Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Nat Immunol ; 20(7): 902-914, 2019 07.
Article in English | MEDLINE | ID: mdl-31209404

ABSTRACT

Lupus nephritis is a potentially fatal autoimmune disease for which the current treatment is ineffective and often toxic. To develop mechanistic hypotheses of disease, we analyzed kidney samples from patients with lupus nephritis and from healthy control subjects using single-cell RNA sequencing. Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid cells, T cells, natural killer cells and B cells that demonstrated both pro-inflammatory responses and inflammation-resolving responses. We found evidence of local activation of B cells correlated with an age-associated B-cell signature and evidence of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, CXCR4 and CX3CR1, were broadly expressed, implying a potentially central role in cell trafficking. Gene expression of immune cells in urine and kidney was highly correlated, which would suggest that urine might serve as a surrogate for kidney biopsies.


Subject(s)
Kidney/immunology , Lupus Nephritis/immunology , Biomarkers , Biopsy , Cluster Analysis , Computational Biology/methods , Epithelial Cells/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunophenotyping , Interferons/metabolism , Kidney/metabolism , Kidney/pathology , Leukocytes/immunology , Leukocytes/metabolism , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Sequence Annotation , Myeloid Cells/immunology , Myeloid Cells/metabolism , Single-Cell Analysis , Transcriptome
3.
Semin Immunol ; 70: 101836, 2023 11.
Article in English | MEDLINE | ID: mdl-37632992

ABSTRACT

The 'immune risk profile' has been shown to predict mortality in the elderly, highlighting the need to better understand age-related immune dysfunction. While aging leads to many defects affecting all arms of the immune system, this review is focused on the accrual of immuno-suppressive CD4 + T cell populations, including FoxP3 + regulatory T cells, and subsets of IL-10-producing T follicular helper cells. New data suggest that such accumulations constitute feedback mechanisms to temper the ongoing progressive low-grade inflammation that develops with age, the so-called "inflammaging", and by doing so, how they have the potential to promote healthier aging. However, they also impair effector immune responses, notably to infections, or vaccines. These studies also reinforce the idea that the aged immune system should not be considered as a poorly functional version of the young one, but more as a dynamic system in which CD4 + T cells, and other immune/non-immune subsets, differentiate, interact with their milieu and function differently than in young hosts. A better understanding of these unique interactions is thus needed to improve effector immune responses in the elderly, while keeping inflammaging under control.


Subject(s)
Aging , Immune System Diseases , Aged , Humans , CD4-Positive T-Lymphocytes , T-Lymphocytes, Regulatory
4.
J Immunol ; 208(3): 651-659, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34996838

ABSTRACT

The precursors of TCRαß+CD8αα+ intraepithelial lymphocytes (IEL) arise in the thymus through a complex process of agonist selection. We and others have shown that the proapoptotic protein, Bim, is critical to limit the number of thymic IEL precursors (IELp), as loss of Bim at the CD4+CD8+ double-positive stage of development drastically increases IELp. The factors determining this cell death versus survival decision remain largely unknown. In this study, we used CD4CreBcl2f/f mice to define the role of the antiapoptotic protein Bcl-2 and CD4CreBcl2f/fBimf/f mice to determine the role of Bcl-2 in opposing Bim to promote survival of IELp. First, in wild-type mice, we defined distinct subpopulations within PD-1+CD122+ IELp, based on their expression of Runx3 and α4ß7. Coexpression of α4ß7 and Runx3 marked IELp that were most dependent upon Bcl-2 for survival. Importantly, the additional loss of Bim restored Runx3+α4ß7+ IELp, showing that Bcl-2 antagonizes Bim to enable IELp survival. Further, the loss of thymic IELp in CD4CreBcl2f/f mice also led to a dramatic loss of IEL in the gut, and the additional loss of Bim restored gut IEL. The loss of gut IEL was due to both reduced seeding by IELp from the thymus as well as a requirement for Bcl-2 for peripheral IEL survival. Together, these findings highlight subset-specific and temporal roles for Bcl-2 in driving the survival of TCRαß+CD8αα+ IEL and thymic IELp.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Intraepithelial Lymphocytes/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Thymus Gland/cytology , Animals , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Survival/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , Female , Intraepithelial Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Stem Cells/cytology , Thymus Gland/immunology
5.
Am J Transplant ; 23(6): 759-775, 2023 06.
Article in English | MEDLINE | ID: mdl-36871629

ABSTRACT

To date, plasma cell (PC)-targeted therapies have been limited by suboptimal PC depletion and antibody rebound. We hypothesized this is partly because of PC residence in protective bone marrow (BM) microenvironments. The purpose of this proof-of-concept study was to examine the effects of the CXCR4 antagonist, plerixafor, on PC BM residence; its safety profile (alone and in combination with a proteasome inhibitor, bortezomib); and the transcriptional effect on BMPCs in HLA-sensitized kidney transplant candidates. Participants were enrolled into 3 groups: group A (n = 4), plerixafor monotherapy; and groups B (n = 4) and C (n = 4), plerixafor and bortezomib combinations. CD34+ stem cell and PC levels increased in the blood after plerixafor treatment. PC recovery from BM aspirates varied depending on the dose of plerixafor and bortezomib. Single-cell RNA sequencing on BMPCs from 3 group C participants pretreatment and posttreatment revealed multiple populations of PCs, with a posttreatment enrichment of oxidative phosphorylation, proteasome assembly, cytoplasmic translation, and autophagy-related genes. Murine studies demonstrated dually inhibiting the proteasome and autophagy resulted in greater BMPC death than did monotherapies. In conclusion, this pilot study revealed anticipated effects of combined plerixafor and bortezomib on BMPCs, an acceptable safety profile, and suggests the potential for autophagy inhibitors in desensitization regimens.


Subject(s)
Heterocyclic Compounds , Kidney Transplantation , Humans , Animals , Mice , Bortezomib/pharmacology , Bortezomib/therapeutic use , Plasma Cells , Bone Marrow , Proteasome Endopeptidase Complex , Boronic Acids/pharmacology , Boronic Acids/therapeutic use , Pyrazines/pharmacology , Pyrazines/therapeutic use , Hematopoietic Stem Cell Mobilization , Pilot Projects , Heterocyclic Compounds/pharmacology , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Receptors, CXCR4
6.
Immunity ; 38(2): 225-36, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23415911

ABSTRACT

It is widely appreciated that T cells increase glycolytic flux during activation, but the role of mitochondrial flux is unclear. Here, we have shown that mitochondrial metabolism in the absence of glucose metabolism is sufficient to support interleukin-2 (IL-2) induction. Furthermore, we used mice with reduced mitochondrial reactive oxygen species (mROS) production in T cells (T-Uqcrfs(-/-) mice) to show that mitochondria are required for T cell activation to produce mROS for activation of nuclear factor of activated T cells (NFAT) and subsequent IL-2 induction. These mice could not induce antigen-specific expansion of T cells in vivo, but Uqcrfs1(-/-) T cells retained the ability to proliferate in vivo under lymphopenic conditions. This suggests that Uqcrfs1(-/-) T cells were not lacking bioenergetically but rather lacked specific ROS-dependent signaling events needed for antigen-specific expansion. Thus, mitochondrial metabolism is a critical component of T cell activation through the production of complex III ROS.


Subject(s)
Mitochondria/metabolism , NFATC Transcription Factors/genetics , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Animals , Cell Proliferation , Electron Transport Complex III/metabolism , Female , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Iron-Sulfur Proteins/deficiency , Iron-Sulfur Proteins/genetics , Lymphocyte Activation , Lymphopenia/immunology , Lymphopenia/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/immunology , NFATC Transcription Factors/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
7.
Am J Transplant ; 21(5): 1691-1698, 2021 05.
Article in English | MEDLINE | ID: mdl-33128812

