Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Trends Immunol ; 41(11): 982-993, 2020 11.
Article in English | MEDLINE | ID: mdl-33036910

ABSTRACT

Recent studies have reported paradoxical roles of inflammation in tumor immunity triggered by PD-1 checkpoint antibody (Ab) blockade. Here, we elaborate on this controversy and propose a new perspective that might help understand this paradox. Since inflammatory cytokines and PD-1 blockade are known to target different subsets of exhausted CD8+ T cells, we propose that the timing at which anti-PD-1 Ab therapy and cytokine modulation occur might determine the fate of exhausted CD8+ T cells and perhaps, the clinical outcome of immunotherapeutic modalities.


Subject(s)
Inflammation , Neoplasms , Programmed Cell Death 1 Receptor , Antibodies, Blocking/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Inflammation/immunology , Neoplasms/drug therapy
2.
Nucleic Acids Res ; 48(22): 12874-12888, 2020 12 16.
Article in English | MEDLINE | ID: mdl-32785615

ABSTRACT

A major proportion of extracellular RNAs (exRNAs) do not copurify with extracellular vesicles (EVs) and remain in ultracentrifugation supernatants of cell-conditioned medium or mammalian blood serum. However, little is known about exRNAs beyond EVs. We have previously shown that the composition of the nonvesicular exRNA fraction is highly biased toward specific tRNA-derived fragments capable of forming RNase-protecting dimers. To solve the problem of stability in exRNA analysis, we developed a method based on sequencing the size exclusion chromatography (SEC) fractions of nonvesicular extracellular samples treated with RNase inhibitors (RI). This method revealed dramatic compositional changes in exRNA population when enzymatic RNA degradation was inhibited. We demonstrated the presence of ribosomes and full-length tRNAs in cell-conditioned medium of a variety of mammalian cell lines. Their fragmentation generates some small RNAs that are highly resistant to degradation. The extracellular biogenesis of some of the most abundant exRNAs demonstrates that extracellular abundance is not a reliable input to estimate RNA secretion rates. Finally, we showed that chromatographic fractions containing extracellular ribosomes are probably not silent from an immunological perspective and could possibly be decoded as damage-associated molecular patterns.


Subject(s)
Extracellular Vesicles/genetics , RNA, Transfer/genetics , RNA/genetics , Ribosomes/genetics , Animals , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , High-Throughput Nucleotide Sequencing , Humans , Ribonucleases/antagonists & inhibitors , Ribonucleases/genetics
3.
J Immunol ; 193(9): 4696-703, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25252962

ABSTRACT

Cell therapy and the use of mAbs that interfere with T cell effector functions constitute promising approaches for the control of allograft rejection. In the current study, we investigated a novel approach combining administration of autologous tolerogenic dendritic cells with short-term treatment with CD3-specific Abs. Permanent acceptance of pancreatic islet allografts was achieved in mice treated with the combination therapy the day before transplantation but not in recipients treated with either therapy alone. The combination treatment induced a marked decrease in T cells infiltrating the allografts and a sustained reduction of antidonor responses. Importantly, CD4(+)Foxp3(+) regulatory T cells appeared to play a crucial role in the long-term graft acceptance. Their frequency increased significantly in the spleen, draining lymph nodes, and transplanted islets and remained elevated over the long term; they exhibited increased donor-specific suppressive functions; and their removal at the time of transplantation abrogated the therapeutic effect of the combined therapy. These results support the therapeutic potential of protocols combining autologous dendritic cells and low-dose CD3 Abs, both currently in clinical development, and that act in synergy to control allogeneic immune responses and favor graft survival in a full-mismatch situation.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD3 Complex/metabolism , Dendritic Cells/immunology , Dendritic Cells/transplantation , Islets of Langerhans Transplantation , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Allografts , Animals , Epitopes/immunology , Graft Survival/drug effects , Graft Survival/immunology , Immunomodulation/drug effects , Immunomodulation/immunology , Islets of Langerhans Transplantation/methods , Mice , Models, Animal , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation Tolerance/drug effects , Transplantation Tolerance/immunology , Transplantation, Autologous
4.
Eur J Immunol ; 43(11): 2832-44, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23852701

ABSTRACT

Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. We have previously shown that either induction of HO-1 or treatment with exogenous CO inhibits LPS-induced maturation of dendritic cells (DCs) and protects in vivo and in vitro antigen-specific inflammation. Here, we evaluated the capacity of HO-1 and CO to regulate antigen presentation on MHC class I and MHC class II molecules by LPS-treated DCs. We observed that HO-1 and CO treatment significantly inhibited the capacity of DCs to present soluble antigens to T cells. Inhibition was restricted to soluble OVA protein, as no inhibition was observed for antigenic OVA-derived peptides, bead-bound OVA protein, or OVA as an endogenous antigen. Inhibition of soluble antigen presentation was not due to reduced antigen uptake by DCs, as endocytosis remained functional after HO-1 induction and CO treatment. On the contrary, CO significantly reduced the efficiency of fusion between late endosomes and lysosomes and not by phagosomes and lysosomes. These data suggest that HO-1 and CO can inhibit the ability of LPS-treated DCs to present exogenous soluble antigens to naïve T cells by blocking antigen trafficking at the level of late endosome-lysosome fusion.


Subject(s)
Antigen Presentation/immunology , Carbon Monoxide/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Animals , Antigen Presentation/drug effects , CD4-Positive T-Lymphocytes/immunology , Carbon Monoxide/pharmacology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis/immunology , Endosomes/drug effects , Heme Oxygenase-1/immunology , Heme Oxygenase-1/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Lysosomes/drug effects , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology
5.
J Chromatogr Sci ; 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38553777

ABSTRACT

Following ICH guidelines for analytical validation, we report a common C18 column stability indicating isocratic reverse phase high performance liquid chromatography method for the determination of the ion channel modulator Bay K8644. Two main forced degradation products and a minor impurity were also tentatively identified by Mass Spectrometry. The mobile phase consisted of a 50/50 acetonitrile/buffer mixture at a flow rate of 2 mL/min. Mean retention time for Bay K8644 was 3.030 minutes. Excellent linearity (r = 0.9998) was achieved in the range 0.10-1.40 µg/mL at 274 nm wavelength. Analytical limits were 16.56 ± 1.04 ng/mL for detection and 55.21 ± 3.48 ng/mL for quantitation respectively. Accuracy and precision studies showed good results (95-105%). Robustness was assessed by varying ±3%, both temperature and flow rate. Five different stress conditions were applied to assess Bay K8644's stability. Only basic and photolytic treatments yielded degradation products, both correctly resolved in a total runtime of 4 minutes. In conclusion, we developed a fast, simple, sensitive, accurate, precise, reliable and stability indicating method for detecting/quantifying Bay K8644, and tentatively characterized its main impurities/forced degradation products.

6.
bioRxiv ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38712104

ABSTRACT

The plasma membrane and the membrane of endosomal vesicles are considered physical barriers preventing extracellular RNA uptake. While naked RNA can be spontaneously internalized by certain cells types, functional delivery of naked RNA into the cytosol has been rarely observed. Here we show that extracellular ribonucleases, mainly derived from cell culture supplements, have so far hindered the study of extracellular RNA functionality. In the presence of active ribonuclease inhibitors (RI), naked bacterial RNA is pro-inflammatory when spiked in the media of dendritic cells and macrophages. In murine cells, this response mainly depends on the action of endosomal Toll-like receptors. However, we also show that naked RNA can perform endosomal escape and engage with cytosolic RNA sensors and ribosomes. For example, naked mRNAs encoding reporter proteins can be spontaneously internalized and translated by a variety of cell types, in an RI-dependent manner. In vivo, RI co-injection enhances the activation induced by naked extracellular RNA on splenic lymphocytes and myeloid-derived leukocytes. Furthermore, naked extracellular RNA is inherently pro-inflammatory in ribonuclease-poor compartments such as the peritoneal cavity. Overall, these results demonstrate that naked RNA is bioactive and does not need encapsulation inside synthetic or biological lipid vesicles for functional uptake, making a case for nonvesicular extracellular RNA-mediated intercellular communication.

7.
J Immunol ; 185(2): 823-33, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20543104

ABSTRACT

Despite accumulating evidence for the importance of allospecific CD8(+) regulatory T cells (Tregs) in tolerant rodents and free immunosuppression transplant recipients, mechanisms underlying CD8(+) Treg-mediated tolerance remain unclear. By using a model of transplantation tolerance mediated by CD8(+) Tregs following CD40Ig treatment in rats, in this study, we show that the accumulation of tolerogenic CD8(+) Tregs and plasmacytoid dendritic cells (pDCs) in allograft and spleen but not lymph nodes was associated with tolerance induction in vascularized allograft recipients. pDCs preferentially induced tolerogenic CD8(+) Tregs to suppress CD4(+) effector cells responses to first-donor Ags in vitro. When tolerogenic CD8(+) Tregs were not in contact with CD4(+) effector cells, suppression was mediated by IDO. Contact with CD4(+) effector cells resulted in alternative suppressive mechanisms implicating IFN-gamma and fibroleukin-2. In vivo, both IDO and IFN-gamma were involved in tolerance induction, suggesting that contact with CD4(+) effector cells is crucial to modulate CD8(+) Tregs function in vivo. In conclusion, CD8(+) Tregs and pDCs interactions were necessary for suppression of CD4(+) T cells and involved different mechanisms modulated by the presence of cell contact between CD8(+) Tregs, pDCs, and CD4(+) effector cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Heart Transplantation/immunology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Adenoviridae/genetics , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Dendritic Cells/cytology , Dendritic Cells/immunology , Flow Cytometry , Genetic Vectors/genetics , Heart Transplantation/methods , Male , Models, Animal , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , Transduction, Genetic , Transplantation, Homologous
8.
Front Cell Dev Biol ; 10: 1038429, 2022.
Article in English | MEDLINE | ID: mdl-36340035

ABSTRACT

Characterizing immune regulatory pathways is critical to understand physiological and pathophysiological processes as well as to identify novel immunotherapeutic targets. The cation channel TMEM176B has emerged in the last years as a potential new immunoregulatory player and pharmacological target. Here, we review how expression data, clinical associations of genetic variants and functional studies support a dual role for TMEM176B in regulating immune responses. Thus, TMEM176B can inhibit effector immune responses in some settings whereas it may also promote immunity by supporting antigen presentation in others. We also discuss a potential role for TMEM176B in regulating type 2 and 3 immunity and comment recent data on modulation of DC biology and inflammasome activation as well as CD8+ T cell responses. Understanding the role of TMEM176B in immunity is critical to propose rational pharmacological approaches targeting this channel.

9.
Immunotherapy ; 14(11): 839-842, 2022 08.
Article in English | MEDLINE | ID: mdl-35757836

ABSTRACT

The Federation of Clinical Immunology Societies (FOCIS) regularly organizes scientific meetings to foster advances in immunology. A new event of this type is FOCIS Goes South, a course and workshop organized by FOCIS Centers of Excellence (FCEs) from across Latin America, which consists of a course on advanced immunology, a flow cytometry workshop and seminars on cutting-edge research in autoimmunity, tolerance, cancer, infectious diseases and vaccines. Due to the COVID-19 pandemic, the second version of FOCIS Goes South, hosted by the Millennium Institute on Immunology and Immunotherapy in Chile, took place virtually from 15 to 18 November 2021, with more than 950 registered participants. The present article summarizes the key findings and insights discussed at FOCIS Goes South 2021.


Subject(s)
Allergy and Immunology , COVID-19 , Neoplasms , COVID-19/therapy , Chile , Humans , Immunotherapy , Pandemics
10.
Sci Adv ; 8(38): eabn6545, 2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36129987

ABSTRACT

Severe COVID-19 is associated with hyperinflammation and weak T cell responses against SARS-CoV-2. However, the links between those processes remain partially characterized. Moreover, whether and how therapeutically manipulating T cells may benefit patients are unknown. Our genetic and pharmacological evidence demonstrates that the ion channel TMEM176B inhibited inflammasome activation triggered by SARS-CoV-2 and SARS-CoV-2-related murine ß-coronavirus. Tmem176b-/- mice infected with murine ß-coronavirus developed inflammasome-dependent T cell dysfunction and critical disease, which was controlled by modulating dysfunctional T cells with PD-1 blockers. In critical COVID-19, inflammasome activation correlated with dysfunctional T cells and low monocytic TMEM176B expression, whereas PD-L1 blockade rescued T cell functionality. Here, we mechanistically link T cell dysfunction and inflammation, supporting a cancer immunotherapy to reinforce T cell immunity in critical ß-coronavirus disease.

11.
Mol Ther ; 18(1): 151-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19888197

ABSTRACT

In the absence of an immune response from the host, intramuscular (IM) injection of recombinant adeno-associated virus (rAAV) results in the permanent expression of the transgene from mouse to primate models. However, recent gene transfer studies into animal models and humans indicate that the risk of transgene and/or capsid-specific immune responses occurs and depends on multiple factors. Among these factors, the route of delivery is important, although poorly addressed in large animal models. Here, we compare the IM and the drug-free regional intravenous (RI) deliveries of rAAV in nonhuman primate (NHP) skeletal muscle monitoring the host immune response toward the transgene. We show that IM is consistently associated with immunotoxicity and the destruction of the genetically modified myofibers, whereas RI allows the stable expression of the transgene. This has important implications for the design of clinical trials for gene transfer in skeletal muscle.


Subject(s)
Dependovirus/genetics , Dependovirus/immunology , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Injections, Intravenous/adverse effects , Muscle, Skeletal/metabolism , Transduction, Genetic/methods , Animals , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/genetics , Immunohistochemistry , In Situ Hybridization , Injections, Intramuscular/adverse effects , Macaca , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/drug effects , Reverse Transcriptase Polymerase Chain Reaction
12.
J Clin Invest ; 117(4): 1096-106, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17404623

ABSTRACT

Treatment with CD40Ig results in indefinite allograft survival in a complete MHC-mismatched heart allograft model in the rat. Here we show that serial second, third, and fourth adoptive transfers of total splenocytes from CD40Ig-treated recipients into secondary recipients led to indefinite donor-specific allograft acceptance. Purification of splenocyte subpopulations from CD40Ig-treated recipients demonstrated that only the adoptively transferred CD8(+)CD45RC(low) subset resulted in donor-specific long-term survival, whereas CD8(+)CD45RC(low) T cells from naive animals did not. Accepted grafts displayed increased indoleamine 2,3-dioxygenase (IDO) expression restricted in the graft to ECs. Coculture of donor ECs with CD8(+)CD45RC(low) T cells purified from CD40Ig-treated animals resulted in donor-specific IDO expression dependent on IFN-gamma. Neutralization of IFN-gamma or IDO triggered acute allograft rejection in both CD40Ig-treated and adoptively transferred recipients. This study demonstrates for what we believe to be the first time that interference in CD40-CD40 ligand (CD40-CD40L) interactions induces allospecific CD8(+) Tregs that maintain allograft survival. CD8(+)CD45RC(low) T cells act through IFN-gamma production, which in turn induces IDO expression by graft ECs. Thus, donor alloantigen-specific CD8(+) Tregs may promote local graft immune privilege through IDO expression.


Subject(s)
Graft Survival/physiology , Heart Transplantation/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Interferon-gamma/physiology , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Graft Survival/drug effects , Graft Survival/immunology , Heart Transplantation/immunology , Immune Tolerance/drug effects , Rats , Rats, Inbred Lew , Transplantation, Homologous/immunology
13.
Curr Opin Organ Transplant ; 15(6): 738-43, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20881497

ABSTRACT

PURPOSE OF REVIEW: We discussed the use of autologous tolerogenic dendritic cell (Tol-DC) therapy in organ transplantation, with a particular emphasis on illustrating the reasons why it is a clinically relevant approach and interpreting the experimental data that support this strategy. RECENT FINDINGS: Various parameters are critical for engineering Tol-DCs as a therapeutic tool to manipulate antigen-specific immune responses. Our group has shown that in rats, mice and nonhuman primates, bone marrow progenitors cultured with low doses of granulocyte macrophage colony-stimulating factor can generate Tol-DCs. Injection of autologous Tol-DCs (the same strain as the recipient) is able to significantly prolong allograft survival. Autologous Tol-DCs are more effective than allogeneic Tol-DCs in prolonging allograft survival. Although the reason of this difference remains unclear, it indicates the practical advantages of autologous Tol-DCs as a therapeutic tool in a clinical setting. When autologous Tol-DCs (not pulsed with donor antigens) are administered along with suboptimal immunosuppression treatment, a synergistic effect is achieved, resulting in donor-specific allograft tolerance. SUMMARY: Autologous Tol-DC therapy is a promising approach to improve long-term allograft survival. This strategy may also help reduce the immunosuppressive load in grafted patients and, therefore, limit the harmful effects of immunosuppressive agents.


Subject(s)
Dendritic Cells/transplantation , Graft Survival/immunology , Organ Transplantation , Transplantation Tolerance/immunology , Animals , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immunosuppression Therapy , Immunosuppressive Agents/administration & dosage , Mice , Primates , Rats , Transplantation, Autologous , Vaccination
14.
Sci Rep ; 9(1): 15919, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31685866

ABSTRACT

Male meiotic germ cell including the spermatozoa represent a great challenge to the immune system, as they appear long after the establishment of normal immune tolerance mechanisms. The capacity of the testes to tolerate autoantigenic germ cells as well as survival of allogeneic organ engrafted in the testicular interstitium have led to consider the testis an immunologically privileged site. Disruption of this immune privilege following trauma, tumor, or autoimmune orchitis often results in male infertility. Strong evidence indicates that indoleamine 2,3-dioxygenase (IDO) has been implicated in fetal and allograft tolerance, tumor immune resistance, and regulation of autoimmune diseases. IDO and tryptophan 2,3-dioxygenase (TDO) catalyze the same rate-limiting step of tryptophan metabolism along a common pathway, which leads to tryptophan starvation and generation of catabolites collectively known as kynurenines. However, the relevance of tryptophan metabolism in testis pathophysiology has not yet been explored. Here we assessed the in vivo role of IDO/TDO in experimental autoimmune orchitis (EAO), a model of autoimmune testicular inflammation and immunologically impaired spermatogenesis. EAO was induced in adult Wistar rats with testicular homogenate and adjuvants. Control (C) rats injected with saline and adjuvants and normal untreated rats (N) were also studied. mRNA expression of IDO decreased in whole testes and in isolated Sertoli cells during EAO. TDO and IDO localization and level of expression in the testis were analyzed by immunostaining and Western blot. TDO is expressed in granulomas from EAO rats, and similar protein levels were observed in N, C, and EAO groups. IDO was detected in mononuclear and endothelial cells and reduced IDO expression was detected in EAO group compared to N and C rats. This phenomenon was concomitant with a significant reduction of IDO activity in EAO testis measured by tryptophan and kynurenine concentrations (HPLC). Finally, in vivo inhibition of IDO with 1-methyl-tryptophan increased severity of the disease, demonstrating down regulation of IDO-based tolerance when testicular immune regulation was disrupted. We present evidence that an IDO-based mechanism is involved in testicular immune privilege.


Subject(s)
Immune Privilege , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Testis/enzymology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Disease Models, Animal , Epididymis/pathology , Immune Privilege/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Kynurenine/analysis , Lymph Nodes/enzymology , Lymph Nodes/metabolism , Male , Orchitis/metabolism , Orchitis/pathology , Rats , Rats, Wistar , Sertoli Cells/cytology , Sertoli Cells/metabolism , Severity of Illness Index , Testis/metabolism , Testis/pathology , Tryptophan/analogs & derivatives , Tryptophan/analysis , Tryptophan/metabolism , Tryptophan/pharmacology , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/metabolism
15.
Cancer Cell ; 35(5): 767-781.e6, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31085177

ABSTRACT

Although immune checkpoint blockers have yielded significant clinical benefits in patients with different malignancies, the efficacy of these therapies is still limited. Here, we show that disruption of transmembrane protein 176B (TMEM176B) contributes to CD8+ T cell-mediated tumor growth inhibition by unleashing inflammasome activation. Lack of Tmem176b enhances the antitumor activity of anti-CTLA-4 antibodies through mechanisms involving caspase-1/IL-1ß activation. Accordingly, patients responding to checkpoint blockade therapies display an activated inflammasome signature. Finally, we identify BayK8644 as a potent TMEM176B inhibitor that promotes CD8+ T cell-mediated tumor control and reinforces the antitumor activity of both anti-CTLA-4 and anti-PD-1 antibodies. Thus, pharmacologic de-repression of the inflammasome by targeting TMEM176B may enhance the therapeutic efficacy of immune checkpoint blockers.


Subject(s)
Antineoplastic Agents/pharmacology , Inflammasomes/drug effects , Inflammasomes/immunology , Membrane Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CHO Cells , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetulus , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/metabolism , Xenopus laevis/metabolism
16.
Transplantation ; 84(8): 1060-3, 2007 Oct 27.
Article in English | MEDLINE | ID: mdl-17989614

ABSTRACT

Cytotoxic T lymphocyte-associated antigen 4 immunoglobulin (CTLA4Ig) leads to transplantation tolerance in mice depending on indoleamine 2,3-dioxygenase (IDO). We have shown that CTLA4Ig induces indefinite heart allograft survival in rats and that nitric oxide (NO) was implicated in the in vitro active tolerogenic mechanisms mediated by dendritic cells (DCs). Here we studied the in vivo tolerogenic mechanisms by which CTLA4Ig induces graft survival in rats receiving a cardiac allograft. Treatment of recipients with the IDO inhibitor 1-methyltryptophan (1-MT) did not abrogate the indefinite graft survival observed with CTLA4Ig alone. This was also the case after administration of the inducible nitric oxide synthase inhibitor aminoguanidine when again, indefinite allograft survival was maintained. However, administration of both inhibitors led to acute rejection. We show that IDO and NO are responsible for the impaired capacity of DCs from CTLA4Ig-treated rats to stimulate allogeneic T cells. In conclusion, we show that NO and IDO mediate CTLA4Ig-induced tolerance in rat allograft recipients.


Subject(s)
Graft Survival , Heart Transplantation , Immunoconjugates/pharmacology , Immunosuppressive Agents/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Nitric Oxide/metabolism , Abatacept , Animals , Dendritic Cells/immunology , Enzyme Inhibitors/pharmacology , Male , Nitric Oxide Synthase Type II/antagonists & inhibitors , Rats , Rats, Inbred Lew , Transplantation, Homologous , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
17.
Sci Rep ; 7(1): 1892, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28507328

ABSTRACT

Antimalarials have demonstrated beneficial effects in Systemic Lupus Erithematosus and Rheumatoid Arthritis. However, the mechanisms and the molecular players targeted by these drugs remain obscure. Although hydroxychloroquine (HCQ) is a known ion channel inhibitor, this property has not been linked to its anti-inflammatory effects. We aimed to study whether HCQ inhibits pro-inflammatory ion channels. Electrophysiology experiments demonstrated that HCQ inhibited Ca++-activated K+ conductance in THP-1 macrophages in a dose-dependent manner. In macrophages, ATP-induced K+ efflux plays a key role in activating the NLRP3 inflammasome. ATP-induced IL-1beta secretion was controlled by the KCa1.1 inhibitor iberiotoxin. NS1619 and NS309 (KCa1.1 and KCa3.1 activators respectively) induced the secretion of IL-1beta. This effect was inhibited by HCQ and also by iberiotoxin and clotrimazol (KCa3.1 inhibitor), arguing against off-target effect. In vitro, HCQ inhibited IL-1beta and caspase 1 activation induced by ATP in a dose-dependent manner. HCQ impaired K+ efflux induced by ATP. In vivo, HCQ inhibited caspase 1-dependent ATP-induced neutrophil recruitment. Our results show that HCQ inhibits Ca++-activated K+ channels. This effect may lead to impaired inflammasome activation. These results are the basis for i) a novel anti-inflammatory mechanism for HCQ and ii) a new strategy to target pro-rheumatic Ca++-activated K+ channels.


Subject(s)
Hydroxychloroquine/pharmacology , Inflammasomes/metabolism , Ion Channel Gating/drug effects , Potassium Channels, Calcium-Activated/metabolism , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Caspase 1/genetics , Caspase 1/metabolism , Humans , Mice
18.
FASEB J ; 19(14): 1957-68, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16319139

ABSTRACT

Heme oxygenase-1 (HO-1) is the rate limiting enzyme of heme catabolism whereas indoleamine 2,3 dioxygenase (IDO) catabolizes tryptophan through the kynurenine pathway. We analyzed the expression and biological effects of these enzymes in rat and human breast cancer cell lines. We show that rat (NMU and 13762) but not human cells (MCF-7 and T47D) express HO-1. When overexpressed, we found this enzyme to have anti-proliferative and proapoptotic effects by antioxidant mechanisms in these four cell lines. We show that IDO is expressed by rat and human breast cancer cells. IDO inhibition with 1-MT and siRNA leads to diminished proliferation in rat cells. In contrast, HO-1 negative human cell lines increase proliferation upon IDO inhibition. Since we also demonstrate that IDO inhibits the anti-proliferative HO-1, we propose that IDO has opposite effects on proliferation depending on the coexpression or not of HO-1. We also describe that HO-1 inhibits IDO at the post-translational level through heme starvation. In vivo, we show that rat normal breast expresses HO-1 and IDO. In contrast, N-nitrosomethylurea-induced breast adenocarcinomas only express IDO. In conclusion, we show that HO-1/IDO cross-regulation modulates apoptosis and proliferation in rat and human breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Heme Oxygenase-1/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Mammary Neoplasms, Animal/pathology , Animals , Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Heme/chemistry , Heme Oxygenase-1/metabolism , Humans , Immunohistochemistry , Lentivirus/genetics , Mammary Neoplasms, Animal/metabolism , Methylnitrosourea/pharmacology , Oxygen/metabolism , Protein Processing, Post-Translational , RNA, Small Interfering/metabolism , Rats , Signal Transduction
19.
Methods Mol Biol ; 1371: 89-100, 2016.
Article in English | MEDLINE | ID: mdl-26530796

ABSTRACT

In the last years, cell therapy has become a promising approach to therapeutically manipulate immune responses in autoimmunity, cancer, and transplantation. Several types of lymphoid and myeloid cells origin have been generated in vitro and tested in animal models. Their efficacy to decrease pharmacological treatment has successfully been established. Macrophages play an important role in physiological and pathological processes. They represent an interesting cell population due to their high plasticity in vivo and in vitro. Here, we describe a protocol to differentiate murine regulatory macrophages in vitro from bone marrow precursors. We also describe several methods to assess macrophage classical functions, as their bacterial killing capacity and antigen endocytosis and degradation. Importantly, regulatory macrophages also display suppressive characteristics, which are addressed by the study of their hypostimulatory T lymphocyte capacity and polyclonal T lymphocyte activation suppression.


Subject(s)
Bone Marrow Cells/cytology , Immunomodulation , Macrophages/immunology , Macrophages/metabolism , Animals , Cell Differentiation , Coculture Techniques , Cytotoxicity, Immunologic , Endocytosis/immunology , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/microbiology , Mice , Monocyte-Macrophage Precursor Cells/cytology
20.
Transplantation ; 100(10): 2079-2089, 2016 10.
Article in English | MEDLINE | ID: mdl-27653226

ABSTRACT

BACKGROUND: Regulatory myeloid cell (RMC) therapy is a promising strategy for the treatment of immunological disorders such as autoimmune disease and allograft transplant rejection. Various RMC subsets can be derived from total bone marrow using different protocols, but their phenotypes often overlap, raising questions about whether they are truly distinct. METHODS: In this study, we directly compared the phenotype and function of 3 types of RMCs, tolerogenic dendritic cells, suppressor macrophages, and myeloid-derived suppressor cells, generated in vitro from the same mouse strain in a single laboratory. RESULTS: We show that the 3 RMC subsets tested in this study share some phenotypic markers, suppress T cell proliferation in vitro and were all able to prolong allograft survival in a model of skin transplantation. However, our results highlight distinct mechanisms of action that are specific to each cell population. CONCLUSIONS: This study shows for the first time a side-by-side comparison of 3 types of RMCs using the same phenotypic and functional assays, thus providing a robust analysis of their similarities and differences.


Subject(s)
Dendritic Cells/physiology , Macrophages/physiology , Myeloid-Derived Suppressor Cells/physiology , Adoptive Transfer , Animals , Graft Survival , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL