Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nature ; 621(7980): 813-820, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37587341

ABSTRACT

Disruption of the lung endothelial-epithelial cell barrier following respiratory virus infection causes cell and fluid accumulation in the air spaces and compromises vital gas exchange function1. Endothelial dysfunction can exacerbate tissue damage2,3, yet it is unclear whether the lung endothelium promotes host resistance against viral pathogens. Here we show that the environmental sensor aryl hydrocarbon receptor (AHR) is highly active in lung endothelial cells and protects against influenza-induced lung vascular leakage. Loss of AHR in endothelia exacerbates lung damage and promotes the infiltration of red blood cells and leukocytes into alveolar air spaces. Moreover, barrier protection is compromised and host susceptibility to secondary bacterial infections is increased when endothelial AHR is missing. AHR engages tissue-protective transcriptional networks in endothelia, including the vasoactive apelin-APJ peptide system4, to prevent a dysplastic and apoptotic response in airway epithelial cells. Finally, we show that protective AHR signalling in lung endothelial cells is dampened by the infection itself. Maintenance of protective AHR function requires a diet enriched in naturally occurring AHR ligands, which activate disease tolerance pathways in lung endothelia to prevent tissue damage. Our findings demonstrate the importance of endothelial function in lung barrier immunity. We identify a gut-lung axis that affects lung damage following encounters with viral pathogens, linking dietary composition and intake to host fitness and inter-individual variations in disease outcome.


Subject(s)
Endothelial Cells , Lung , Orthomyxoviridae Infections , Receptors, Aryl Hydrocarbon , Animals , Humans , Mice , Apelin/metabolism , Diet , Endothelial Cells/metabolism , Endothelium/cytology , Endothelium/metabolism , Epithelial Cells/metabolism , Erythrocytes/metabolism , Influenza, Human/immunology , Influenza, Human/metabolism , Intestines/metabolism , Leukocytes/metabolism , Ligands , Lung/immunology , Lung/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Receptors, Aryl Hydrocarbon/metabolism
2.
Circulation ; 137(5): 488-503, 2018 01 30.
Article in English | MEDLINE | ID: mdl-28775077

ABSTRACT

BACKGROUND: Cardiac transplantation is an excellent treatment for end-stage heart disease. However, rejection of the donor graft, in particular, by chronic rejection leading to cardiac allograft vasculopathy, remains a major cause of graft loss. The lymphatic system plays a crucial role in the alloimmune response, facilitating trafficking of antigen-presenting cells to draining lymph nodes. The encounter of antigen-presenting cells with T lymphocytes in secondary lymphoid organs is essential for the initiation of alloimmunity. Donor lymphatic vessels are not anastomosed to that of the recipient during transplantation. The pathophysiology of lymphatic disruption is unknown, and whether this disruption enhances or hinders the alloimmune responses is unclear. Although histological analysis of lymphatic vessels in donor grafts can yield information on the structure of the lymphatics, the function following cardiac transplantation is poorly understood. METHODS: Using single-photon emission computed tomography/computed tomography lymphoscintigraphy, we quantified the lymphatic flow index following heterotrophic cardiac transplantation in a murine model of chronic rejection. RESULTS: Ten weeks following transplantation of a minor antigen (HY) sex-mismatched heart graft, the lymphatic flow index was significantly increased in comparison with sex-matched controls. Furthermore, the enhanced lymphatic flow index correlated with an increase in donor cells in the mediastinal draining lymph nodes; increased lymphatic vessel area; and graft infiltration of CD4+, CD8+ T cells, and CD68+ macrophages. CONCLUSIONS: Chronic rejection results in increased lymphatic flow from the donor graft to draining lymph nodes, which may be a factor in promoting cellular trafficking, alloimmunity, and cardiac allograft vasculopathy.


Subject(s)
Cell Movement , Graft Rejection/immunology , Heart Transplantation , Lymph/immunology , Lymphatic Vessels/immunology , Allografts , Animals , Chronic Disease , Disease Models, Animal , Female , Graft Rejection/diagnostic imaging , Graft Rejection/pathology , Graft Survival , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility , Lymphangiogenesis , Lymphatic Vessels/diagnostic imaging , Lymphatic Vessels/pathology , Lymphoscintigraphy/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Single Photon Emission Computed Tomography Computed Tomography , Time Factors
3.
J Immunol ; 190(9): 4848-60, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23536635