ABSTRACT

Kidney transplant recipients administered belatacept-based maintenance immunosuppression present with a more favorable metabolic profile, reduced incidence of de novo donor-specific antibodies (DSAs), and improved renal function and long-term patient/graft survival relative to individuals receiving calcineurin inhibitor (CNI)-based immunosuppression. However, the rates and severity of acute rejection (AR) are greater with the approved belatacept-based regimen than with CNI-based immunosuppression. Although these early co-stimulation blockade-resistant rejections are typically steroid sensitive, the higher rate of cellular AR has led many transplant centers to adopt immunosuppressive regimens that differ from the approved label. This article summarizes the available data on these alternative de novo belatacept-based maintenance regimens. Steroid-sparing, belatacept-based immunosuppression (following T cell-depleting induction therapy) has been shown to yield AR rates comparable to those seen with CNI-based regimens. Concomitant treatment with belatacept plus a mammalian target of rapamycin inhibitor (mTORi; sirolimus or everolimus) has yielded AR rates ranging from 0 to 4%. Because the optimal induction agent and number of induction doses; blood levels of mTORi; and dose, duration, and use of corticosteroids have yet to be determined, larger prospective clinical trials are needed to establish the optimal alternative belatacept-based regimen for minimizing early cellular AR occurrence.


Subject(s)
Kidney Transplantation , Abatacept/therapeutic use , Calcineurin Inhibitors , Graft Rejection/drug therapy , Graft Rejection/etiology , Graft Rejection/prevention & control , Graft Survival , Humans , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Prospective Studies , Transplant Recipients
8.
Immunol Rev ; 277(1): 21-43, 2017 05.
Article in English | MEDLINE | ID: mdl-28462527

ABSTRACT

T cells play a critical role in immune responses as they specifically recognize peptide/MHC complexes with their T-cell receptors and initiate adaptive immune responses. While T cells are critical for performing appropriate effector functions and maintaining immune memory, they also can cause autoimmunity or neoplasia if misdirected or dysregulated. Thus, T cells must be tightly regulated from their development onward. Maintenance of appropriate T-cell homeostasis is essential to promote protective immunity and limit autoimmunity and neoplasia. This review will focus on the role of cell death in maintenance of T-cell homeostasis and outline novel therapeutic strategies tailored to manipulate cell death to limit T-cell survival (eg, autoimmunity and transplantation) or enhance T-cell survival (eg, vaccination and immune deficiency).


Subject(s)
Autoimmune Diseases/immunology , Graft Rejection/immunology , Immunologic Deficiency Syndromes/immunology , Immunotherapy/methods , T-Lymphocytes/physiology , Animals , Autoimmune Diseases/therapy , Cell Death , Cell Survival , Graft Rejection/prevention & control , Homeostasis , Humans , Immunologic Deficiency Syndromes/therapy , Transplantation , Vaccination
9.
Am J Transplant ; 20(2): 411-421, 2020 02.
Article in English | MEDLINE | ID: mdl-31550069

ABSTRACT

Proteasome inhibitor-based strategies hold promise in transplant but have yielded varying results. Carfilzomib, a second-generation proteasome inhibitor, may possess advantages over bortezomib, the first-generation proteasome inhibitors. The purpose of this study was to evaluate the safety, toxicity, and preliminary efficacy of carfilzomib in highly HLA-sensitized kidney transplant candidates. Renal transplant candidates received escalating doses of carfilzomib followed by plasmapheresis (group A) or an identical regimen with additional plasmapheresis once weekly before carfilzomib dosing. Thirteen participants received carfilzomib, which was well tolerated with most adverse events classified as low grade. The safety profile was similar to bortezomib desensitization; however, neurotoxicity was not observed with carfilzomib. Toxicity resulted in permanent dose reduction in 1 participant but caused no withdrawals or deaths. HLA antibodies were substantially reduced with carfilzomib alone, and median maximal immunodominant antibody reduction was 72.8% (69.8% for group A, P = .031, 80.1% for group B, P = .938). After depletion, rebound occurred rapidly and antibody levels returned to baseline between days 81 and 141. Bone marrow studies revealed that approximately 69.2% of plasma cells were depleted after carfilzomib monotherapy. Carfilzomib monotherapy-based desensitization provides an acceptable safety and toxicity profile while leading to significant bone marrow plasma cell depletion and anti-HLA antibody reduction.


Subject(s)
Graft Rejection/prevention & control , Immunosuppression Therapy/methods , Immunosuppressive Agents/administration & dosage , Kidney Transplantation , Oligopeptides/administration & dosage , Proteasome Inhibitors/administration & dosage , Adolescent , Adult , Aged , Biomarkers/blood , Bone Marrow/immunology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Graft Rejection/immunology , HLA Antigens/immunology , Humans , Immunosuppressive Agents/therapeutic use , Isoantibodies/blood , Male , Middle Aged , Oligopeptides/therapeutic use , Plasma Cells/immunology , Prospective Studies , Proteasome Inhibitors/therapeutic use , Treatment Outcome , Young Adult
10.
Proc Natl Acad Sci U S A ; 114(24): E4782-E4791, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28533414

ABSTRACT

Antigen-activated lymphocytes undergo extraordinarily rapid cell division in the course of immune responses. We hypothesized that this unique aspect of lymphocyte biology leads to unusual genomic stress in recently antigen-activated lymphocytes and that targeted manipulation of DNA damage-response (DDR) signaling pathways would allow for selective therapeutic targeting of pathological T cells in disease contexts. Consistent with these hypotheses, we found that activated mouse and human T cells display a pronounced DDR in vitro and in vivo. Upon screening a variety of small-molecule compounds, we found that potentiation of p53 (via inhibition of MDM2) or impairment of cell cycle checkpoints (via inhibition of CHK1/2 or WEE1) led to the selective elimination of activated, pathological T cells in vivo. The combination of these strategies [which we termed "p53 potentiation with checkpoint abrogation" (PPCA)] displayed therapeutic benefits in preclinical disease models of hemophagocytic lymphohistiocytosis and multiple sclerosis, which are driven by foreign antigens or self-antigens, respectively. PPCA therapy targeted pathological T cells but did not compromise naive, regulatory, or quiescent memory T-cell pools, and had a modest nonimmune toxicity profile. Thus, PPCA is a therapeutic modality for selective, antigen-specific immune modulation with significant translational potential for diverse immune-mediated diseases.


Subject(s)
DNA Damage/immunology , Immune System Diseases/therapy , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Etoposide/administration & dosage , Humans , Immune System Diseases/immunology , Lymphocyte Activation , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/therapy , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/immunology , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/immunology
11.
PLoS Pathog ; 13(8): e1006507, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28796839

ABSTRACT

Cytomegalovirus (CMV) causes a persistent, lifelong infection. CMV persists in a latent state and undergoes intermittent subclinical viral reactivation that is quelled by ongoing T cell responses. While T cells are critical to maintain control of infection, the immunological factors that promote CMV persistence remain unclear. Here, we investigated the role of regulatory T cells (Treg) in a mouse model of latent CMV infection using Foxp3-diphtheria toxin receptor (Foxp3-DTR) mice. Eight months after infection, MCMV had established latency in the spleen, salivary gland, lung, and pancreas, which was accompanied by an increased frequency of Treg. Administration of diphtheria toxin (DT) after establishment of latency efficiently depleted Treg and drove a significant increase in the numbers of functional MCMV-specific CD4+ and CD8+ T cells. Strikingly, Treg depletion decreased the number of animals with reactivatable latent MCMV in the spleen. Unexpectedly, in the same animals, ablation of Treg drove a significant increase in viral reactivation in the salivary gland that was accompanied with augmented local IL-10 production by Foxp3-CD4+T cells. Further, neutralization of IL-10 after Treg depletion significantly decreased viral load in the salivary gland. Combined, these data show that Treg have divergent control of MCMV infection depending upon the tissue. In the spleen, Treg antagonize CD8+ effector function and promote viral persistence while in the salivary gland Treg prevent IL-10 production and limit viral reactivation and replication. These data provide new insights into the organ-specific roles of Treg in controlling the reactivation of latent MCMV infection.


Subject(s)
Cytomegalovirus Infections/immunology , T-Lymphocytes, Regulatory/immunology , Virus Activation/immunology , Virus Latency/immunology , Animals , Cytomegalovirus/immunology , Disease Models, Animal , Flow Cytometry , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction
12.
J Immunol ; 198(1): 257-269, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27852740

ABSTRACT

CD8αα TCRαß+ intestinal intraepithelial lymphocytes play a critical role in promoting intestinal homeostasis, although mechanisms controlling their development and peripheral homeostasis remain unclear. In this study, we examined the spatiotemporal role of Bim in the thymic selection of CD8αα precursors and the fate of these cells in the periphery. We found that T cell-specific expression of Bim during early/cortical, but not late/medullary, thymic development controls the agonist selection of CD8αα precursors and limits their private TCRß repertoire. During this process, agonist-selected double-positive cells lose CD4/8 coreceptor expression and masquerade as double-negative (DN) TCRαßhi thymocytes. Although these DN thymocytes fail to re-express coreceptors after OP9-DL1 culture, they eventually mature and accumulate in the spleen where TCR and IL-15/STAT5 signaling promotes their conversion to CD8αα cells and their expression of gut-homing receptors. Adoptive transfer of splenic DN cells gives rise to CD8αα cells in the gut, establishing their precursor relationship in vivo. Interestingly, Bim does not restrict the IL-15-driven maturation of CD8αα cells that is critical for intestinal homeostasis. Thus, we found a temporal and tissue-specific role for Bim in limiting thymic agonist selection of CD8αα precursors and their TCRß repertoire, but not in the maintenance of CD8αα intraepithelial lymphocytes in the intestine.


Subject(s)
Bcl-2-Like Protein 11/biosynthesis , Cell Differentiation/immunology , T-Lymphocyte Subsets/cytology , Thymocytes/cytology , Adoptive Transfer , Animals , Bcl-2-Like Protein 11/immunology , Flow Cytometry , Intestines/cytology , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , Thymocytes/immunology
13.
Proc Natl Acad Sci U S A ; 113(38): 10631-6, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27582468

ABSTRACT

DNMT3a is a de novo DNA methyltransferase expressed robustly after T-cell activation that regulates plasticity of CD4(+) T-cell cytokine expression. Here we show that DNMT3a is critical for directing early CD8(+) T-cell effector and memory fate decisions. Whereas effector function of DNMT3a knockout T cells is normal, they develop more memory precursor and fewer terminal effector cells in a T-cell intrinsic manner compared with wild-type animals. Rather than increasing plasticity of differentiated effector CD8(+) T cells, loss of DNMT3a biases differentiation of early effector cells into memory precursor cells. This is attributed in part to ineffective repression of Tcf1 expression in knockout T cells, as DNMT3a localizes to the Tcf7 promoter and catalyzes its de novo methylation in early effector WT CD8(+) T cells. These data identify DNMT3a as a crucial regulator of CD8(+) early effector cell differentiation and effector versus memory fate decisions.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Immunologic Memory/genetics , T Cell Transcription Factor 1/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , DNA Methylation/genetics , DNA Methylation/immunology , DNA Methyltransferase 3A , Lymphocyte Activation/immunology , Mice , Promoter Regions, Genetic
14.
Eur J Immunol ; 46(10): 2333-2339, 2016 10.
Article in English | MEDLINE | ID: mdl-27457412

ABSTRACT

Virtual memory (VM) CD8+ T cells are present in unimmunized mice, yet possess T-cell receptors specific for foreign antigens. To date, VM cells have only been characterized in C57BL/6 mice. Here, we assessed the cytokine requirements for VM cells in C57BL/6 and BALB/c mice. As reported previously, VM cells in C57BL/6 mice rely mostly on IL-15 and marginally on IL-4. In stark contrast, VM cells in BALB/c mice rely substantially on IL-4 and marginally on IL-15. Further, NKT cells are the likely source of IL-4, because CD1d-deficient mice on a BALB/c background have significantly fewer VM cells. Notably, this NKT/IL-4 axis contributes to appropriate effector and memory T-cell responses to infection in BALB/c mice, but not in C57BL/6 mice. However, the effects of IL-4 are manifest prior to, rather than during, infection. Thus, cytokine-mediated control of the precursor population affects the development of virus-specific CD8+ T-cell memory. Depending upon the genetic background, different cytokines encountered before infection may influence the subsequent ability to mount primary and memory anti-viral CD8+ T-cell responses.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interleukin-15/metabolism , Interleukin-4/metabolism , Lymphocytic choriomeningitis virus/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD1d/genetics , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Immunity, Cellular/genetics , Immunologic Memory/genetics , Interleukin-15/genetics , Interleukin-4/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , T-Lymphocyte Subsets/virology
15.
Crit Care Med ; 45(4): e426-e432, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27635771

ABSTRACT

OBJECTIVES: Heterogeneity in sepsis-related pathobiology presents a significant challenge. Resolving this heterogeneity presents an opportunity to understand pathobiology and improve patient care. Olfactomedin-4 is a neutrophil subset marker and may contribute to sepsis heterogeneity. Our objective was to evaluate the expression of olfactomedin-4 and characterize neutrophil heterogeneity in children with septic shock. DESIGN: Single-center, prospective cohort, as well as secondary analysis of existing transcriptomic and proteomic databases. SETTING: Tertiary care PICU. PATIENTS: Patients from 5 days to 18 years old with septic shock were enrolled. Data collected included the expression of olfactomedin-4 messenger RNA, serum protein concentrations, and percentage of neutrophils that express olfactomedin-4. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Secondary analysis of existing transcriptomic data demonstrated that olfactomedin-4 is the most highly expressed gene in nonsurvivors of pediatric septic shock, compared with survivors. Secondary analysis of an existing proteomic database corroborated these observations. In a prospectively enrolled cohort, we quantified the percentage of olfactomedin-4+ neutrophils in patients with septic shock. Patients with a complicated course, defined as greater than or equal to two organ failures at day 7 of septic shock or 28-day mortality, had a higher percentage of olfactomedin-4+ neutrophils, compared with those without a complicated course. By logistic regression, the percentage of olfactomedin-4+ neutrophils was independently associated with increased risk of a complicated course (odds ratio, 1.09; 95% CI, 1.01-1.17; p = 0.024). CONCLUSIONS: Olfactomedin-4 identifies a subpopulation of neutrophils in patients with septic shock, and those with a high percentage of olfactomedin-4+ neutrophils are at higher risk for greater organ failure burden and death. Olfactomedin-4 might serve as a marker of a pathogenic neutrophil subset in patients with septic shock.


Subject(s)
Granulocyte Colony-Stimulating Factor/analysis , Multiple Organ Failure/blood , Neutrophils/chemistry , RNA, Messenger/blood , Shock, Septic/blood , Adolescent , Biomarkers/blood , Child , Child, Preschool , Databases, Genetic , Female , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/genetics , Humans , Infant , Infant, Newborn , Leukocyte Count , Male , Multiple Organ Failure/etiology , Prospective Studies , Proteome , Shock, Septic/complications , Shock, Septic/genetics , Transcriptome
16.
Cell Immunol ; 313: 25-31, 2017 03.
Article in English | MEDLINE | ID: mdl-28063598

ABSTRACT

Following burn injury, a key factor for patients susceptible to opportunistic infections is immune suppression. Butyrate levels are important in maintaining a functional immune system and these levels can be altered after injury. The acid sphingomyelinase (Asm) lipid signaling system has been implicated in a T cell actions with some evidence of being influenced by butyrate. Here, we hypothesized that burn-injury changes in butyrate levels would mediate Asm activity and, consequently, T cell homeostasis. We demonstrate that burn injury temporally decreases butyrate levels. We further determined that T cell Asm activity is increased by butyrate and decreased after burn injury. We additionally observed decreased T cell numbers in Asm-deficient, burn-injured, and microbiota-depleted mice. Finally, we demonstrate that butyrate reduced T cell death in an Asm-dependent manner. These data suggest that restoration of butyrate after burn injury may ameliorate the T cell lost observed in burn-injured patients by Asm regulation.


Subject(s)
Burns/immunology , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes/immunology , Animals , Apoptosis , Butyrates/metabolism , Cells, Cultured , Disease Models, Animal , Gene Knockout Techniques , Homeostasis , Humans , Male , Mice , Mice, Inbred Strains , Signal Transduction , Sphingomyelin Phosphodiesterase/genetics
17.
Immunity ; 28(3): 370-80, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328744

ABSTRACT

Severe congenital neutropenia (SCN) is characterized by a deficiency of mature neutrophils, leading to recurrent bacterial and fungal infections. Although mutations in Elastase-2, neutrophil (ELA2) predominate in human SCN, mutation of Ela2 in mice does not recapitulate SCN. The growth factor independent-1 (GFI1) transcription factor regulates ELA2. Mutations in GFI1 are associated with human SCN, and genetic deletion of Gfi1 results in murine neutropenia. We examined whether human SCN-associated GFI1N382S mutant proteins are causal in SCN and found that GFI1 functions as a rate-limiting granulopoietic molecular switch. The N382S mutation inhibited GFI1 DNA binding and resulted in a dominant-negative block to murine granulopoiesis. Moreover, Gfi1N382S selectively derepressed the monopoietic cytokine CSF1 and its receptor. Gfi1N382S-expressing Csf1-/- cells formed neutrophils. These results reveal a common transcriptional program that underlies both human and murine myelopoiesis, and that is central to the pathogenesis of SCN associated with mutations in GFI1. This shared transcriptional pathway may provide new avenues for understanding SCN caused by mutations in other genes and for clinical intervention into human neutropenias.


Subject(s)
DNA-Binding Proteins/genetics , Granulocytes/cytology , Hematopoiesis/genetics , Macrophage Colony-Stimulating Factor/metabolism , Neutropenia/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Cell Lineage , Electrophoretic Mobility Shift Assay , Flow Cytometry , Hematopoietic Stem Cells/cytology , Humans , Immunoblotting , Immunoprecipitation , Mice , Mutation , Neutropenia/congenital , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
18.
J Immunol ; 195(3): 944-52, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26109645

ABSTRACT

Regulatory T cells (Tregs), a subset of CD4(+) T cells, dramatically accumulate with age in humans and mice and contribute to age-related immune suppression. Recently, we showed that a majority of accumulating Tregs in aged mice expressed low levels of CD25, and their accrual is associated with declining levels of IL-2 in aged mice. In this study, we further investigated the origin of CD25(lo) Tregs in aged mice. First, aged Tregs had high expression of neuropilin-1 and Helios, and had a broad Vß repertoire. Next, we analyzed the gene expression profile of Tregs, naive T cells, and memory T cells in aged mice. We found that the gene expression profile of aged CD25(lo) Tregs were more related to young CD25(lo) Tregs than to either naive or memory T cells. Further, the gene expression profile of aged Tregs was consistent with recently described "effector" Tregs (eTregs). Additional analysis revealed that nearly all Tregs in aged mice were of an effector phenotype (CD44(hi)CD62L(lo)) and could be further characterized by high levels of ICOS and CD69. ICOS contributed to Treg maintenance in aged mice, because in vivo Ab blockade of ICOSL led to a loss of eTregs, and this loss was rescued in Bim-deficient mice. Further, serum levels of IL-6 increased with age and contributed to elevated expression of ICOS on aged Tregs. Finally, Treg accrual was significantly blunted in aged IL-6-deficient mice. Together, our data show a role for IL-6 in promoting eTreg accrual with age likely through maintenance of ICOS expression.


Subject(s)
Aging/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-6/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Apoptosis Regulatory Proteins/genetics , Base Sequence , Bcl-2-Like Protein 11 , Cell Death , Cell Survival , DNA-Binding Proteins/biosynthesis , Gene Expression Profiling , Hyaluronan Receptors/biosynthesis , Immunologic Memory/genetics , Immunologic Memory/immunology , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-6/blood , Interleukin-6/genetics , L-Selectin/biosynthesis , Lectins, C-Type/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropilin-1/biosynthesis , Proto-Oncogene Proteins/genetics , Sequence Analysis, DNA , Transcription Factors/biosynthesis
19.
J Immunol ; 192(1): 84-91, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24259502

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is an inborn disorder of immune regulation caused by mutations affecting perforin-dependent cytotoxicity. Defects in this pathway impair negative feedback between cytotoxic lymphocytes and APCs, leading to prolonged and pathologic activation of T cells. Etoposide, a widely used chemotherapeutic drug that inhibits topoisomerase II, is the mainstay of treatment for HLH, although its therapeutic mechanism remains unknown. We used a murine model of HLH, involving lymphocytic choriomeningitis virus infection of perforin-deficient mice, to study the activity and mechanism of etoposide for treating HLH and found that it substantially alleviated all symptoms of murine HLH and allowed prolonged survival. This therapeutic effect was relatively unique among chemotherapeutic agents tested, suggesting distinctive effects on the immune response. We found that the therapeutic mechanism of etoposide in this model system involved potent deletion of activated T cells and efficient suppression of inflammatory cytokine production. This effect was remarkably selective; etoposide did not exert a direct anti-inflammatory effect on macrophages or dendritic cells, and it did not cause deletion of quiescent naive or memory T cells. Finally, etoposide's immunomodulatory effects were similar in wild-type and perforin-deficient animals. Thus, etoposide treats HLH by selectively eliminating pathologic, activated T cells and may have usefulness as a novel immune modulator in a broad array of immunopathologic disorders.


Subject(s)
Etoposide/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Apoptosis/drug effects , Apoptosis/immunology , DNA-Binding Proteins/genetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Etoposide/administration & dosage , Interferon-gamma/biosynthesis , Lymphocytic choriomeningitis virus , Lymphohistiocytosis, Hemophagocytic/genetics , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Knockout , Topoisomerase II Inhibitors/administration & dosage , Topoisomerase II Inhibitors/pharmacology
20.
J Immunol ; 192(1): 73-83, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24277699

ABSTRACT

The current clinical approach for treating autoimmune diseases is to broadly blunt immune responses as a means of preventing autoimmune pathology. Among the major side effects of this strategy are depressed beneficial immunity and increased rates of infections and tumors. Using the experimental autoimmune encephalomyelitis model for human multiple sclerosis, we report a novel alternative approach for purging autoreactive T cells that spares beneficial immunity. The moderate and temporally limited use of etoposide, a topoisomerase inhibitor, to eliminate encephalitogenic T cells significantly reduces the onset and severity of experimental autoimmune encephalomyelitis, dampens cytokine production and overall pathology, while dramatically limiting the off-target effects on naive and memory adaptive immunity. Etoposide-treated mice show no or significantly ameliorated pathology with reduced antigenic spread, yet have normal T cell and T-dependent B cell responses to de novo antigenic challenges as well as unimpaired memory T cell responses to viral rechallenge. Thus, etoposide therapy can selectively ablate effector T cells and limit pathology in an animal model of autoimmunity while sparing protective immune responses. This strategy could lead to novel approaches for the treatment of autoimmune diseases with both enhanced efficacy and decreased treatment-associated morbidities.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphocyte Depletion , T-Lymphocytes/immunology , Animals , Apoptosis/drug effects , Apoptosis/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Etoposide/administration & dosage , Etoposide/pharmacology , Female , Immunologic Memory , Lymphocyte Activation/drug effects , Lymphocytic choriomeningitis virus/immunology , Mice , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Recurrence , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL