Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
JCI Insight ; 5(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32369450

ABSTRACT

Tumor-associated macrophages (TAMs) contribute to the maintenance of a strong immunosuppressive environment, supporting tumor progression and resistance to treatment. To date, the mechanisms that drive acquisition of these immunosuppressive features are still poorly defined. Heme oxygenase-1 (HO-1) is the rate-limiting enzyme that catabolizes free heme. It displays important cytoprotective, antiinflammatory, and antioxidant properties. A growing body of evidence suggests that HO-1 may also promote tumor development. Herein, we show that HO-1 is highly expressed in monocytic cells in the tumor microenvironment (TME) once they differentiate into TAMs. Deletion of HO-1 in the myeloid compartment enhances the beneficial effects of a therapeutic antitumor vaccine by restoring CD8+ T cell proliferation and cytotoxicity. We further show that induction of HO-1 plays a major role in monocyte education by tumor cells by modulating their transcriptional and epigenetic programs. These results identify HO-1 as a valuable therapeutic target to reprogram the TME and synergize with current cancer therapies to facilitate antitumor response.


Subject(s)
Heme Oxygenase-1/immunology , Immune Tolerance , Membrane Proteins/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Heme Oxygenase-1/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Neoplasms/genetics , Tumor Microenvironment/genetics , Tumor-Associated Macrophages/pathology
3.
Front Immunol ; 11: 579151, 2020.
Article in English | MEDLINE | ID: mdl-33537027

ABSTRACT

Graft-versus-host disease (GVHD) remains a major clinical drawback of allogeneic hematopoietic stem cell transplantation (HSCT). Here, we investigated how the stress responsive heme catabolizing enzyme heme oxygenase-1 (HO-1, encoded by HMOX1) regulates GVHD in response to allogeneic hematopoietic stem cell transplantation in mice and humans. We found that deletion of the Hmox1 allele, specifically in the myeloid compartment of mouse donor bone marrow, promotes the development of aggressive GVHD after allogeneic transplantation. The mechanism driving GVHD in mice transplanted with allogeneic bone marrow lacking HO-1 expression in the myeloid compartment involves enhanced T cell alloreactivity. The clinical relevance of these observations was validated in two independent cohorts of HSCT patients. Individuals transplanted with hematopoietic stem cells from donors carrying a long homozygous (GT)n repeat polymorphism (L/L) in the HMOX1 promoter, which is associated with lower HO-1 expression, were at higher risk of developing severe acute GVHD as compared to donors carrying a short (GT)n repeat (S/L or S/S) polymorphism associated with higher HO-1 expression. In this study, we showed the unique importance of donor-derived myeloid HO-1 in the prevention of lethal experimental GVHD and we corroborated this observation by demonstrating the association between human HMOX1 (GT)n microsatellite polymorphisms and the incidence of severe acute GVHD in two independent HSCT patient cohorts. Donor-derived myeloid HO-1 constitutes a potential therapeutic target for HSCT patients and large-scale prospective studies in HSCT patients are necessary to validate the HO-1 L/L genotype as an independent risk factor for developing severe acute GVHD.


Subject(s)
Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Myeloid-Derived Suppressor Cells/transplantation , Adult , Animals , Disease Models, Animal , Female , Genetic Predisposition to Disease , Graft vs Host Disease/enzymology , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Heme Oxygenase-1/genetics , Homozygote , Humans , Male , Membrane Proteins/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microsatellite Repeats , Middle Aged , Myeloid-Derived Suppressor Cells/enzymology , Phenotype , Polymorphism, Genetic , Retrospective Studies , Risk Assessment , Risk Factors , Severity of Illness Index
4.
J Clin Lipidol ; 11(4): 1101-1105, 2017.
Article in English | MEDLINE | ID: mdl-28711549

ABSTRACT

We report a new case of hypobetalipoproteinemia in a 44-year-old man of Peruvian origin exhibiting a heterozygous PCSK9 missense mutation (c.946 G>T, p. Gly316Cys). In vitro functional studies demonstrated that this mutation leads to a loss of function of PCSK9 on low-density lipoprotein receptor degradation. This patient exhibited liver steatosis; he was neither diabetic, nor obese or alcoholic, but is a carrier of 2 polymorphisms, p.Ile148Met (rs738409) and p.Glu167Lys (rs58542926) on PNPLA3 and TM6SF2 gene, respectively, previously shown to be associated with nonalcoholic steatosis and fibrosis evolution. These data suggested that if a resistance to hepatic steatosis mediated by the PCSK9 deficiency exists, as demonstrated in mice, it is not sufficient to prevent hepatic fatty accumulation in the case of genetic factors predisposing to nonalcoholic fatty liver disease.


Subject(s)
Cholesterol/metabolism , Genetic Predisposition to Disease/genetics , Loss of Function Mutation , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Polymorphism, Genetic , Proprotein Convertase 9/genetics , Adult , Humans , Male , Non-alcoholic Fatty Liver Disease/metabolism
5.
PLoS One ; 8(11): e79984, 2013.
Article in English | MEDLINE | ID: mdl-24278228

ABSTRACT

Activation of innate immunity through Toll-like receptors (TLR) can abrogate transplantation tolerance by revealing hidden T cell alloreactivity. Separately, the cholinergic anti-inflammatory pathway has the capacity to dampen macrophage activation and cytokine release during endotoxemia and ischemia reperfusion injury. However, the relevance of the α7 nicotinic acetylcholine receptor (α7nAChR)-dependent anti-inflammatory pathway in the process of allograft rejection or maintenance of tolerance remains unknown. The aim of our study is to investigate whether the cholinergic pathway could impact T cell alloreactivity and transplant outcome in mice. For this purpose, we performed minor-mismatched skin allografts using donor/recipient combinations genetically deficient for the α7nAChR. Minor-mismatched skin grafts were not rejected unless the mice were housed in an environment with endogenous pathogen exposure or the graft was treated with direct application of imiquimod (a TLR7 ligand). The α7nAChR-deficient recipient mice showed accelerated rejection compared to wild type recipient mice under these conditions of TLR activation. The accelerated rejection was associated with enhanced IL-17 and IFN-γ production by alloreactive T cells. An α7nAChR-deficiency in the donor tissue facilitated allograft rejection but not in recipient mice. In addition, adoptive T cell transfer experiments in skin-grafted lymphopenic animals revealed a direct regulatory role for the α7nAChR on T cells. Taken together, our data demonstrate that the cholinergic pathway regulates alloreactivity and transplantation tolerance at multiple levels. One implication suggested by our work is that, in an organ transplant setting, deliberate α7nAChR stimulation of brain dead donors might be a valuable approach for preventing donor tissue inflammation prior to transplant.


Subject(s)
Allografts , Graft Rejection , Inflammation/prevention & control , Skin Transplantation , Toll-Like Receptors/physiology , alpha7 Nicotinic Acetylcholine Receptor/physiology , Adoptive Transfer , Animals , Base Sequence , Cells, Cultured , DNA Primers , Female , Flow Cytometry , Inflammation/metabolism , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/transplantation
6.
PLoS One ; 8(10): e76040, 2013.
Article in English | MEDLINE | ID: mdl-24146810

ABSTRACT

Several evidences suggest that regulatory T cells (Treg) promote Th17 differentiation. Based on this hypothesis, we tested the effect of IL-17A neutralization in a model of skin transplantation in which long-term graft survival depends on a strong in vivo Treg expansion induced by transient exogenous IL-2 administration. As expected, IL-2 supplementation prevented rejection of MHC class II disparate skin allografts but, surprisingly, not in IL-17A-deficient recipients. We attested that IL-17A was not required for IL-2-mediated Treg expansion, intragraft recruitment or suppressive capacities. Instead, IL-17A prevented allograft rejection by inhibiting Th1 alloreactivity independently of Tregs. Indeed, T-bet expression of naive alloreactive CD4+ T cells and the subsequent Th1 immune response was significantly enhanced in IL-17A deficient mice. Our results illustrate for the first time a protective role of IL-17A in CD4+-mediated allograft rejection process.


Subject(s)
Graft Rejection/prevention & control , Graft Survival/immunology , Interleukin-17/immunology , Interleukin-2/pharmacology , Skin Transplantation , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Cell Communication , Cell Proliferation , Gene Expression , Graft Rejection/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Interleukin-17/deficiency , Mice , Mice, Inbred C57BL , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
7.
PLoS One ; 8(7): e70236, 2013.
Article in English | MEDLINE | ID: mdl-23936171

ABSTRACT

Primary graft dysfunction (PGD) and bronchiolitis obliterans (BO) are the leading causes of morbidity and mortality after lung transplantation. Reports from clinical and rodent models suggest the implication of IL-17A in either PGD or BO. We took advantage of the heterotopic trachea transplantation model in mice to study the direct role of IL-17A in post-transplant airway lesions. Across full MHC barrier, early lesions were controlled in IL-17A(-/-) or anti-IL17 treated recipients. In contrast, IL-17A deficiency did not prevent subsequent obliterative airway disease (OAD). Interestingly, this early protection occurred also in syngeneic grafts and was accompanied by a decrease in cellular stress, as attested by lower HSP70 mRNA levels, suggesting the involvement of IL-17A in ischemia-reperfusion injury (IRI). Furthermore, persistence of multipotent CK14(+) epithelial stem cells underlined allograft protection afforded by IL-17A deficiency or neutralisation. Recipient-derived γδ(+) and CD4(+) T cells were the major source of IL-17A. However, lesions still occurred in the absence of each subset, suggesting a high redundancy between the innate and adaptive IL-17A producing cells. Notably, a double depletion significantly diminished lesions. In conclusion, this work implicated IL-17A as mediator of early post-transplant airway lesions and could be considered as a potential therapeutic target in clinical transplantation.


Subject(s)
Interleukin-17/metabolism , Trachea/metabolism , Trachea/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Interleukin-17/deficiency , Interleukin-17/genetics , Lung Transplantation/adverse effects , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Reperfusion Injury/complications , Trachea/transplantation , Transplantation, Heterotopic , Transplantation, Homologous
8.
Proc Natl Acad Sci U S A ; 108(18): 7431-6, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502497

ABSTRACT

Squamous cell carcinoma (SCC) is the second most frequent skin cancer. The cellular origin of SCC remains controversial. Here, we used mouse genetics to determine the epidermal cell lineages at the origin of SCC. Using mice conditionally expressing a constitutively active KRas mutant (G12D) and an inducible CRE recombinase in different epidermal lineages, we activated Ras signaling in different cellular compartments of the skin epidermis and determined from which epidermal compartments Ras activation induces squamous tumor formation. Expression of mutant KRas in hair follicle bulge stem cells (SCs) and their immediate progeny (hair germ and outer root sheath), but not in their transient amplifying matrix cells, led to benign squamous skin tumor (papilloma). Expression of KRas(G12D) in interfollicular epidermis also led to papilloma formation, demonstrating that squamous tumor initiation is not restricted to the hair follicle lineages. Whereas no malignant tumor was observed after KRas(G12D) expression alone, expression of KRas(G12D) combined with the loss of p53 induced invasive SCC. Our studies demonstrate that different epidermal lineages including bulge SC are competent to initiate papilloma formation and that multiple genetic hits in the context of oncogenic KRas are required for the development of invasive SCC.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Cell Lineage/physiology , Epidermal Cells , Skin Neoplasms/physiopathology , Animals , Bromodeoxyuridine , Carcinoma, Squamous Cell/genetics , DNA Primers/genetics , Flow Cytometry , Hair Follicle/cytology , Integrases/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics , Stem Cells/metabolism
9.
J Immunol ; 185(6): 3417-25, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20733201

ABSTRACT

Th17-mediated immune responses have been recently identified as novel pathogenic mechanisms in a variety of conditions; however, their importance in allograft rejection processes is still debated. In this paper, we searched for MHC or minor Ag disparate models of skin graft rejection in which Th17 immune responses might be involved. We found that T cell-derived IL-17 is critical for spontaneous rejection of minor but not major Ag-mismatched skin grafts. IL-17 neutralization was associated with a lack of neutrophil infiltration and neutrophil depletion delayed rejection, suggesting neutrophils as an effector mechanism downstream of Th17 cells. Regulatory T cells (Tregs) appeared to be involved in Th17 reactivity. We found that in vivo Treg depletion prevented IL-17 production by recipient T cells. An adoptive cotransfer of Tregs with naive monospecific antidonor T cells in lymphopenic hosts biased the immune response toward Th17. Finally, we observed that IL-6 was central for balancing Tregs and Th17 cells as demonstrated by the prevention of Th17 differentiation, the enhanced Treg/Th17 ratio, and a net impact of rejection blockade in the absence of IL-6. In conclusion, the ability of Tregs to promote the Th17/neutrophil-mediated pathway of rejection that we have described should be considered as a potential drawback of Treg-based cell therapy.


Subject(s)
Graft Rejection/genetics , Interleukin-17/physiology , Minor Histocompatibility Antigens/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Cells, Cultured , Female , Gene Knock-In Techniques , Graft Rejection/immunology , Graft Rejection/pathology , Interleukin-17/deficiency , Interleukin-17/genetics , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Signal Transduction/genetics , Signal Transduction/immunology , Skin Transplantation/immunology , Skin Transplantation/pathology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism
10.
J Immunol ; 183(7): 4284-91, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734216

ABSTRACT

In bone marrow-transplanted patients, chronic graft-versus-host disease is a complication that results from the persistent stimulation of recipient minor histocompatibility Ag (mHA)-specific T cells contained within the graft. In this study, we developed a mouse model where persistent stimulation of donor T cells by recipient's mHA led to multiorgan T cell infiltration. Exposure to systemic mHA, however, deeply modified T cell function and chronically stimulated T cells developed a long-lasting state of unresponsiveness, or immune adaptation, characterized by their inability to mediate organ immune damages in vivo. However, analysis of the gene expression profile of adapted CD4+ T cells revealed the specific coexpression of genes known to promote differentiation and function of Th1 effector cells as well as genes coding for proteins that control T cell activity, such as cell surface-negative costimulatory molecules and regulatory cytokines. Strikingly, blockade of negative costimulation abolished T cell adaptation and stimulated strong IFN-gamma production and severe multiorgan wasting disease. Negative costimulation was also shown to control lethal LPS-induced toxic shock in mice with adapted T cells, as well as the capacity of adapted T cells to reject skin graft. Our results demonstrate that negative costimulation is the molecular mechanism used by CD4+ T cells to adapt their activity in response to persistent antigenic stimulation. The effector function of CD4+ T cells that have adapted to chronic Ag presentation can be activated by stimuli strong enough to overcome regulatory signals delivered to the T cells by negative costimulation.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Immune Tolerance , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cells, Cultured , Female , H-Y Antigen/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Molecular Sequence Data , Skin Transplantation/immunology
11.
Transplant Rev (Orlando) ; 23(1): 11-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19027613

ABSTRACT

Interleukin (IL) 17 is a proinflammatory cytokine already known to play a defense role against microbes and a pathogenic role in a number of autoimmune diseases. Although IL-17 can be produced by a variety of cells including neutrophils, CD8+, NK, and gamma-delta T cells, the concept of IL-17-secreting CD4+ T helper cells (Th17), distinct from Th1 and Th2, recently emerged. Herein, we discuss arguments in favor of a Th17-mediated alternative pathway of allograft rejection based on clinical and experimental observations drawn from the literature. We also discuss the complex interplays among regulatory T cells and Th17 cells in the allogeneic context.


Subject(s)
Interleukin-17/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transplantation Immunology , Transplantation, Homologous/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Humans , Immunologic Deficiency Syndromes/immunology , Mice , Neutrophils/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Toll-Like Receptors/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...