ABSTRACT

Modification of allogeneic dendritic cells (DCs) through drug treatment results in DCs with in vitro hallmarks of tolerogenicity. Despite these observations, using murine MHC-mismatched skin and heart transplant models, donor-derived drug-modified DCs not only failed to induce tolerance but also accelerated graft rejection. The latter was inhibited by injecting the recipient with anti-CD8 Ab, which removed both CD8(+) T cells and CD8(+) DCs. The discrepancy between in vitro and in vivo data could be explained, partly, by the presentation of drug-modified donor DC MHC alloantigens by recipient APCs and activation of recipient T cells with indirect allospecificity, leading to the induction of alloantibodies. Furthermore, allogeneic MHC molecules expressed by drug-treated DCs were rapidly processed and presented in peptide form by recipient APCs in vivo within hours of DC injection. Using TCR-transgenic T cells, Ag presentation of injected OVA-pulsed DCs was detectable for ≤ 3 d, whereas indirect presentation of MHC alloantigen by recipient APCs led to activation of T cells within 14 h and was partially inhibited by reducing the numbers of CD8(+) DCs in vivo. In support of this observation when mice lacking CD8(+) DCs were pretreated with drug-modified DCs prior to transplantation, skin graft rejection kinetics were similar to those in non-DC-treated controls. Of interest, when the same mice were treated with anti-CD40L blockade plus drug-modified DCs, skin graft survival was prolonged, suggesting endogenous DCs were responsible for T cell priming. Altogether, these findings highlight the risks and limitations of negative vaccination using alloantigen-bearing "tolerogenic" DCs.


Subject(s)
Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Isoantigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology , Skin Transplantation/immunology , Tissue Donors , Transplantation
4.
J Am Soc Nephrol ; 23(9): 1474-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22797180

ABSTRACT

Renal ischemia reperfusion injury triggers complement activation, but whether and how the small proinflammatory fragments C3a and C5a contribute to the pathogenesis of this injury remains to be elucidated. Using C3aR-, C5aR-, or C3aR/C5aR-deficient mice and models of renal ischemia-reperfusion injury, we found that deficiency of either or both of these receptors protected mice from injury, but the C3aR/C5aR- and C5aR-deficient mice were most protected. Protection from injury was associated with less cellular infiltration and lower mRNA levels of kidney injury molecule-1, proinflammatory mediators, and adhesion molecules in postischemic kidneys. Furthermore, chimera studies showed that the absence of C3aR and C5aR on renal tubular epithelial cells or circulating leukocytes attenuated renal ischemia-reperfusion injury. In vitro, C3a and C5a stimulation induced inflammatory mediators from both renal tubular epithelial cells and macrophages after hypoxia/reoxygenation. In conclusion, although both C3a and C5a contribute to renal ischemia-reperfusion injury, the pathogenic role of C5a in this injury predominates. These data also suggest that expression of C3aR and C5aR on both renal and circulating leukocytes contributes to the pathogenesis of renal ischemia-reperfusion injury.


Subject(s)
Complement C3a/metabolism , Complement C5a/metabolism , Kidney/blood supply , Kidney/metabolism , Reperfusion Injury/metabolism , Animals , Complement C3a/deficiency , Complement C3a/genetics , Complement C5a/deficiency , Complement C5a/genetics , Cytokines/metabolism , Female , Hepatitis A Virus Cellular Receptor 1 , Kidney/pathology , Leukocytes/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Regional Blood Flow/physiology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction/physiology , Up-Regulation/physiology
5.
J Am Soc Nephrol ; 21(8): 1344-53, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20651167

ABSTRACT

Interaction between C5a, a product of complement activation, and its receptor (C5aR) upregulates antigen-specific T cell responses by modulating the activation of antigen-presenting cells and T cells. Whether this C5a-C5aR interaction contributes to the immune responses that promote renal allograft rejection is unknown. Here, we found that deficiency of C5aR in both graft and recipient reduced allospecific T cell responses and prolonged renal allograft survival. In addition, lack of C5aR impaired the function of donor and recipient antigen-presenting cells and inhibited the response of recipient T cells to allostimulation. Furthermore, deficiency of C5aR in both graft and recipient reduced early inflammation in the grafts, with less cellular infiltration around the vessels and fewer F4/80 positive cells in the peritubular interstitium. These data demonstrate that C5aR is critical for a full adaptive immune response and mediates renal allograft rejection. Engagement of C5aR on dendritic cells and T cells modulates their function, enhancing allospecific T cell responses that lead to allograft rejection. Targeting C5a signaling may have therapeutic potential for T cell-mediated graft rejection.


Subject(s)
Graft Survival , Kidney Transplantation , Receptor, Anaphylatoxin C5a/deficiency , Animals , Graft Survival/immunology , Kidney Transplantation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , Time Factors
6.
J Immunol Methods ; 483: 112794, 2020 08.
Article in English | MEDLINE | ID: mdl-32428450

ABSTRACT

A commonly employed method to determine the function of a particular cell population and to assess its contribution to the overall system in vivo is to selectively deplete that population and observe the effects. Using monoclonal antibodies to deliver toxins to target cells can achieve this with a high degree of efficiency. Here, we describe an in vivo model combining the use of immunotoxins and multidrug resistant (MDR) gene deficient mice so that only MDR deficient cells expressing the target molecule would be depleted while target molecule expressing, but MDR sufficient, cells are spared. This allows targeted depletion at a higher degree of specificity than has been previously achieved. We have applied this technique to study trogocytosis, the intercellular transfer of cell surface molecules, but this principle could also be adapted using technology already available for use in other fields of study.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytotoxicity, Immunologic/drug effects , Genes, MDR/physiology , Immunotoxins/toxicity , Lymphocyte Depletion/methods , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/deficiency , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Female , Graft Survival/drug effects , Heart Transplantation , Histocompatibility Antigens Class II/immunology , Immunoconjugates/toxicity , Immunoglobulin Fab Fragments/toxicity , Kidney Transplantation , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Ribosome Inactivating Proteins, Type 1/toxicity , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Transplantation Tolerance/drug effects
7.
Exp Clin Transplant ; 17(3): 330-338, 2019 06.
Article in English | MEDLINE | ID: mdl-30880652

ABSTRACT

OBJECTIVES: Tertiary lymphoid organs are formed at sites of chronic inflammation and are thought to contribute to the immune response. Here, we aimed to characterize the structure and function of tertiary lymphoid organs in a model of murine kidney allotransplant to understand their role in alloimmunity. MATERIALS AND METHODS: We transplanted 4 C57BL/6 mouse kidneys (isograft group) and 17 DBA/2 mouse kidneys into C57BL/6 mouse recipients. Three DBA/2-to-C57BL/6 transplant mice that rejected their grafts acutely (before 10 days posttransplant) were excluded from the study. The 14 surviving DAB2 grafts were retrieved at day 45 posttransplant and evaluated histologically. The presence of antibody-secreting cells and circulating levels of donor-specific antibodies were also evaluated. RESULTS: We found that tertiary lymphoid organs can be associated with a beneficial response in a kidney allotransplant model. Characterization of B-cell subsets within tertiary lymphoid organs in mouse kidney allografts revealed naive, plasma, and memory B cells, which were mostly grouped within or in close proximity of tertiary lymphoid organs. Staining for intracellular immunoglobulin G showed that many of the B cells within tertiary lymphoid organs were capable of producing antibodies. Although allospecific antibodies were found in the serum of recipient mice and were deposited in the transplanted kidneys, graft function was not affected in this model. CONCLUSIONS: B cells within tertiary lymphoid organs are functional and contribute to the humoral arm of the alloresponse. However, tertiary lymphoid organs are not necessarily associated with graft rejection, suggesting that protective mechanisms are at play.


Subject(s)
Allografts/immunology , B-Lymphocytes/immunology , Immunity, Humoral , Kidney Transplantation , Tertiary Lymphoid Structures/immunology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
8.
EJNMMI Res ; 5(1): 69, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26611870

ABSTRACT

BACKGROUND: Macrophages represent a critical cell type in host defense, development and homeostasis. The ability to image non-invasively pro-inflammatory macrophage infiltrate into a transplanted organ may provide an additional tool for the monitoring of the immune response of the recipient against the donor graft. We therefore decided to image in vivo sialoadhesin (Sn, Siglec 1 or CD169) using anti-Sn mAb (SER-4) directly radiolabelled with (99m)Tc pertechnetate. METHODS: We used a heterotopic heart transplantation model where allogeneic or syngeneic heart grafts were transplanted into the abdomen of recipients. In vivo nanosingle-photon emission computed tomography (SPECT/CT) imaging was performed 7 days post transplantation followed by biodistribution and histology. RESULTS: In wild-type mice, the majority of (99m)Tc-SER-4 monoclonal antibody cleared from the blood with a half-life of 167 min and was located predominantly on Sn(+) tissues in the spleen, liver and bone marrow. The biodistribution in the transplantation experiments confirmed data derived from the non-invasive SPECT/CT images, with significantly higher levels of (99m)Tc-SER-4 observed in allogeneic grafts (9.4 (±2.7) %ID/g) compared to syngeneic grafts (4.3 (±10.3) %ID/g) (p = 0.0022) or in mice which received allogeneic grafts injected with (99m)Tc-IgG isotype control (5.9 (±0.6) %ID/g) (p = 0.0185). The transplanted heart to blood ratio was also significantly higher in recipients with allogeneic grafts receiving (99m)Tc-SER-4 as compared to recipients with syngeneic grafts (p = 0.000004) or recipients with allogeneic grafts receiving (99m)Tc-IgG isotype (p = 0.000002). CONCLUSIONS: Here, we demonstrate that imaging of Sn(+) macrophages in inflammation may provide an important additional and non-invasive tool for the monitoring of the pathophysiology of cellular immunity in a transplant model.

9.
Transplantation ; 92(11): 1194-201, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22089665

ABSTRACT

BACKGROUND: The role of the CD70-specific antibody and the mechanisms by which it extends transplant survival are not known. METHODS: Fully major histocompatibility complex-mismatched heterotopic heart transplantation (BALB/c to C57BL/6) was performed. Treated mice received intraperitoneal injections of wild-type (WT) CD70-specific antibody (FR70) or IgG1 or IgG2a chimeric antibodies on days 0, 2, 4, and 6 posttransplantation. RESULTS: WT FR70 antibody significantly extended heart transplant survival to 19 days compared with untreated mice (median survival time [MST]=10 days). Graft survival using the nondepleting IgG1 antibody was significantly shorter (MST=14 days), whereas the survival using depleting IgG2a antibody (MST=18) was similar to that using WT FR70. The FR70 and IgG2a antibodies demonstrated a greater efficiency of fixing mouse complement over the IgG1 variant in vitro. CD4 and CD8 T-cell graft infiltration was reduced with treatment; however, this was most pronounced with WT FR70 and IgG2a antibody therapy compared with the IgG1 chimeric variant. Circulating donor-specific IgG alloantibodies were initially reduced with WT FR70 treatment (day 8 posttransplantation) but increased at days 15 and 20 posttransplantation to the level detected in untreated controls. CONCLUSION: We conclude that WT (FR70) and the IgG2a depleting variant of CD70-specific antibody reduce graft infiltrating CD4 and CD8 T cells, transiently reduce serum alloantibody levels, and extend graft survival. In contrast, the nondepleting IgG1 variant of this antibody showed lower efficacy. These data suggest that a depleting mechanism of action and not merely costimulation blockade plays a substantial role in the therapeutic effects of CD70-specific antibody.


Subject(s)
Antibodies/pharmacology , Antibody Specificity/immunology , CD27 Ligand/immunology , Graft Survival/drug effects , Heart Transplantation/immunology , Immunoglobulin Fc Fragments/pharmacology , Animals , Antibodies/administration & dosage , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Heart Transplantation/pathology , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Injections, Intraperitoneal , Isoantibodies/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Treatment Outcome
10.
Transplantation ; 90(3): 270-8, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20571468

ABSTRACT

BACKGROUND: It is well established that primed/memory T cells play a critical role in heart transplant rejection. This contributes to the challenges faced in the transplant clinic because current treatments that are efficient in controlling naïve T cell alloresponses have limited efficacy on primed T cell responders. METHODS: Fully MHC-mismatched heart transplantation was performed from BALB/c to C57BL/6 mice presensitized with BALB/c splenocytes 14 days pretransplantation. A combination therapy comprising CD70-, CD154-, and CD8-specific antibodies (Abs) was administered at day 0 and 4 posttransplantation with rapamycin on days 0 to 4. RESULTS: The Ab combination therapy extended heart transplant survival in presensitized recipients from median survival time 8 days (MST) to MST 78 days. A decrease in the number of splenic interferon-gamma-secreting cells measured by ELISpot assay was seen in the treated group compared with the untreated controls. However, graft-infiltrating CD8+ and CD4+ T cells persisted despite treatment and the number of intragraft CD4+ T cells increased at day 30 posttransplantation. When an additional "rescue therapy" comprising the same Abs was readministered at days 30, 60, and 90 posttransplantation, T cell infiltration was reduced and indefinite graft survival was observed. Furthermore, rescue therapy resulted in gradual decrease in titer and, by day 90 posttransplantation, the complete loss of the preexisting, donor-specific Abs. CONCLUSION: We conclude that our Ab combination therapy extends allograft survival in presensitized recipients. When combined with intermittent Ab-mediated rescue therapy, this results in indefinite allograft survival and a loss of the preexisting, donor-specific Abs from the circulation.


Subject(s)
Antibodies/therapeutic use , Graft Rejection/prevention & control , Graft Survival/drug effects , Heart Transplantation/immunology , Immunosuppressive Agents/therapeutic use , Isoantibodies/immunology , T-Lymphocytes/drug effects , Transplantation Tolerance/drug effects , Animals , CD27 Ligand/immunology , CD40 Ligand/immunology , CD8 Antigens/immunology , Drug Therapy, Combination , Female , Forkhead Transcription Factors/metabolism , Graft Occlusion, Vascular/immunology , Graft Rejection/immunology , Histocompatibility , Immunologic Memory/drug effects , Interferon-gamma/blood , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Sirolimus/therapeutic use , Spleen/immunology , Spleen/transplantation , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL