Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Host Microbe ; 22(4): 484-493.e5, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29024642

ABSTRACT

Immunity to intestinal helminth infections has been well studied, but the mechanism of helminth killing prior to expulsion remains unclear. Here we identify epithelial-cell-derived phospholipase A2 group 1B (PLA2g1B) as a host-derived endogenous anthelmintic. PLA2g1B is elevated in resistant mice and is responsible for killing tissue-embedded larvae. Despite comparable activities of other essential type-2-dependent immune mechanisms, Pla2g1b-/- mice failed to expel the intestinal helminths Heligmosomoides polygyrus or Nippostrongylus brasiliensis. Expression of Pla2g1b by epithelial cells was dependent upon intestinal microbiota, adaptive immunity, and common-gamma chain-dependent signaling. Notably, Pla2g1b was downregulated in susceptible mice and inhibited by IL-4R-signaling in vitro, uncoupling parasite killing from expulsion mechanisms. Resistance was restored in Pla2g1b-/- mice by treating infective H. polygyrus L3 larvae with PLA2g1B, which reduced larval phospholipid abundance. These findings uncover epithelial-cell-derived Pla2g1b as an essential mediator of helminth killing, highlighting a previously overlooked mechanism of anti-helminth immunity.


Subject(s)
Group IB Phospholipases A2/immunology , Intestinal Mucosa/immunology , Nematospiroides dubius/immunology , Nippostrongylus/immunology , Phospholipids/metabolism , Strongylida Infections/immunology , Adaptive Immunity , Animals , Gastrointestinal Microbiome/immunology , Group IB Phospholipases A2/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Larva/immunology , Mice , Mice, Knockout , Primary Cell Culture
2.
J Exp Med ; 214(6): 1809-1826, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28507062

ABSTRACT

Immunity to intestinal helminth infections requires the rapid activation of T helper 2 cells (Th2 cells). However, simultaneous expansion of CD4+Foxp3+ regulatory T cells (T reg cells) impedes protective responses, resulting in chronic infections. The ratio between T reg and effector T cells can therefore determine the outcome of infection. The redifferentiation of T reg cells into Th cells has been identified in hyperinflammatory diseases. In this study, we asked whether ex-T reg Th2 cells develop and contribute to type-2 immunity. Using multigene reporter and fate-reporter systems, we demonstrate that a significant proportion of Th2 cells derive from Foxp3+ cells after Heligmosomoides polygyrus infection and airway allergy. Ex-Foxp3 Th2 cells exhibit characteristic Th2 effector functions and provide immunity to H. polygyrus Through selective deletion of Il4ra on Foxp3+ cells, we further demonstrate IL-4 is required for the development of ex-Foxp3 Th2 cells. Collectively, our findings indicate that converting T reg cells into Th2 cells could concomitantly enhance Th2 cells and limit T reg cell-mediated suppression.


Subject(s)
Forkhead Transcription Factors/metabolism , Immunity , Interleukin-4/metabolism , Intestines/immunology , Intestines/parasitology , Nematospiroides dubius/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Cell Polarity , Gene Expression Profiling , Immunity/genetics , Mice, Inbred C57BL , Receptors, Interleukin-4/metabolism , Signal Transduction , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Lymphocytes, Regulatory/immunology
3.
J Allergy Clin Immunol ; 139(2): 655-666.e7, 2017 02.
Article in English | MEDLINE | ID: mdl-27484038

ABSTRACT

BACKGROUND: The molecular and cellular pathways driving the pathogenesis of severe asthma are poorly defined. Tumor progression locus 2 (TPL-2) (COT, MAP3K8) kinase activates the MEK1/2-extracellular-signal regulated kinase 1/2 MAP kinase signaling pathway following Toll-like receptor, TNFR1, and IL-1R stimulation. OBJECTIVE: TPL-2 has been widely described as a critical regulator of inflammation, and we sought to investigate the role of TPL-2 in house dust mite (HDM)-mediated allergic airway inflammation. METHODS: A comparative analysis of wild-type and Map3k8-/- mice was conducted. Mixed bone marrow chimeras, conditional knockout mice, and adoptive transfer models were also used. Differential cell counts were performed on the bronchoalveolar lavage fluid, followed by histological analysis of lung sections. Flow cytometry and quantitative PCR was used to measure type 2 cytokines. ELISA was used to assess the production of IgE, type 2 cytokines, and Ccl24. RNA sequencing was used to characterize dendritic cell (DC) transcripts. RESULTS: TPL-2 deficiency led to exacerbated HDM-induced airway allergy, with increased airway and tissue eosinophilia, lung inflammation, and IL-4, IL-5, IL-13, and IgE production. Increased airway allergic responses in Map3k8-/- mice were not due to a cell-intrinsic role for TPL-2 in T cells, B cells, or LysM+ cells but due to a regulatory role for TPL-2 in DCs. TPL-2 inhibited Ccl24 expression in lung DCs, and blockade of Ccl24 prevented the exaggerated airway eosinophilia and lung inflammation in mice given HDM-pulsed Map3k8-/- DCs. CONCLUSIONS: TPL-2 regulates DC-derived Ccl24 production to prevent severe type 2 airway allergy in mice.


Subject(s)
Asthma/immunology , Chemokine CCL24/metabolism , Dendritic Cells/immunology , Eosinophils/immunology , Lung/immunology , MAP Kinase Kinase Kinases/metabolism , Pneumonia/immunology , Proto-Oncogene Proteins/metabolism , Animals , Antigens, Dermatophagoides/immunology , Cytokines/metabolism , Immunoglobulin E/blood , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Pyroglyphidae/immunology , Signal Transduction , Th2 Cells/immunology
4.
Proc Natl Acad Sci U S A ; 113(5): E568-76, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787865

ABSTRACT

There is a paucity of new therapeutic targets to control allergic reactions and forestall the rising trend of allergic diseases. Although a variety of immune cells contribute to allergy, cytokine-secreting αß(+)CD4(+) T-helper 2 (TH2) cells orchestrate the type-2-driven immune response in a large proportion of atopic asthmatics. To identify previously unidentified putative targets in pathogenic TH2 cells, we performed in silico analyses of recently published transcriptional data from a wide variety of pathogenic TH cells [Okoye IS, et al. (2014) Proc Natl Acad Sci USA 111(30):E3081-E3090] and identified that transcription intermediary factor 1 regulator-alpha (Tif1α)/tripartite motif-containing 24 (Trim24) was predicted to be active in house dust mite (HDM)- and helminth-elicited Il4(gfp+)αß(+)CD4(+) TH2 cells but not in TH1, TH17, or Treg cells. Testing this prediction, we restricted Trim24 deficiency to T cells by using a mixed bone marrow chimera system and found that T-cell-intrinsic Trim24 is essential for HDM-mediated airway allergy and antihelminth immunity. Mechanistically, HDM-elicited Trim24(-/-) T cells have reduced expression of many TH2 cytokines and chemokines and were predicted to have compromised IL-1-regulated signaling. Following this prediction, we found that Trim24(-/-) T cells have reduced IL-1 receptor (IL-1R) expression, are refractory to IL-1ß-mediated activation in vitro and in vivo, and fail to respond to IL-1ß-exacerbated airway allergy. Collectively, these data identify a previously unappreciated Trim24-dependent requirement for IL-1R expression on TH2 cells and an important nonredundant role for T-cell-intrinsic Trim24 in TH2-mediated allergy and antihelminth immunity.


Subject(s)
Hypersensitivity/immunology , Nuclear Proteins/physiology , Receptors, Interleukin-1/metabolism , Th2 Cells/immunology , Transcription Factors/physiology , Animals , Helminths/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/genetics , Th2 Cells/metabolism , Transcription Factors/genetics
5.
PLoS Pathog ; 11(7): e1004994, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26147567

ABSTRACT

Parasitic helminths establish chronic infections in mammalian hosts. Helminth/Plasmodium co-infections occur frequently in endemic areas. However, it is unclear whether Plasmodium infections compromise anti-helminth immunity, contributing to the chronicity of infection. Immunity to Plasmodium or helminths requires divergent CD4+ T cell-driven responses, dominated by IFNγ or IL-4, respectively. Recent literature has indicated that Th cells, including Th2 cells, have phenotypic plasticity with the ability to produce non-lineage associated cytokines. Whether such plasticity occurs during co-infection is unclear. In this study, we observed reduced anti-helminth Th2 cell responses and compromised anti-helminth immunity during Heligmosomoides polygyrus and Plasmodium chabaudi co-infection. Using newly established triple cytokine reporter mice (Il4gfpIfngyfpIl17aFP635), we demonstrated that Il4gfp+ Th2 cells purified from in vitro cultures or isolated ex vivo from helminth-infected mice up-regulated IFNγ following adoptive transfer into Rag1-/- mice infected with P. chabaudi. Functionally, Th2 cells that up-regulated IFNγ were transcriptionally re-wired and protected recipient mice from high parasitemia. Mechanistically, TCR stimulation and responsiveness to IL-12 and IFNγ, but not type I IFN, was required for optimal IFNγ production by Th2 cells. Finally, blockade of IL-12 and IFNγ during co-infection partially preserved anti-helminth Th2 responses. In summary, this study demonstrates that Th2 cells retain substantial plasticity with the ability to produce IFNγ during Plasmodium infection. Consequently, co-infection with Plasmodium spp. may contribute to the chronicity of helminth infection by reducing anti-helminth Th2 cells and converting them into IFNγ-secreting cells.


Subject(s)
Coinfection/immunology , Interferon-gamma/metabolism , Interleukin-12/immunology , Malaria/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Cell Separation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Nematospiroides dubius/immunology , Plasmodium chabaudi/immunology , Polymerase Chain Reaction
6.
Proc Natl Acad Sci U S A ; 111(30): E3081-90, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024218

ABSTRACT

Allergic diseases, orchestrated by hyperactive CD4(+) Th2 cells, are some of the most common global chronic diseases. Therapeutic intervention relies upon broad-scale corticosteroids with indiscriminate impact. To identify targets in pathogenic Th2 cells, we took a comprehensive approach to identify the microRNA (miRNA) and mRNA transcriptome of highly purified cytokine-expressing Th1, Th2, Th9, Th17, and Treg cells both generated in vitro and isolated ex vivo from allergy, infection, and autoimmune disease models. We report here that distinct regulatory miRNA networks operate to regulate Th2 cells in house dust mite-allergic or helminth-infected animals and in vitro Th2 cells, which are distinguishable from other T cells. We validated several miRNA (miR) candidates (miR-15a, miR-20b, miR-146a, miR-155, and miR-200c), which targeted a suite of dynamically regulated genes in Th2 cells. Through in-depth studies using miR-155(-/-) or miR-146a(-/-) T cells, we identified that T-cell-intrinsic miR-155 was required for type-2 immunity, in part through regulation of S1pr1, whereas T-cell-intrinsic miR-146a was required to prevent overt Th1/Th17 skewing. These data identify miR-155, but not miR-146a, as a potential therapeutic target to alleviate Th2-medited inflammation and allergy.


Subject(s)
Helminthiasis, Animal/immunology , Hypersensitivity/immunology , MicroRNAs/immunology , Th2 Cells/immunology , Animals , Gene Expression Profiling , Helminthiasis, Animal/genetics , Helminthiasis, Animal/pathology , Hypersensitivity/genetics , Hypersensitivity/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Pyroglyphidae/immunology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , Sphingosine-1-Phosphate Receptors , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/pathology
7.
Immunity ; 41(1): 89-103, 2014 07 17.
Article in English | MEDLINE | ID: mdl-25035954

ABSTRACT

Foxp3(+) T regulatory (Treg) cells prevent inflammatory disease but the mechanistic basis of suppression is not understood completely. Gene silencing by RNA interference can act in a cell-autonomous and non-cell-autonomous manner, providing mechanisms of intercellular regulation. Here, we demonstrate that non-cell-autonomous gene silencing, mediated by miRNA-containing exosomes, is a mechanism employed by Treg cells to suppress T-cell-mediated disease. Treg cells transferred microRNAs (miRNA) to various immune cells, including T helper 1 (Th1) cells, suppressing Th1 cell proliferation and cytokine secretion. Use of Dicer-deficient or Rab27a and Rab27b double-deficient Treg cells to disrupt miRNA biogenesis or the exosomal pathway, respectively, established a requirement for miRNAs and exosomes for Treg-cell-mediated suppression. Transcriptional analysis and miRNA inhibitor studies showed that exosome-mediated transfer of Let-7d from Treg cell to Th1 cells contributed to suppression and prevention of systemic disease. These studies reveal a mechanism of Treg-cell-mediated suppression mediated by miRNA-containing exosomes.


Subject(s)
Exosomes/genetics , MicroRNAs/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Antigens, CD19/immunology , B-Lymphocytes/immunology , Cell Proliferation , Cytokines/metabolism , DEAD-box RNA Helicases/genetics , Exosomes/immunology , Exosomes/metabolism , Female , Forkhead Transcription Factors/immunology , Gene Transfer, Horizontal/genetics , Inflammation/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA Interference , Ribonuclease III/genetics , Th17 Cells/immunology , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
9.
Open Biol ; 3(1): 120157, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23345540

ABSTRACT

Following thymic output, αß⁺CD4⁺ T cells become activated in the periphery when they encounter peptide-major histocompatibility complex. A combination of cytokine and co-stimulatory signals instructs the differentiation of T cells into various lineages and subsequent expansion and contraction during an appropriate and protective immune response. Our understanding of the events leading to T-cell lineage commitment has been dominated by a single fate model describing the commitment of T cells to one of several helper (T(H)), follicular helper (T(FH)) or regulatory (T(REG)) phenotypes. Although a single lineage-committed and dedicated T cell may best execute a single function, the view of a single fate for T cells has recently been challenged. A relatively new paradigm in αß⁺CD4⁺ T-cell biology indicates that T cells are much more flexible than previously appreciated, with the ability to change between helper phenotypes, between helper and follicular helper, or, most extremely, between helper and regulatory functions. In this review, we comprehensively summarize the recent literature identifying when T(H) or T(REG) cell plasticity occurs, provide potential mechanisms of plasticity and ask if T-cell plasticity is beneficial or detrimental to immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Lineage/immunology , Humans , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Regulatory/cytology
10.
Am J Pathol ; 179(5): 2382-96, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21924228

ABSTRACT

Pulmonary infections and pneumonitis occur frequently after hematopoietic stem cell transplantation. Using a syngeneic mouse model of bone marrow transplantation (BMT), we have previously demonstrated that BMT mice are more susceptible to acute gammaherpesvirus 68 (MHV-68) replication at day 7 after infection. By day 21, the virus is latent in lungs of BMT and control mice, and there is no difference in viral load. Despite similar latent viral load, BMT mice develop severe pneumonitis associated with reduced oxygen saturation, fibrosis, peripheral inflammation, hyaline membranes, and foamy alveolar macrophages, a phenotype that persists for 7 weeks after infection. BMT mice demonstrate increased bronchoalveolar lavage (BAL) cells, and this population is enriched in neutrophils and T cells. Alternatively, activated macrophages appear earlier than do classically activated macrophages. BAL fluid from BMT mice at day 21 after infection contains increased levels of hydrogen peroxide, nitrite, and transforming growth factor-ß (TGF-ß). Mice expressing the dominant-negative transgene dn-TGFßRII in multiple cell types were used as BMT donors. BMT mice with T-cell dnTGFßRII are largely protected from the pneumonitis phenotype, whereas mice with CD11c-dnTGFßRII BMT mice are only modestly protected from pneumonitis. Protection in BMT mice with T-cell dnTGFßRII is associated with decreased TGF-ß derived from parenchymal cells in the BAL fluid, lower nitrite levels, and reduced apoptosis, whereas alternatively activated macrophage markers are unchanged.


Subject(s)
Bone Marrow Transplantation/adverse effects , Gammaherpesvirinae , Herpesviridae Infections , Pneumonia/virology , Pulmonary Fibrosis/virology , Transforming Growth Factor beta/physiology , Animals , Apoptosis/physiology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/virology , Macrophage Activation/physiology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Signal Transduction/physiology , T-Lymphocytes, Regulatory/physiology , Transgenes , Viral Load
11.
Fibrogenesis Tissue Repair ; 4: 18, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21810214

ABSTRACT

BACKGROUND: We have shown previously that murine gammaherpesvirus 68 (γHV68) infection exacerbates established pulmonary fibrosis. Because Toll-like receptor (TLR)-9 may be important in controlling the immune response to γHV68 infection, we examined how TLR-9 signaling effects exacerbation of fibrosis in response to viral infection, using models of bleomycin- and fluorescein isothiocyanate-induced pulmonary fibrosis in wild-type (Balb/c) and TLR-9-/- mice. RESULTS: We found that in the absence of TLR-9 signaling, there was a significant increase in collagen deposition following viral exacerbation of fibrosis. This was not associated with increased viral load in TLR-9-/- mice or with major alterations in T helper (Th)1 and Th2 cytokines. We examined alveolar epithelial-cell apoptosis in both strains, but this could not explain the altered fibrotic outcomes. As expected, TLR-9-/- mice had a defect in the production of interferon (IFN)-ß after viral infection. Balb/c fibroblasts infected with γHV68 in vitro produced more IFN-ß than did infected TLR-9-/- fibroblasts. Accordingly, in vitro infection of Balb/c fibroblasts resulted in reduced proliferation rates whereas infection of TLR-9-/- fibroblasts did not. Finally, therapeutic administration of CpG oligodeoxynucleotides ameliorated bleomycin-induced fibrosis in wild-type mice. CONCLUSIONS: These results show a protective role for TLR-9 signaling in murine models of lung fibrosis, and highlight differences in the biology of TLR-9 between mice and humans.

12.
Immunol Res ; 50(1): 78-86, 2011 May.
Article in English | MEDLINE | ID: mdl-21170739

ABSTRACT

Infectious complications are a serious cause of morbidity and mortality following hematopoietic stem cell transplantation (HSCT), and the lung is a particular target organ post-transplant. Our laboratory has used a murine bone marrow transplant model to study alterations in immunity that occur as a result of transplantation. Our studies focus on immune responses that occur following immune cell reconstitution in the absence of immunosuppressive drug therapy or graft-versus-host disease. We have found that impaired clearance of both bacterial and viral pulmonary infections is related to specific alterations in immune cell function and cytokine production. Our data offer insight into mechanisms that contribute to opportunistic infections in HSCT recipients.


Subject(s)
Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/immunology , Lung Diseases/immunology , Animals , Disease Models, Animal , Hematopoietic Stem Cell Transplantation , Humans , Immunity, Innate/immunology , Mice , Opportunistic Infections/etiology , Opportunistic Infections/immunology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Pseudomonas Infections/etiology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , T-Lymphocytes/immunology , Viruses/immunology
13.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L274-85, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21036917

ABSTRACT

Although γherpesvirus infections are associated with enhanced lung fibrosis in both clinical and animal studies, there is limited understanding about fibrotic effects of γherpesviruses on cell types present in the lung, particularly during latent infection. Wild-type mice were intranasally infected with a murine γherpesvirus (γHV-68) or mock-infected with saline. Twenty-eight days postinfection (dpi), ∼14 days following clearance of the lytic infection, alveolar macrophages (AMs), mesenchymal cells, and CD19-enriched cell populations from the lung and spleen express M(3) and/or glycoprotein B (gB) viral mRNA and harbor viral genome. AMs from infected mice express more transforming growth factor (TGF)-ß(1), CCL2, CCL12, TNF-α, and IFN-γ than AMs from mock-infected mice. Mesenchymal cells express more total TGF-ß(1), CCL12, and TNF-α than mesenchymal cells from mock-infected mice. Lung and spleen CD19-enriched cells express more total TGF-ß(1) 28 dpi compared with controls. The CD19-negative fraction of the spleen overexpresses TGF-ß(1) and harbors viral genome, but this likely represents infection of monocytes. Purified T cells from the lung harbor almost no viral genome. Purified T cells overexpress IL-10 but not TGF-ß(1). Intracellular cytokine staining demonstrated that lung T cells at 28 dpi produce IFN-γ but not IL-4. Thus infection with a murine γherpesvirus is sufficient to upregulate profibrotic and proinflammatory factors in a variety of lung resident and circulating cell types 28 dpi. Our results provide new information about possible contributions of these cells to fibrogenesis in the lungs of individuals harboring a γherpesvirus infection and may help explain why γHV-68 infection can augment or exacerbate fibrotic responses in mice.


Subject(s)
Cytokines/biosynthesis , Herpesviridae Infections/immunology , Pulmonary Fibrosis/etiology , Rhadinovirus/pathogenicity , Tumor Virus Infections/immunology , Animals , Base Sequence , Chemokine CCL2/biosynthesis , DNA Primers/genetics , DNA, Viral/genetics , Disease Models, Animal , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Lung/immunology , Lung/virology , Macrophage Activation , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Male , Mesoderm/immunology , Mesoderm/virology , Mice , Mice, Inbred C57BL , Monocyte Chemoattractant Proteins/biosynthesis , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/virology , Spleen/immunology , Spleen/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Transforming Growth Factor beta1/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Virus Infections/genetics , Tumor Virus Infections/virology , Viral Load
14.
Transplantation ; 90(11): 1139-44, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20671593

ABSTRACT

Transforming growth factor (TGF)-ß is a pleiotropic cytokine with beneficial and detrimental effects posthematopoietic stem-cell transplantation. TGF-ß is increased in specific sites postengraftment and can suppress immune responses and maintain peripheral tolerance. Thus, TGF-ß may promote allograft acceptance. However, TGF-ß is also the central pathogenic cytokine in fibrotic disease and likely promotes pneumonitis. Although TGF-ß can enhance leukocyte recruitment and IgA production, it inhibits both innate and adaptive immune cell function and antiviral host defense posthematopoietic stem-cell transplantation. This review will focus on the current understanding of TGF-ß biology and the numerous ways it can impact outcomes posttransplant.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Transforming Growth Factor beta/immunology , Animals , Cell Survival , Graft Rejection/immunology , Graft Survival , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Pulmonary Fibrosis/immunology , T-Lymphocytes, Regulatory/immunology , Time Factors , Transplantation Tolerance , Transplantation, Homologous , Treatment Outcome
15.
J Immunol ; 184(9): 5130-40, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20348421

ABSTRACT

Patients receiving hematopoietic stem cell transplantation or bone marrow transplantation (BMT) as therapy for various malignancies or autoimmune diseases have an increased risk for infectious complications posttransplant, especially in the lung. We have used BMT in mice and murine gammaherpesvirus, gammaHV-68, to study the efficacy of adaptive immune responses post-BMT. Five weeks posttransplant, mice have fully reconstituted their hematopoietic lineages in both the lung and periphery. When challenged with virus, however, BMT mice have a reduced ability to clear lytic virus from the lung. Defective viral control in BMT mice is not related to impaired leukocyte recruitment or defective APC function. Rather, BMT mice are characterized by defective CD4 cell proliferation, skewing of effector CD4 T cells from a Th1 to a Th17 phenotype, and an immunosuppressive lung environment at the time of infection that includes overexpression of TGF-beta1 and PGE(2) and increased numbers of regulatory T cells. Neither indomethacin treatment to block PG synthesis nor anti-CD25 depletion of regulatory T cells improved antiviral host defense post-BMT. Transplanting mice with transgenic bone marrow expressing a dominant-negative TGF-betaRII under the permissive CD4 promoter created mice in which effector CD4 and CD8 cells were unresponsive to TGF-beta1. Mice with TGF-beta1-nonresponsive effector T cells had restored antiviral immunity and improved Th1 responses post-BMT. Thus, our results indicate that overexpression of TGF-beta1 following myeloablative conditioning post-BMT results in impaired effector T cell responses to viral infection.


Subject(s)
Bone Marrow Transplantation/immunology , Herpesviridae Infections/immunology , Pneumonia, Viral/immunology , Rhadinovirus/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/adverse effects , Transforming Growth Factor beta1/biosynthesis , Tumor Virus Infections/immunology , Animals , Bone Marrow Transplantation/adverse effects , Cell Differentiation/immunology , Cell Proliferation , Growth Inhibitors/adverse effects , Growth Inhibitors/biosynthesis , Growth Inhibitors/genetics , Herpesviridae Infections/therapy , Herpesviridae Infections/virology , Interleukin-17/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia, Viral/therapy , Pneumonia, Viral/virology , Protein Serine-Threonine Kinases/administration & dosage , Protein Serine-Threonine Kinases/therapeutic use , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/administration & dosage , Receptors, Transforming Growth Factor beta/therapeutic use , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/virology , Th1 Cells/immunology , Th1 Cells/pathology , Th1 Cells/virology , Transforming Growth Factor beta1/genetics , Transplantation Conditioning/methods , Tumor Virus Infections/therapy , Tumor Virus Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...