARMA1. Phenotypic differences observed between Vulpo and Zerda mutants suggest a role for the CARD of CARMA1 independent of Bcl-10 activation of downstream pathways. We conclude that our forward genetic approach demonstrates a critical role for the CARD function of CARMA1 in Treg cell development in vivo.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Cell Differentiation , Ethylnitrosourea/pharmacology , Mutagens/pharmacology , Mutation , T-Lymphocytes, Regulatory/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoimmunity/genetics , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/metabolism , Cells, Cultured , Genotype , Heredity , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phenotype , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism
11.
J Immunol ; 191(5): 2319-27, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23913965

ABSTRACT

Regulatory T cells (Tregs) play a pivotal role in the maintenance of immune tolerance and hold great promise as cell therapy for a variety of immune-mediated diseases. However, the cellular mechanisms that regulate Treg maintenance and homeostasis have yet to be fully explored. Although Tregs express granzyme-B (GrB) to suppress effector T cells via direct killing, the mechanisms by which they protect themselves from GrB-mediated self-inflicted damage are unknown. To our knowledge, we show for the first time that both induced Tregs and natural Tregs (nTregs) increase their intracellular expression of GrB and its endogenous inhibitor, serine protease inhibitor 6 (Spi6) upon activation. Subcellular fractionation and measurement of GrB activity in the cytoplasm of Tregs show that activated Spi6(-/-) Tregs had significantly higher cytoplasmic GrB activity. We observed an increase in GrB-mediated apoptosis in Spi6(-/-) nTregs and impaired suppression of alloreactive T cells in vitro. Spi6(-/-) Tregs were rescued from apoptosis by the addition of a GrB inhibitor (Z-AAD-CMK) in vitro. Furthermore, adoptive transfer experiments showed that Spi6(-/-) nTregs were less effective than wild type nTregs in suppressing graft-versus-host disease because of their impaired survival, as shown in our in vivo bioluminescence imaging. Finally, Spi6-deficient recipients rejected MHC class II-mismatch heart allografts at a much faster rate and showed a higher rate of apoptosis among Tregs, as compared with wild type recipients. To our knowledge, our data demonstrate, for the first time, a novel role for Spi6 in Treg homeostasis by protecting activated Tregs from GrB-mediated injury. These data could have significant clinical implications for Treg-based therapy in immune-mediated diseases.


Subject(s)
Granzymes/immunology , Homeostasis/immunology , Membrane Proteins/immunology , Serine Endopeptidases/immunology , Serpins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis/immunology , Flow Cytometry , Graft vs Host Disease/immunology , Granzymes/metabolism , Heart Transplantation/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Serine Endopeptidases/metabolism , Serpins/metabolism , T-Lymphocytes, Regulatory/metabolism
12.
Immunol Lett ; 152(1): 65-76, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23624075

ABSTRACT

Serine proteases control a wide variety of physiological and pathological processes in multi-cellular organisms, including blood clotting, cancer, cell death, osmo-regulation, tissue re-modeling and immunity to infection. T lymphocytes are required for adaptive cell mediated immunity and serine proteases are not only important for effector function but also homeostatic regulation of cell numbers. Serine Protease Inhibitors (Serpins) are the physiological regulators of serine proteases activity. In this review, I will discuss the role of serpins in controlling the recognition of antigen, effector function and homeostatic control of T lymphocytes through the inhibition of physiological serine protease targets. An emerging view of serpins is that they are important promoters of cellular viability through their inhibition of executioner proteases. This will be discussed in the context of the T lymphocyte survival during effector responses and the development and persistence of long-lived memory T cells. The potent anti-apoptotic properties of serpins can also work against adaptive cell immunity by protecting viruses and tumors from eradication by cytotoxic T cells (CTL). Recent insights from knock-out mouse models demonstrate that these serpins also are required for hematological progenitor cells and so are critical for the development of lineages other than T lymphocytes. Given the emerging role of serpins in multiple aspects of lymphocyte immunity and blood development I will review the progress to date in developing new immunotherapeutic approaches based directly on serpins or knowledge gained from identifying their physiologically relevant protease targets.


Subject(s)
Serine Proteinase Inhibitors/physiology , Serpins/physiology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Homeostasis , Humans , Immunologic Memory , Immunologic Surveillance , Immunotherapy , Lymphocyte Activation , Mice , Mice, Knockout
13.
Biomaterials ; 34(14): 3697-705, 2013 May.
Article in English | MEDLINE | ID: mdl-23422590

ABSTRACT

Cytotoxic T lymphocytes (CTL) and natural killer (NK) cells protect vertebrates by killing infected or transformed cells using granzyme B (GrB) to induce apoptosis. However, GrB-induced apoptosis of target cells causes inflammatory disease and chronic transplant rejection and so is an important disease target. The aim of this study was to prevent apoptosis of the target cells by delivering a plasmid encoding GrB inhibitor proteinase inhibitor-9 (PI-9) using cationic polymers as a non-viral vector. Polyethyleneimine (PEI, branched, Mn 10 kDa) gives a high degree of gene transfection efficiency in many types of cell lines, but it is highly cytotoxic. To reduce this cytotoxicity, we modified PEI by blocking primary amine groups through nucleophilic addition between primary amine and a protected mannose-functionalized cyclic carbonate (MTC-ipman), generating a carbamate linkage through the ring-opening of the cyclic carbonate. Deprotection of the mannose yielded a PEI polymer that is decorated with the carbohydrate. PEI with 7 or 20 of 67 primary amine groups substituted by the carbohydrate had similar gene binding ability compared to unmodified PEI, leading to almost 100% transfection efficiency of a GFP-reporter plasmid in HEK293T human embryonic kidney cells. Furthermore, modification of PEI resulted in a decrease in the cytotoxicity of PEI/DNA complexes. However, PEI with all primary amine groups blocked was unable to form a complex with DNA, and so reporter transfection was negligible. The PI-9 encoding plasmid was transfected into HEK293T cells effectively using the modified PEIs with the optimal degree of primary amine substitution, protecting up to 80% HEK293T cells from killing by human natural killer-like leukemic YT cells. Therefore, these carbamate-mannose modified PEI/PI-9 encoding plasmid complexes have potential clinical utility in the prevention of chronic transplant rejection and inflammatory disease caused by GrB.


Subject(s)
Carbamates/chemistry , Granzymes/antagonists & inhibitors , Mannose/chemistry , Polyethyleneimine/chemistry , Polymers/chemistry , T-Lymphocytes, Cytotoxic/drug effects , Cell Line , DNA/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Plasmids/chemistry , Polymers/chemical synthesis , Transfection
14.
J Exp Med ; 210(2): 225-32, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23319700

ABSTRACT

Erythropoietin (EPO) and its cell surface receptor (EPOR) are essential for red blood cell production and exert important cytoprotective effects on select vascular, immune, and cancer cells. To discover novel EPO action modes, we profiled the transcriptome of primary erythroid progenitors. We report Serpina3g/Spi2A as a major new EPO/EPOR target for the survival of erythroid progenitors. In knockout mice, loss of Spi2A worsened anemia caused by hemolysis, radiation, or transplantation. EPO-induced erythropoiesis also was compromised. In particular, maturing erythroblasts required Spi2A for cytoprotection, with iron and reactive oxygen species as cytotoxic agents. Spi2A defects were ameliorated by cathepsin-B/L inhibition, and by genetic co-deletion of lysosomal cathepsin B. Pharmacological inhibition of cathepsin B/L enhanced EPO-induced red cell formation in normal mice. Overall, we define an unexpected EPO action mode via an EPOR-Spi2A serpin-cathepsin axis in maturing erythroblasts, with lysosomal cathepsins as novel therapeutic targets.


Subject(s)
Cathepsins/antagonists & inhibitors , Erythroblasts/cytology , Erythroblasts/metabolism , Erythropoiesis/physiology , Erythropoietin/physiology , Anemia/genetics , Anemia/metabolism , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin B/deficiency , Cathepsin B/genetics , Cathepsin L/antagonists & inhibitors , Erythropoiesis/drug effects , Lysosomes/metabolism , Mice , Mice, Knockout , Receptors, Erythropoietin/physiology , Serpins/deficiency , Serpins/genetics , Serpins/physiology , Signal Transduction , Transcriptome
15.
J Immunol ; 189(3): 1133-43, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22745374

ABSTRACT

The persistence of memory T lymphocytes confers lifelong protection from pathogens. Memory T cells survive and undergo homeostatic proliferation (HSP) in the absence of Ag, although the cell-intrinsic mechanisms by which cytokines drive the HSP of memory T cells are not well understood. In this study we report that lysosome stability limits the long-term maintenance of memory CD8(+) T cell populations. Serine protease inhibitor (Spi) 2A, an anti-apoptotic cytosolic cathepsin inhibitor, is induced by both IL-15 and IL-7. Mice deficient in Spi2A developed fewer memory phenotype CD44(hi)CD8(+) T cells with age, which underwent reduced HSP in the bone marrow. Spi2A was also required for the maintenance of central memory CD8(+) T cell populations after acute infection with lymphocytic choriomeningitis virus. Spi2A-deficient Ag-specific CD8(+) T cell populations declined more than wild-type competitors after viral infection, and they were eroded further after successive infections. Spi2A protected memory cells from lysosomal breakdown by inhibiting cathepsin B. The impaired maintenance of Spi2A-deficient memory CD8(+) T cells was rescued by concomitant cathepsin B deficiency, demonstrating that cathepsin B was a physiological target of Spi2A in memory CD8(+) T cell survival. Our findings support a model in which protection from lysosomal rupture through cytokine-induced expression of Spi2A determines the long-term persistence of memory CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cathepsin B/physiology , Immunologic Memory , Animals , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/metabolism , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Female , Homeostasis/genetics , Homeostasis/immunology , Immunologic Memory/genetics , Lysosomes/enzymology , Lysosomes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Serpins/deficiency , Serpins/physiology
16.
J Immunol ; 188(8): 3920-7, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22407915

ABSTRACT

The homeostasis of the immune system is tightly controlled by both cell-extrinsic and -intrinsic mechanisms. These regulators, not all known to date, drive cells in and out of quiescence when and where required to allow the immune system to function. In this article, we describe a deficiency in deoxycytidine kinase (DCK), one of the major enzymes of the nucleoside salvage pathway, which affects peripheral T cell homeostatic proliferation and survival. As a result of an N-ethyl-N-nitrosourea-induced mutation in the last α helix of DCK, a functionally null protein has been generated in the mouse and affects the composition of the hematopoietic system. Both B and T lymphocyte development is impaired, leading to a state of chronic lymphopenia and to a significant increase in the number of myeloid cells and erythrocytes. In the periphery, we found that mutant lymphocytes adopt a CD44(high)CD62L(low) memory phenotype, with high levels of proliferation and apoptosis. These phenotypes are notably the result of a cell-extrinsic-driven lymphopenia-induced proliferation as wild-type cells transferred into DCK-deficient recipients adopt the same profile. In addition, DCK also regulates lymphocyte quiescence in a cell-intrinsic manner. These data establish dCK as a new regulator of hematopoietic integrity and lymphocyte quiescence and survival.


Subject(s)
Deoxycytidine Kinase/immunology , Nucleosides/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/immunology , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Proliferation , Cell Survival , Deoxycytidine Kinase/genetics , Erythrocytes/immunology , Erythrocytes/metabolism , Ethylnitrosourea/toxicity , Gene Silencing , Immunity, Innate , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myeloid Cells/immunology , Myeloid Cells/metabolism , Nucleosides/genetics , Nucleosides/metabolism , Sequence Analysis, DNA , T-Lymphocytes/metabolism
17.
J Immunol ; 188(3): 1057-63, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22227570

ABSTRACT

How dendritic cells (DC) present Ag to cytotoxic T cells (CTL) without themselves being killed through contact-mediated cytotoxicity (so-called kiss of death) has proved to be controversial. Using mice deficient in serine protease inhibitor 6 (Spi6), we show that Spi6 protects DC from the kiss of death by inhibiting granzyme B (GrB) delivered by CTL. Infection of Spi6 knockout mice with lymphocytic choriomeningitis virus revealed impaired survival of CD8α DC. The impaired survival of Spi6 knockout CD8α DC resulted in impaired priming and expansion of both primary and memory lymphocytic choriomeningitis virus-specific CTL, which could be corrected by GrB deficiency. The rescue in the clonal burst obtained by GrB elimination demonstrated that GrB was the physiological target through which Spi6 protected DC from CTL. We conclude that the negative regulation of DC priming of CD8 T lymphocyte immunity by CTL killing is mitigated by the physiological inhibition of GrB by Spi6.


Subject(s)
Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Membrane Proteins/physiology , Serine Endopeptidases/physiology , Serpins/physiology , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Granzymes/immunology , Mice , Mice, Knockout , Serine Proteinase Inhibitors/physiology , T-Lymphocytes, Cytotoxic/immunology
18.
J Immunol ; 187(5): 2252-60, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21795594

ABSTRACT

Clinical trials using allogeneic mesenchymal stem cells (MSCs) are ongoing for the purpose of providing therapeutic benefit for a variety of human disorders. Pertinent to their clinical use are the accessibility to sufficient quantities of these cells allowing for repetitive administration, as well as a better understanding of the specific mechanisms by which allogeneic MSCs evade host immune responses that in turn influence their life span following administration. In this report, we sought to characterize and compare human peripheral blood MSCs (hPB-MSCs) with bone marrow-derived MSCs. hPB-MSCs met the established criteria to characterize this cellular lineage, including capacity for self-renewal, differentiation into tissues of mesodermal origin, and expression of phenotypic surface markers. In addition, hPB-MSCs suppressed alloreactive proliferation as well as the production of proinflammatory cytokines. Examination of the mechanisms by which allogeneic MSCs evade the host immune response, which is crucial for their therapeutic use, demonstrated that constitutive expression of serine protease inhibitor 9 (PI-9) on hPB-MSCs and bone marrow-derived MSCs is a major defense mechanism against granzyme B-mediated destruction by NK cells. Similarly, MSCs treated with small interfering RNA for PI-9 increased MSC cellular death, whereas expression of transgenic PI-9 following retroviral transduction protected MSCs. These data significantly advance our understanding of the immunomodulatory role for hPB-MSCs as well as the mechanisms by which they evade host immune responses. These findings contribute to the development of MSC-based therapies for diseases.


Subject(s)
Cytotoxicity, Immunologic/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Serpins/immunology , Bone Marrow Cells/cytology , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/methods , Cell Differentiation/immunology , Cell Proliferation , Cell Separation , Flow Cytometry , Humans , Killer Cells, Natural/immunology , Peripheral Blood Stem Cell Transplantation/methods , Serpins/metabolism
19.
Blood ; 117(4): 1176-83, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21076046

ABSTRACT

Clinical trials using mesenchymal stem cells (MSCs) have been initiated worldwide. An improved understanding of the mechanisms by which allogeneic MSCs evade host immune responses is paramount to regulating their survival after administration. This study has focused on the novel role of serine protease inhibitor (SPI) in the escape of MSCs from host immunosurveillance through the inhibition of granzyme B (GrB). Our data indicate bone marrow-derived murine MSCs express SPI6 constitutively. MSCs from mice deficient for SPI6 (SPI6(-/-)) exhibited a 4-fold higher death rate by primed allogeneic cytotoxic T cells than did wild-type MSCs. A GrB inhibitor rescued SPI6(-/-) MSCs from cytotoxic T-cell killing. Transduction of wild-type MSCs with MigR1-SPI6 also protected MSCs from cytotoxic T cell-mediated death in vitro. In addition, SPI6(-/-) MSCs displayed a shorter lifespan than wild-type MSCs when injected into an allogeneic host. We conclude that SPI6 protects MSCs from GrB-mediated killing and plays a pivotal role in their survival in vivo. Our data could serve as a basis for future SPI-based strategies to regulate the survival and function of MSCs after administration and to enhance the efficacy of MSC-based therapy for diseases.


Subject(s)
Immune Evasion/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/metabolism , Serine Endopeptidases/genetics , Serpins/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Granzymes/antagonists & inhibitors , Granzymes/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/metabolism , Serpins/metabolism
20.
Immunol Rev ; 235(1): 147-58, 2010 May.
Article in English | MEDLINE | ID: mdl-20536561

ABSTRACT

Serine proteases control a wide variety of physiological and pathological processes in multi-cellular organisms, including blood clotting, cancer, cell death, osmoregulation, tissue remodeling, and immunity to infection. Cytotoxic T lymphocytes (CTLs) are required for adaptive cell-mediated immunity to intracellular pathogens by killing infected cells and through the development of memory T cells. Serine proteases not only allow a CTL to kill but also impose homeostatic control on CTL number. Serine protease inhibitors (serpins) are the physiological regulators of serine proteases' activity. In this review, I discuss the role of serpins in controlling the recognition of antigen, effector function, and homeostatic control of CTLs through the inhibition of physiological serine protease targets. An emerging view of serpins is that they are important promoters of cellular viability through their inhibition of executioner proteases. This view is discussed in the context of the T-lymphocyte survival during effector responses and the development and persistence of long-lived memory T cells. Given the important role serpins play in CTL immunity, I discuss the potential for developing new immunotherapeutic approaches based directly on serpins or knowledge gained from identifying their physiologically relevant protease targets.


Subject(s)
Cytotoxicity, Immunologic , Serine Proteases/metabolism , Serpins/metabolism , T-Lymphocytes, Cytotoxic/enzymology , Animals , Apoptosis , Humans , Immunologic Memory , Immunotherapy/methods , Serine Proteases/immunology , Serpins/immunology , Serpins/therapeutic use , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...