Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Blood Cancer J ; 14(1): 75, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697976

ABSTRACT

Follicular lymphoma (FL), the most common indolent non-Hodgkin lymphoma, constitutes a paradigm of immune tumor microenvironment (TME) contribution to disease onset, progression, and heterogenous clinical outcome. Here we present the first FL-Patient Derived Lymphoma Spheroid (FL-PDLS), including fundamental immune actors and features of TME in FL lymph nodes (LNs). FL-PDLS is organized in disc-shaped 3D structures composed of proliferating B and T cells, together with macrophages with an intermediate M1/M2 phenotype. FL-PDLS recapitulates the most relevant B-cell transcriptional pathways present in FL-LN (proliferation, epigenetic regulation, mTOR, adaptive immune system, among others). The T cell compartment in the FL-PDLS preserves CD4 subsets (follicular helper, regulatory, and follicular regulatory), also encompassing the spectrum of activation/exhaustion phenotypes in CD4 and CD8 populations. Moreover, this system is suitable for chemo and immunotherapy testing, recapitulating results obtained in the clinic. FL-PDLS allowed uncovering that soluble galectin-9 limits rituximab, rituximab, plus nivolumab/TIM-3 antitumoral activities. Blocking galectin-9 improves rituximab efficacy, highlighting galectin-9 as a novel immunotherapeutic target in FL. In conclusion, FL-PDLS maintains the crosstalk between malignant B cells and the immune LN-TME and constitutes a robust and multiplexed pre-clinical tool to perform drug screening in a patient-derived system, advancing toward personalized therapeutic approaches.


Subject(s)
Galectins , Lymph Nodes , Lymphoma, Follicular , Tumor Microenvironment , Humans , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Lymphoma, Follicular/therapy , Lymph Nodes/pathology , Lymph Nodes/immunology , Tumor Microenvironment/immunology , Spheroids, Cellular , Immunotherapy/methods , Signal Transduction , Tumor Cells, Cultured
2.
Leukemia ; 37(6): 1311-1323, 2023 06.
Article in English | MEDLINE | ID: mdl-37031299

ABSTRACT

Mantle cell lymphoma (MCL), a rare and aggressive B-cell non-Hodgkin lymphoma, mainly develops in the lymph node (LN) and creates a protective and immunosuppressive niche that facilitates tumor survival, proliferation and chemoresistance. To capture disease heterogeneity and tumor microenvironment (TME) cues, we have developed the first patient-derived MCL spheroids (MCL-PDLS) that recapitulate tumor oncogenic pathways and immune microenvironment in a multiplexed system that allows easy drug screening, including immunotherapies. MCL spheroids, integrated by tumor B cells, monocytes and autologous T-cells self-organize in disc-shaped structures, where B and T-cells maintain viability and proliferate, and monocytes differentiate into M2-like macrophages. RNA-seq analysis demonstrated that tumor cells recapitulate hallmarks of MCL-LN (proliferation, NF-kB and BCR), with T cells exhibiting an exhaustion profile (PD1, TIM-3 and TIGIT). MCL-PDLS reproduces in vivo responses to ibrutinib and demonstrates that combination of ibrutinib with nivolumab (anti-PD1) may be effective in ibrutinib-resistant cases by engaging an immune response with increased interferon gamma and granzyme B release. In conclusion, MCL-PDLS recapitulates specific MCL-LN features and in vivo responses to ibrutinib, representing a robust tool to study MCL interaction with the immune TME and to perform drug screening in a patient-derived system, advancing toward personalized therapeutic approaches.


Subject(s)
Lymphoma, Mantle-Cell , Humans , Adult , Cell Line, Tumor , Lymphoma, Mantle-Cell/pathology , Drug Resistance, Neoplasm , Adenine/therapeutic use , Tumor Microenvironment
3.
Leukemia ; 35(9): 2635-2649, 2021 09.
Article in English | MEDLINE | ID: mdl-33731849

ABSTRACT

Microenvironment contributes to follicular lymphoma (FL) pathogenesis and impacts survival with macrophages playing a controversial role. In the present study, using FL primary samples and HK follicular dendritic cells (FDC) to mimic the germinal center, together with mouse models, we have analyzed the three-way crosstalk of FL-FDC-macrophages and derived therapeutic opportunities. Ex vivo primary FL-FDC co-cultures (n = 19) and in vivo mouse co-xenografts demonstrated that FL-FDC crosstalk favors tumor growth and, via the secretion of CCL2 and CSF-1, promotes monocyte recruitment, differentiation, and polarization towards an M2-like protumoral phenotype. Moreover, FL-M2 co-cultures displayed enhanced angiogenesis, dissemination, and immunosuppression. Analysis of the CSF-1/CSF-1R pathway uncovered that CSF-1 was significantly higher in serum from grade 3A FL patients, and that high CSF-1R expression in FL biopsies correlated with grade 3A, reduced overall survival and risk of transformation. Furthermore, CSF-1R inhibition with pexidartinib (PLX3397) preferentially affected M2-macrophage viability and polarization program disrupting FL-M2 positive crosstalk. In vivo CSF1-R inhibition caused M2 reduction and repolarization towards M1 macrophages and antitumor effect cooperating with anti-CD20 rituximab. In summary, these results support the role of macrophages in FL pathogenesis and indicate that CSF-1R may be a relevant prognostic factor and a novel therapeutic target cooperating with anti-CD20 immunotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, Follicular/pathology , Macrophages/pathology , Monocytes/pathology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tumor Microenvironment , Aminopyridines/pharmacology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Differentiation , Cell Proliferation , Humans , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation , Pyrroles/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancers (Basel) ; 13(4)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562694

ABSTRACT

Follicular Lymphoma (FL), the most common indolent non-Hodgkin's B cell lymphoma, is a paradigm of the immune microenvironment's contribution to disease onset, progression, and heterogeneity. Over the last few years, state-of-the-art technologies, including whole-exome sequencing, single-cell RNA sequencing, and mass cytometry, have precisely dissected the specific cellular phenotypes present in the FL microenvironment network and their role in the disease. In this already complex picture, the presence of recurring mutations, including KMT2D, CREBBP, EZH2, and TNFRSF14, have a prominent contributory role, with some of them finely tuning this exquisite dependence of FL on its microenvironment. This precise characterization of the enemy (FL) and its allies (microenvironment) has paved the way for the development of novel therapies aimed at dismantling this contact network, weakening tumor cell support, and reactivating the host's immune response against the tumor. In this review, we will describe the main microenvironment actors, together with the current and future therapeutic approaches targeting them.

5.
Blood Adv ; 4(17): 4217-4231, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32898249

ABSTRACT

Despite idelalisib approval in relapsed follicular lymphoma (FL), a complete characterization of the immunomodulatory consequences of phosphatidylinositol 3-kinase δ (PI3Kδ) inhibition, biomarkers of response, and potential combinatorial therapies in FL remain to be established. Using ex vivo cocultures of FL patient biopsies and follicular dendritic cells (FDCs) to mimic the germinal center (n = 42), we uncovered that PI3Kδ inhibition interferes with FDC-induced genes related to angiogenesis, extracellular matrix formation, and transendothelial migration in a subset of FL samples, defining an 18-gene signature fingerprint of idelalisib sensitivity. A common hallmark of idelalisib found in all FL cases was its interference with the CD40/CD40L pathway and induced proliferation, together with the downregulation of proteins crucial for B-T-cell synapses, leading to an inefficient cross talk between FL cells and the supportive T-follicular helper cells (TFH). Moreover, idelalisib downmodulates the chemokine CCL22, hampering the recruitment of TFH and immunosupressive T-regulatory cells to the FL niche, leading to a less supportive and tolerogenic immune microenvironment. Finally, using BH3 profiling, we uncovered that FL-FDC and FL-macrophage cocultures augment tumor addiction to BCL-XL and MCL-1 or BFL-1, respectively, limiting the cytotoxic activity of the BCL-2 inhibitor venetoclax. Idelalisib restored FL dependence on BCL-2 and venetoclax activity. In summary, idelalisib exhibits a patient-dependent activity toward angiogenesis and lymphoma dissemination. In all FL cases, idelalisib exerts a general reshaping of the FL immune microenvironment and restores dependence on BCL-2, predisposing FL to cell death, providing a mechanistic rationale for investigating the combination of PI3Kδ inhibitors and venetoclax in clinical trials.


Subject(s)
Antineoplastic Agents , Lymphoma, Follicular , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Humans , Lymphoma, Follicular/drug therapy , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Tumor Microenvironment
6.
Haematologica ; 105(4): 1032-1041, 2020 04.
Article in English | MEDLINE | ID: mdl-31296574

ABSTRACT

CD38 is expressed in several types of non-Hodgkin lymphoma (NHL) and constitutes a promising target for antibody-based therapy. Daratumumab (Darzalex) is a first-in-class anti-CD38 antibody approved for the treatment of relapsed/refractory (R/R) multiple myeloma (MM). It has also demonstrated clinical activity in Waldenström macroglobulinaemia and amyloidosis. Here, we have evaluated the activity and mechanism of action of daratumumab in preclinical in vitro and in vivo models of mantle cell lymphoma (MCL), follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL), as monotherapy or in combination with standard chemo-immunotherapy. In vitro, daratumumab engages Fc-mediated cytotoxicity by antibody-dependent cell cytotoxicity and antibody-dependent cell phagocytosis in all lymphoma subtypes. In the presence of human serum, complement-dependent cell cytotoxicity was marginally engaged. We demonstrated by Selective Plane Illumination Microscopy that daratumumab fully penetrated a three-dimensional (3D) lymphoma organoid and decreased organoid volume. In vivo, daratumumab completely prevents tumor outgrowth in models of MCL and FL, and shows comparable activity to rituximab in a disseminated in vivo model of blastic MCL. Moreover, daratumumab improves overall survival (OS) in a mouse model of transformed CD20dim FL, where rituximab showed limited activity. Daratumumab potentiates the antitumor activity of CHOP and R-CHOP in MCL and FL xenografts. Furthermore, in a patient-derived DLBCL xenograft model, daratumumab anti-tumor activity was comparable to R-CHOP and the addition of daratumumab to either CHOP or R-CHOP led to full tumor regression. In summary, daratumumab constitutes a novel therapeutic opportunity in certain scenarios and these results warrant further clinical development.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphoma, Non-Hodgkin/therapy , Adult , B-Lymphocytes , Humans , Immunotherapy , Rituximab
7.
Hum Pathol ; 70: 6-13, 2017 12.
Article in English | MEDLINE | ID: mdl-28601659

ABSTRACT

In endometrioid endometrial carcinomas (EECs), microcystic, elongated, and fragmented (MELF) myoinvasion is associated with easily overlooked lymph node metastases; however, the role of immunohistochemistry in their detection and their clinical significance have not been addressed. We identified MELF in 43 of 101 (43%) myoinvasive EECs. Nodes were removed in 49 (49%), 25 with MELF and 24 without MELF. Metastases were initially reported in 3 of the former (12%) and 2 of the latter (8%). All negative nodes were reviewed, and cytokeratin immunohistochemistry was performed. Three metastases were identified in the MELF group but none in the EECs without MELF. By immunohistochemistry, metastatic nodal isolated tumor cells (ITCs) were found in 6 of the remaining 19 MELF-positive cases. In contrast, lymph node metastases were detected in only 2 of the 22 EECs without MELF. MELF-positive cases had more lymph node metastases (P=.03) than myoinvasive EECs without MELF. At follow-up, all 6 patients with grade 1-2 EECs and nodal ITCs/micrometastases were alive (5 no evidence of disease and 1 with perineal disease). In contrast, 3 of 4 patients with grade 3 EECs and nodal ITCs/micrometastases died of disease, and the other patient was alive with tumor. In MELF, the frequency of ITCs/micrometastases in apparently negative lymph nodes is high. In patients with grade 1-2 EEC who had not received chemotherapy, the presence of nodal ITCs/micrometastases did not affect survival. In contrast, in high-grade tumors, ITCs/micrometastases were associated with unfavorable prognosis. Immunohistochemistry should be done in MELF-positive cases to detect occult lymph node metastases, especially in high-grade tumors.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Endometrioid/chemistry , Carcinoma, Endometrioid/secondary , Endometrial Neoplasms/chemistry , Endometrial Neoplasms/pathology , Immunohistochemistry , Lymph Nodes/chemistry , Aged , Aged, 80 and over , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/therapy , Endometrial Neoplasms/mortality , Endometrial Neoplasms/therapy , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Myometrium/chemistry , Myometrium/pathology , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Micrometastasis , Predictive Value of Tests , Retrospective Studies , Risk Factors , Treatment Outcome
8.
Hum Pathol ; 56: 180-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27346574

ABSTRACT

Carcinogenesis is a multistep process in which cancer cells and tumor stroma cells play important roles. T lymphocytes are immune constituents of tumor stroma and play a crucial function in anti-tumor response. By immunohistochemistry and flow cytometry, we studied T cytotoxic (CTLs) and T helper lymphocyte distribution and percentage in the tumor microenvironment and peripheral blood from 35 patients with endometrioid endometrial carcinomas (EEC). We also studied 23 healthy donors' blood samples as a control group. Tumor and non-tumoral endometrium samples were obtained. Immunohistochemistry revealed a high number of CTLs and T helper lymphocytes in the tumor stroma of myoinvasive EECs. T lymphocytes were mostly located in the invasive front. By flow cytometry, the percentages of CTLs and T helper lymphocytes were significantly higher in the tumor compared with the non-neoplastic endometrium (P = .0492 and P = .002). The mean fluorescence intensity of CD8 staining was lower in the tumor compared to the non-neoplastic endometrium (P = .001). There was also reduction of the mean fluorescence intensity of CD8 staining on peripheral blood from patients with grade 3 EECs compare to the peripheral blood from healthy donors (P = .0093). No alterations in the expression of granzymes A and B were found in the CTLs from the EEC cases. Finally, in a proteome profiler cytokine array we found that the growth differentiation factor 15 (GDF15) increased in blood in parallel to the tumor grade. EECs are capable of down-regulating CD8 expression of CTLs. Most likely, this effect is mediated by a soluble molecule present in plasma and is not a result of anergy or exhaustion state.


Subject(s)
Biomarkers, Tumor/analysis , CD8 Antigens/analysis , Carcinoma, Endometrioid/immunology , Endometrial Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Carcinoma, Endometrioid/blood , Carcinoma, Endometrioid/pathology , Cytokines/blood , Down-Regulation , Endometrial Neoplasms/blood , Endometrial Neoplasms/pathology , Female , Growth Differentiation Factor 15/blood , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Middle Aged , Neoplasm Grading , T-Lymphocytes, Cytotoxic/pathology , Tumor Microenvironment
9.
J Immunol ; 192(1): 418-26, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24307736

ABSTRACT

LPS induces the expression of NO synthase 2 (nos2) in macrophages. The expression of this molecule is one of the hallmarks of classical activation. In this paper, we describe that trichostatin A (TSA), which inhibits deacetylase activity, blocks LPS-dependent nos2 expression. TSA specifically inhibits LPS-dependent genes of secondary response, which require new protein synthesis for their induction but not those belonging to the primary response, which do not depend on this process. Deacetylase activity acts at the transcriptional level because RNA polymerase II was not bound after LPS stimulus when we added TSA. A link between the global acetylation caused by HDAC inhibitor and gene promoter recruitment of CDK8 was found. This Mediator complex subunit associates with Med 12, Med13, and cyclin C to form a submodule that is a transcriptional negative regulator. We also found that TSA reduces C/EBPß phosphorylation without affecting its binding to DNA. Taken together, these results shed light on the molecular mechanisms involved in the transcriptional regulation of LPS-treated macrophages and on how TSA targets critical LPS-induced genes, such as nos2 and tnf-α, in inflammatory macrophage response.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/metabolism , Animals , Cyclin-Dependent Kinase 8/metabolism , Gene Expression Regulation/drug effects , Gene Order , Gene Silencing , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/metabolism , Signal Transduction/drug effects , TATA-Box Binding Protein/metabolism , Transcription Initiation, Genetic , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Eur J Immunol ; 42(11): 3028-37, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22865229

ABSTRACT

The amount of arginine available at inflammatory loci is a limiting factor for the growth of several cells of the immune system. IL-4-induced activation of macrophages produced arginase-1, which converts arginine into ornithine, a precursor of polyamines and proline. Trichostatin A (TSA), a pan-inhibitor of histone deacetylases (HDACs), inhibited IL-4-induced arginase-1 expression. TSA showed promoter-specific effects on the IL-4-responsive genes. While TSA inhibited the expression of arginase-1, fizz1, and mrc1, other genes, such as ym,1 mgl1, and mgl2, were not affected. The inhibition of arginase-1 occurred at the transcriptional level with the inhibition of polymerase II binding to the promoter. IL-4 induced STAT6 phosphorylation and binding to DNA. These activities were not affected by TSA treatment. However, TSA inhibited C/EBPß DNA binding. This inhibitor induced acetylation on lysine residues 215-216, which are critical for DNA binding. Finally, using macrophages from STAT6 KO mice we showed that STAT6 is required for the DNA binding of C/EBPß. These results demonstrate that the acetylation/deacetylation balance strongly influences the expression of arginase-1, a gene of alternative activation of macrophages. These findings also provide a molecular mechanism to explain the control of gene expression through deacetylase activity.


Subject(s)
Arginase/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/metabolism , Histone Deacetylase Inhibitors/pharmacology , Interleukin-4/pharmacology , Macrophages/immunology , Acetylation , Animals , Arginase/genetics , Arginase/immunology , CCAAT-Enhancer-Binding Protein-beta/immunology , Hydroxamic Acids/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , STAT6 Transcription Factor/immunology , Statistics, Nonparametric
12.
Mol Immunol ; 47(4): 825-32, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19897249

ABSTRACT

The closest region of the promoter of MHC II genes and particularly three conserved boxes (X, Y and S) are fundamental for the transcriptional regulation. A second set of conserved sequences is present approximately 1200-1500 bp upstream in opposite orientation. In transient transfection experiments in IFN-gamma-treated macrophages and in B lymphocytes, we determined the expression of a fragment of 2035 bp of the I-Abeta gene, which contains the upstream boxes. Mutation of the distal boxes increased induction, thereby suggesting a repressive effect on transcription. In vitro, the proximal and distal ends of I-Abeta promoter were ligated in the presence of nuclear extracts from untreated macrophages but not when the extracts were obtained from IFN-gamma-stimulated cells. The mutation of distal or proximal boxes resulted in a decrease in the ligation assay. The addition of recombinant CIITA to untreated nuclear extracts decreased the capacity of the promoter to be ligated. Finally, we observed increased capacity to ligate the promoter in extracts from B cells lacking CIITA, but not from B cells lacking RFXANK. These results allow us to postulate a model where the proteins in the proximal and distal conserved sequences interact. When CIITA is induced, these proteins make an enhanceosome, allowing chromatin to open and initiate transcription.


Subject(s)
Histocompatibility Antigens Class II/genetics , Locus Control Region/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Trans-Activators/metabolism , Animals , Cell Line , Conserved Sequence , Humans , Mice , Models, Genetic , Nucleic Acid Conformation
13.
J Immunol ; 180(9): 5898-906, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424709

ABSTRACT

After interaction with its receptor, GM-CSF induces phosphorylation of the beta-chain in two distinct domains in macrophages. One induces activation of mitogen-activated protein kinases and the PI3K/Akt pathway, and the other induces JAK2-STAT5. In this study we describe how trichostatin A (TSA), which inhibits deacetylase activity, blocks JAK2-STAT5-dependent gene expression but not the expression of genes that depend on the signal transduction induced by the other domain of the receptor. TSA treatment inhibited the GM-CSF-dependent proliferation of macrophages by interfering with c-myc and cyclin D1 expression. However, M-CSF-dependent proliferation, which requires ERK1/2, was unaffected. Protection from apoptosis, which involves Akt phosphorylation and p21(waf-1) expression, was not modified by TSA. GM-CSF-dependent expression of MHC class II molecules was inhibited because CIITA was not induced. The generation of dendritic cells was also impaired by TSA treatment because of the inhibition of IRF4, IRF2, and RelB expression. TSA mediates its effects by preventing the recruitment of RNA polymerase II to the promoter of STAT5 target genes and by inhibiting their expression. However, this drug did not affect STAT5A or STAT5B phosphorylation or DNA binding. These results in GM-CSF-treated macrophages reveal a relationship between histone deacetylase complexes and STAT5 in the regulation of gene expression.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Histone Deacetylases/immunology , Macrophages/immunology , STAT5 Transcription Factor/immunology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cyclin D , Cyclin-Dependent Kinase Inhibitor p21/immunology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/immunology , Cyclins/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Interferon Regulatory Factor-2/immunology , Interferon Regulatory Factor-2/metabolism , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/immunology , RNA Polymerase II/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
14.
J Immunol ; 176(10): 5918-24, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16670299

ABSTRACT

Arginine is processed by macrophages in response to the cytokines to which these cells are exposed. Th1-type cytokines induce NO synthase 2, which metabolizes arginine into nitrites, while the Th2-type cytokines produce arginase, which converts arginine into polyamines and proline. Activation of bone marrow-derived macrophages by these two types of cytokines increases L-arginine transport only through the y(+) system. Analysis of the expression of the genes involved in this system showed that Slc7A1, encoding cationic amino acid transporters (CAT)1, is constitutively expressed and is not modified by activating agents, while Slc7A2, encoding CAT2, is induced during both classical and alternative activation. Macrophages from Slc7A2 knockout mice showed a decrease in L-arginine transport in response to the two kinds of cytokines. However, while NO synthase 2 and arginase expression were unmodified in these cells, the catabolism of arginine was impaired by both pathways, producing smaller amounts of nitrites and also of polyamines and proline. In addition, the induction of Slc7A2 expression was independent of the arginine available and of the enzymes that metabolize it. In conclusion, the increased arginine transport mediated by activators is strongly regulated by CAT2 expression, which could limit the function of macrophages.


Subject(s)
Arginine/metabolism , Cationic Amino Acid Transporter 2/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Signal Transduction/immunology , Animals , Biological Transport, Active/genetics , Biological Transport, Active/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cationic Amino Acid Transporter 2/physiology , Cells, Cultured , Macrophage Activation/genetics , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...