Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Immunology ; 168(1): 83-95, 2023 01.
Article in English | MEDLINE | ID: mdl-36054607

ABSTRACT

Integrin-mediated T-cell adhesion and migration is a crucial step in immune response and autoimmune diseases. However, the underlying signalling mechanisms are not fully elucidated. In this study, we examined the implication of purinergic signalling, which has been associated with T-cell activation, in the adhesion and migration of human Th17 cells across fibronectin, a major matrix protein associated with inflammatory diseases. We showed that the adhesion of human Th17 cells to fibronectin induces, via ß1 integrin, a sustained release of adenosine triphosphate (ATP) from the mitochondria through the pannexin-1 hemichannels. Inhibition of ATP release or its degradation with apyrase impaired the capacity of the cells to attach and migrate across fibronectin. Inhibition studies identified a major role for the purinergic receptor P2X4 in T-cell adhesion and migration but not for P2X7 or P2Y11 receptors. Blockade of P2X4 but not P2X7 or P2Y11 receptors reduced cell adhesion and migration by inhibiting activation of ß1 integrins, which is essential for ligand binding. Furthermore, we found that ß1 integrin-induced ATP release, P2X4 receptor transactivation, cell adhesion and migration were dependent on the focal adhesion kinase Pyk2 but not FAK. Finally, P2X4 receptor inhibition also blocked fibronectin-induced Pyk2 activation suggesting the existence of a positive feedback loop of activation between ß1 integrin/Pyk2 and P2X4 purinergic signalling pathways. Our findings uncovered an unrecognized link between ß1 integrin and P2X4 receptor signalling pathways for promoting T-cell adhesion and migration across the extracellular matrix.


Subject(s)
Fibronectins , Integrin beta1 , Humans , Integrin beta1/metabolism , Fibronectins/metabolism , Focal Adhesion Kinase 2/metabolism , Receptors, Purinergic P2X4 , Th17 Cells , Cell Adhesion , Adenosine Triphosphate/metabolism
2.
Int J Mol Sci ; 23(15)2022 Aug 02.
Article in English | MEDLINE | ID: mdl-35955693

ABSTRACT

Synovial fluids from rheumatoid and psoriatic arthritis patients have high levels of PLA1A. The current study was to understand PLA1A functions in the pathophysiology of rheumatic diseases. We generated Pla1a−/− mice to assess their phenotype and the impact of PLA1A deficiency on the development of mannan-induced psoriatic arthritis (MIP). Mice were evaluated routinely for the induced symptoms. On the day of sacrifice, blood samples were collected for hematology analysis and prepared for plasma. Livers were collected. Lymph node immune cells were analyzed using flow cytometry. We performed µCT scans of hind paws from naïve and mannan-induced female mice. Cytokines/chemokines were quantified using Luminex in hind paw tissues and plasma of female mice. Pla1a−/− mice showed a slight increase in circulating and lymph node lymphocytes. CD4+ T cells contributed most to this increase in lymph nodes (p = 0.023). In the MIP model, the lymph node ratios of CD3+ to CD19+ and CD4+ to CD8+ were higher in Pla1a−/− mice. Pla1a−/− mice were less susceptible to MIP (p < 0.001) and showed reduced bone erosions. Pla1a−/− mice also showed reduced IL-17, KC, IP-10, MIP-1ß, LIF, and VEGF in hind paw tissues as compared to WT mice (p < 0.05). These findings indicated that PLA1A deficiency protected from the development of the MIP disease. The data suggested that PLA1A could contribute to MIP through increased activation of lymphocytes, possibly those producing IL-17.


Subject(s)
Arthritis, Psoriatic , Interleukin-17 , Animals , Arthritis, Psoriatic/genetics , Cytokines , Female , Mannans , Mice , Phospholipases A1
3.
J Immunol ; 208(5): 1115-1127, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35165166

ABSTRACT

Purinergic signaling plays a major role in T cell activation leading to IL-2 production and proliferation. However, it is unclear whether purinergic signaling contributes to the differentiation and activation of effector T cells. In this study, we found that the purinergic receptor P2X4 was associated with human Th17 cells but not with Th1 cells. Inhibition of P2X4 receptor with the specific antagonist 5-BDBD and small interfering RNA inhibited the development of Th17 cells and the production of IL-17 by effector Th17 cells stimulated via the CD3/CD28 pathway. Our results showed that P2X4 was required for the expression of retinoic acid-related orphan receptor C, which is the master regulator of Th17 cells. In contrast, inhibition of P2X4 receptor had no effect on Th1 cells and on the production of IFN-γ and it did not affect the expression of the transcription factor T-bet (T-box transcription factor). Furthermore, inhibition of P2X4 receptor reduced the production of IL-17 but not of IFN-γ by effector/memory CD4+ T cells isolated from patients with rheumatoid arthritis. In contrast to P2X4, inhibition of P2X7 and P2Y11 receptors had no effects on Th17 and Th1 cell activation. Finally, treatment with the P2X4 receptor antagonist 5-BDBD reduced the severity of collagen-induced arthritis in mice by inhibiting Th17 cell expansion and activation. Our findings provide novel insights into the role of purinergic signaling in T cell activation and identify a critical role for the purinergic receptor P2X4 in Th17 activation and in autoimmune arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/immunology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/metabolism , Th17 Cells/immunology , Animals , Arthritis, Rheumatoid/pathology , Benzodiazepinones/pharmacology , Cell Differentiation/immunology , Cells, Cultured , Humans , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred DBA , Orphan Nuclear Receptors , RNA Interference , RNA, Small Interfering/genetics , Receptors, Purinergic P2X4/genetics , T-Box Domain Proteins/biosynthesis , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology
4.
Cancers (Basel) ; 13(14)2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34298726

ABSTRACT

Cell adhesion plays a critical role in the development of chemoresistance, which is a major issue in anti-cancer therapies. In this study, we have examined the role of the VLA-4 integrin, a major adhesion molecule of the immune system, in the chemoresistance of T-ALL cells. We found that attachment of Jurkat and HSB-2 T-ALL cells to VCAM-1, a VLA-4 ligand, inhibits doxorubicin-induced apoptosis. However, their adhesion to fibronectin, which is mainly mediated via VLA-5, had no effect. Even the presence of the chemoattractant SDF1α (Stromal cell-derived factor-1α), which enhances the adhesion of T-ALL cells to fibronectin, did not modify the sensitivity of the cells attached on fibronectin towards doxorubicin-induced apoptosis. Mechanistically, we found that VLA-4 promoted T-ALL chemoresistance by inducing doxorubicin efflux. Our results showed that cell adhesion to both fibronectin and VCAM-1-induced Focal adhesion kinase (FAK) phosphorylation in T-ALL cells. However, only cell adhesion to VCAM-1 led to PYK2 phosphorylation. Inhibition studies indicated that FAK is not involved in doxorubicin efflux and chemoresistance, whereas PYK2 inhibition abrogated both VLA-4-induced doxorubicin efflux and chemoresistance. Together, these results indicate that the VLA-4/PYK2 pathway could participate in T-ALL chemoresistance and its targeting could be beneficial to limit/avoid chemoresistance and patient relapse.

5.
J Immunol ; 206(3): 505-514, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33361205

ABSTRACT

High concentrations of the damage-associated molecular patterns S100A8 and S100A9 are found in skin and serum from patients suffering from psoriasis, an IL-17-related disease. Notably, although the expression of these proteins correlates with psoriatic disease severity, the exact function of S100A8 and S100A9 in psoriasis pathogenesis remains unclear. In this study, we investigated the role of S100A8 and S100A9 in psoriasis-associated skin hyperplasia and immune responses using S100a8-/- and S100a9-/- mice in an imiquimod-induced model of psoriasis. We found that S100a8-/- and S100a9-/- psoriatic mice exhibit worsened clinical symptoms relative to wild-type mice and increased expression of S100A9 and S100A8 proteins in keratinocytes, respectively. In addition, the loss of S100A8 enhances proliferation of keratinocytes and disrupts keratinocyte differentiation. We further detected elevated production of IL-17A and -F from CD4+ T cells in the absence of S100A8 and S100A9, as well as increased infiltration of neutrophils in the skin. In addition, treatment with anti-IL-17A and -F was found to reduce psoriasis symptoms and skin hyperplasia in S100a8-/- and S100a9-/- mice. These data suggest that S100A8 and S100A9 regulate psoriasis by inhibiting production of IL-17A and -F, thereby, to our knowledge, providing new insights into their biological functions.


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Interleukin-17/metabolism , Psoriasis/metabolism , Psoriasis/pathology , Skin/pathology , Th17 Cells/immunology , Animals , Antibodies, Blocking/metabolism , Calgranulin A/genetics , Calgranulin B/genetics , Cells, Cultured , Disease Models, Animal , Humans , Hyperplasia , Imiquimod , Interleukin-17/immunology , Mice, Inbred C57BL , Mice, Knockout , Psoriasis/chemically induced
6.
PLoS One ; 15(8): e0235753, 2020.
Article in English | MEDLINE | ID: mdl-32745080

ABSTRACT

CD154 plays a major role in the pathogenesis of several autoimmune and inflammatory diseases. In addition to CD40, soluble CD154 (sCD154) binds to other receptors namely αIIbß3, αMß2, α5ß1 and αvß3 integrins. We have previously reported that binding of sCD154 to α5ß1 integrin expressed on several human T cell lines is capable of inhibiting Fas-induced cell death. In the current study, we show that such effect of the sCD154/α5ß1 interaction is not restricted to the cell death response induced by Fas but could also be exhibited toward other death signals such as TRAIL and TNF- α. We also demonstrate that sCD154 is capable of inhibiting Fas-mediated death of human activated T cells, more importantly of CD4+ than CD8+ T ones. Our data also show that membrane-bound CD154 and α5ß1 integrin expressed on the surface of distinct cells failed to influence cell death responses. However, when membrane-bound CD154 and α5ß1 are expressed on the surface of same cell, their interaction was capable of down regulating cell death. CD154 was shown to co-localize with the α5ß1 integrin on the surface of these cells. These data strongly suggest a cis-type of interaction between CD154 and α5ß1 when both are expressed on the same cell surface, rather than a trans-interaction which usually implicates the ligand and its receptor each expressed on the surface of a distinct cell. Taken together, these findings add to the list of roles through which CD154 is contributing to the pathogenesis of autoimmune-inflammatory diseases, i.e. by protecting T cells from death and enhancing their survival.


Subject(s)
CD40 Ligand/metabolism , Integrin alpha5beta1/metabolism , T-Lymphocytes/cytology , CD40 Ligand/analysis , Cell Death , HEK293 Cells , Humans , Inflammation/metabolism , Integrin alpha5beta1/analysis , Jurkat Cells , Protein Interaction Maps , T-Lymphocytes/metabolism
7.
Mediators Inflamm ; 2020: 2713074, 2020.
Article in English | MEDLINE | ID: mdl-32322163

ABSTRACT

Chemoattractant sensing, adhesiveness, and migration are critical events underlying the recruitment of neutrophils (PMNs) to sites of inflammation or infection. Defects in leukocyte adhesion or migration result in immunodeficiency disorders characterized by recurrent infections. In this study, we evaluated the role of Arf6 on PMN adhesion in vitro and on migration to inflammatory sites using PMN-Arf6 conditional knockout (cKO) mice. In PMN-like PLB-985 silenced for Arf6 fMLP-mediated adhesion to the ß2 integrin ligands, ICAM-1 and fibrinogen or the ß1/ß2 integrin ligand fibronectin was significantly reduced. Furthermore, overexpression of wild-type Arf6 promoted basal and fMLP-induced adhesion to immobilized integrin ligands, while overexpression of the dominant-negative Arf6 has the opposite effects. Using the Elane-Cre deleting mouse strains, we report that the level of Arf6 deletion in inflammatory PMNs isolated from the dorsal air pouches was stronger when compared to naïve cells isolated from the bone marrow. In PMN-Arf6 cKO mice, the recruitment of PMNs into the dorsal air pouch injected with LPS or the chemoattractant fMLP was significantly diminished. Impaired cell migration correlated with reduced cell surface expression of CD11a and CD11b in Arf6 cKO PMNs. Our results highlight that Arf6 regulates the activity and possibly the recycling of PMN integrins, and this compromises PMN migration to inflammatory sites.


Subject(s)
Neutrophils/cytology , Neutrophils/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blood Cell Count , Blotting, Western , Cell Line , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Flow Cytometry , Genetic Vectors/genetics , Humans , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Mice , Mice, Knockout , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Sci Rep ; 9(1): 19455, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31857649

ABSTRACT

Chemoresistance is a major hurdle in anti-cancer therapy. Growing evidence indicates that integrin-mediated cell adhesion to extracellular matrix plays a major role in chemoresistance. However, the underlying mechanisms are not fully understood. We have previously shown that the collagen-binding integrin α2ß1 promoted doxorubicin resistance in acute T cell lymphoblastic leukemia (T-ALL). In this study, we found that acute myeloid leukemia (AML) cell lines also express α2ß1 integrin and collagen promoted their chemoresistance as well. Furthermore, we found that high levels of α2 integrin correlate with worse overall survival in AML. Our results showed that doxorubicin-induced apoptosis in leukemic cells is associated with activation of Ras-related C3 botulinum toxin substrate 1 (Rac1) and that collagen inhibited this pathway. The protective effect of collagen is associated with the inhibition of Rac1-induced DNA damage as evaluated by the comet assay and the phosphorylated levels of histone H2AX (γ-H2AX). Together these results show that by inhibiting pro-apoptotic Rac1, α2ß1 integrin can be a major pathway protecting leukemic cells from genotoxic agents and may thus represent an important therapeutic target in anti-cancer treatment.


Subject(s)
Collagen/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/drug therapy , rac1 GTP-Binding Protein/metabolism , Apoptosis/drug effects , Cell Adhesion , DNA Damage/drug effects , Doxorubicin/therapeutic use , Extracellular Matrix/metabolism , Histones/metabolism , Humans , Integrin alpha2beta1/metabolism , Jurkat Cells , Leukemia, Myeloid, Acute/pathology , Phosphorylation , Signal Transduction/drug effects
9.
PLoS One ; 14(8): e0221528, 2019.
Article in English | MEDLINE | ID: mdl-31437241

ABSTRACT

Expressed strongly by myeloid cells, damage-associated molecular pattern (DAMP) proteins S100A8 and S100A9 are found in the serum of patients with infectious and autoimmune diseases. Compared to S100A9, the role of S100A8 is controversial. We investigated its biological activity in collagen-induced arthritis using the first known viable and fertile S100a8-deficient (S100a8-/-) mouse. Although comparable to the wild type (WT) in terms of lymphocyte distribution in blood and in the primary and secondary lymphoid organs, S100a8-/- mice had increased numbers of neutrophils, monocytes and dendritic cells in the blood and bone marrow, and these all expressed myeloid markers such as CD11b, Ly6G and CD86 more strongly. Granulocyte-macrophage common precursors were increased in S100a8-/- bone marrow and yielded greater numbers of macrophages and dendritic cells in culture. The animals also developed more severe arthritic disease leading to aggravated osteoclast activity and bone destruction. These findings were correlated with increased inflammatory cell infiltration and cytokine secretion in the paws. This study suggests that S100A8 is an anti-inflammatory DAMP that regulates myeloid cell differentiation, thereby mitigating the development of experimental arthritis.


Subject(s)
Arthritis, Experimental/pathology , Calgranulin A/deficiency , Myelopoiesis , Animals , Arthritis, Experimental/diagnostic imaging , Bone Marrow/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calgranulin A/metabolism , Cartilage/pathology , Cell Differentiation , Dendritic Cells/metabolism , Female , Gene Deletion , Mice , Myeloid Cells/pathology
10.
Biochim Biophys Acta Mol Basis Dis ; 1865(10): 2595-2605, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31271845

ABSTRACT

Extracellular nucleotides are released as constitutive danger signals by various cell types and activate nucleotide (P2) receptors such as P2Y6 receptor. P2Y6 activation on monocytes induces the secretion of the chemokine CXCL8 which may propagate intestinal inflammation. Also, P2Y6 expression is increased in infiltrating T cells of Crohn's disease patients. As inflammatory bowel disease (IBD) is associated with immune cell recruitment, we hypothesised that P2Y6 would participate to the establishment of inflammation in this disease. To address this, we used P2Y6 deficient (P2ry6--/-) mice in the dextran sodium sulfate (DSS) murine model of IBD. In disagreement with our hypothesis, P2Y6 deficient mice were more susceptible to inflammation induced by DSS than WT mice. DSS treated-P2ry6-/- mice showed increased histological damage and increased neutrophil and macrophage infiltration that correlated with increased mRNA levels of the chemokines KC and MCP-1. DSS treated-P2ry6-/- mice exhibited also higher levels of Th17/Th1 lymphocytes in their colon which correlated with increased levels of IFN-γ and IL-17A in the sera as well as increased mRNA levels of IFN-γ, IL-17A, IL-6, IL-23 and IL-1ß in P2ry6-/- colons. This inflammation was also accompanied by a decreased cell proliferation and goblet cell number. Importantly, injection of anti-IL-17 intraperitoneally partially protected P2ry6-/- mice from DSS-induced colitis. Taken together, in the absence of P2Y6, an exacerbated intestinal inflammation to DSS was observed which correlated with increased recruitment of Th17/Th1 lymphocytes. These data suggest a protective role of P2Y6 expressed on leukocytes in intestinal inflammation.


Subject(s)
Inflammation/metabolism , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Th17 Cells/metabolism , Animals , Cell Proliferation , Chemokine CCL2/metabolism , Chemokines/metabolism , Colitis/chemically induced , Colitis/metabolism , Colon/metabolism , Cytokines/blood , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Female , Inflammation/chemically induced , Inflammation/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interleukin-8/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/metabolism , Th17 Cells/immunology , Transcriptome
11.
Cell Death Dis ; 10(5): 357, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043590

ABSTRACT

Growing evidence indicates that cell adhesion to extracellular matrix (ECM) plays an important role in cancer chemoresistance. Leukemic T cells express several adhesion receptors of the ß1 integrin subfamily with which they interact with ECM. However, the role of ß1 integrins in chemoresistance of T-cell acute lymphoblastic leukemia (T-ALL) is still ill defined. In this study, we demonstrate that interactions of human T-ALL cell lines and primary blasts with three-dimensional matrices including Matrigel and collagen type I gel promote their resistance to doxorubicin via ß1 integrin. The blockade of ß1 integrin with a specific neutralizing antibody sensitized xenografted CEM leukemic cells to doxorubicin, diminished the leukemic burden in the bone marrow and resulted in the extension of animal survival. Mechanistically, Matrigel/ß1 integrin interaction enhanced T-ALL chemoresistance by promoting doxorubicin efflux through the activation of the ABCC1 drug transporter. Finally, our findings showed that Matrigel/ß1 interaction enhanced doxorubicin efflux and chemoresistance by activating the FAK-related proline-rich tyrosine kinase 2 (PYK2) as both PYK2 inhibitor and siRNA diminished the effect of Matrigel. Collectively, these results support the role of ß1 integrin in T-ALL chemoresistance and suggest that the ß1 integrin pathway can constitute a therapeutic target to avoid chemoresistance and relapsed-disease in human T-ALL.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Focal Adhesion Kinase 2/genetics , Gene Expression Regulation, Leukemic , Integrin beta1/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , Collagen/chemistry , Collagen/metabolism , Collagen Type I/chemistry , Collagen Type I/metabolism , Drug Combinations , Drug Resistance, Neoplasm/genetics , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/metabolism , Humans , Integrin beta1/metabolism , Jurkat Cells , Laminin/chemistry , Laminin/metabolism , Mice, Nude , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Primary Cell Culture , Proteoglycans/chemistry , Proteoglycans/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays
12.
Front Immunol ; 9: 2269, 2018.
Article in English | MEDLINE | ID: mdl-30374344

ABSTRACT

ß1 integrins are critical for T cell migration, survival and costimulation. The integrin α2ß1, which is a receptor for collagen, also named VLA-2, is a major costimulatory pathway of effector T cells and has been implicated in arthritis pathogenesis. Herein, we have examined its ability to promote methotrexate (MTX) resistance by enhancing effector T cells survival. Our results show that attachment of anti-CD3-activated human polarized Th17 cells to collagen but not to fibronectin or laminin led to a significant reduction of MTX-induced apoptosis. The anti-CD3+collagen-rescued cells still produce significant amounts of IL-17 and IFNγ upon their reactivation indicating that their inflammatory nature is preserved. Mechanistically, we found that the prosurvival role of anti-CD3+collagen involves activation of the MTX transporter ABCC1 (ATP Binding Cassette subfamily C Member 1). Finally, the protective effect of collagen/α2ß1 integrin on MTX-induced apoptosis also occurs in memory CD4+ T cells isolated from rheumatoid arthritis (RA) patients suggesting its clinical relevance. Together these results show that α2ß1 integrin promotes MTX resistance of effector T cells, and suggest that it could contribute to the development of MTX resistance that is seen in RA.


Subject(s)
Apoptosis/drug effects , Integrin alpha2beta1/immunology , Methotrexate/pharmacology , Th17 Cells/drug effects , Apoptosis/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cells, Cultured , Collagen/pharmacology , Fibronectins/pharmacology , Humans , Integrin alpha2beta1/metabolism , Laminin/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Protective Agents/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Th17 Cells/immunology
13.
J Cell Biochem ; 118(9): 2819-2827, 2017 09.
Article in English | MEDLINE | ID: mdl-28198034

ABSTRACT

T cell migration across extracellular matrix (ECM) is an important step of the adaptive immune response but is also involved in the development of inflammatory autoimmune diseases. Currently, the molecular mechanisms regulating the motility of effector T cells in ECM are not fully understood. Activation of p38 MAPK has been implicated in T cell activation and is critical to the development of immune and inflammatory responses. In this study, we examined the implication of p38 MAPK in regulating the migration of human Th17 cells through collagen. Using specific inhibitor and siRNA, we found that p38 is necessary for human Th17 migration in three-dimensional (3D) collagen and that 3D collagen increases p38 phosphorylation. We also provide evidence that the collagen receptor, discoidin domain receptor 1 (DDR1), which promotes Th17 migration in 3D collagen, is involved in p38 activation. Together, our findings suggest that targeting DDR1/p38 MAPK pathway could be beneficial for the treatment of Th17-mediated inflammatory diseases. J. Cell. Biochem. 118: 2819-2827, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Cell Movement/immunology , Collagen/chemistry , MAP Kinase Signaling System/immunology , Th17 Cells/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Discoidin Domain Receptor 1/immunology , Enzyme Activation/immunology , Humans
14.
PLoS One ; 11(7): e0158987, 2016.
Article in English | MEDLINE | ID: mdl-27391025

ABSTRACT

CD154, a critical regulator of the immune response, is usually associated with chronic inflammatory, autoimmune diseases as well as malignant disorders. In addition to its classical receptor CD40, CD154 is capable of binding other receptors, members of the integrin family, the αIIbß3, αMß2 and α5ß1. Given the role attributed to integrins and particularly the ß1 integrins in inhibiting apoptotic events in normal as well as malignant T cells, we were highly interested in investigating the role of the CD154/α5ß1 interaction in promoting survival of malignant T cells contributing as such to tumor development and/or propagation. To support our hypothesis, we first show that soluble CD154 binds to the T-cell acute lymphoblastic leukemia cell line, Jurkat E6.1 in a α5ß1-dependent manner. Binding of soluble CD154 to α5ß1 integrin of Jurkat cells leads to the activation of key survival proteins, including the p38 and ERK1/2 mitogen-activated protein kinases (MAPKs), phosphoinositide 3 kinase (PI-3K), and Akt. Interestingly, soluble CD154 significantly inhibits Fas-mediated apoptosis in T cell leukemia-lymphoma cell lines, Jurkat E6.1 and HUT78 cells, an important hallmark of T cell survival during malignancy progression. These anti-apoptotic effects were mainly mediated by the activation of the PI-3K/Akt pathway but also involved the p38 and the ERK1/2 MAPKs cascades. Our data also demonstrated that the CD154-triggered inhibition of the Fas-mediated cell death response was dependent on a suppression of caspase-8 cleavage, but independent of de novo protein synthesis or alterations in Fas expression on cell surface. Together, our results highlight the impact of the CD154/α5ß1 interaction in T cell function/survival and identify novel targets for the treatment of malignant disorders, particularly of T cell origin.


Subject(s)
CD40 Ligand/immunology , Gene Expression Regulation/immunology , MAP Kinase Signaling System/immunology , Receptors, Vitronectin/immunology , T-Lymphocytes/immunology , fas Receptor/immunology , Caspase 8/immunology , Cell Death/immunology , Cell Survival/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Humans , Jurkat Cells
15.
Oncotarget ; 7(29): 44975-44990, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27391444

ABSTRACT

Effector T cell migration through the tissue extracellular matrix (ECM) is an important step of the adaptive immune response and in the development of inflammatory diseases. However, the mechanisms involved in this process are still poorly understood. In this study, we addressed the role of a collagen receptor, the discoidin domain receptor 1 (DDR1), in the migration of Th17 cells. We showed that the vast majority of human Th17 cells express DDR1 and that silencing DDR1 or using the blocking recombinant receptor DDR1:Fc significantly reduced their motility and invasion in three-dimensional (3D) collagen. DDR1 promoted Th17 migration by activating RhoA/ROCK and MAPK/ERK signaling pathways. Interestingly, the RhoA/ROCK signaling module was required for MAPK/ERK activation. Finally, we showed that DDR1 is important for the recruitment of Th17 cells into the mouse dorsal air pouch containing the chemoattractant CCL20. Collectively, our results indicate that DDR1, via the activation of RhoA/ROCK/MAPK/ERK signaling axis, is a key pathway of effector T cell migration through collagen of perivascular tissues. As such, DDR1 can contribute to the development of Th17-dependent inflammatory diseases.


Subject(s)
Cell Movement/physiology , Discoidin Domain Receptor 1/metabolism , MAP Kinase Signaling System/physiology , Th17 Cells/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
16.
J Immunol Res ; 2015: 235170, 2015.
Article in English | MEDLINE | ID: mdl-26609537

ABSTRACT

Polymorphonuclear neutrophils (PMNs) are key innate immune cells that represent the first line of defence against infection. They are the first leukocytes to migrate from the blood to injured or infected sites. This process involves molecular mechanisms that coordinate cell polarization, delivery of receptors, and activation of integrins at the leading edge of migrating PMNs. These phagocytes actively engulf microorganisms or form neutrophil extracellular traps (NETs) to trap and kill pathogens with bactericidal compounds. Association of the NADPH oxidase complex at the phagosomal membrane for production of reactive oxygen species (ROS) and delivery of proteolytic enzymes into the phagosome initiate pathogen killing and removal. G protein-dependent signalling pathways tightly control PMN functions. In this review, we will focus on the small monomeric GTPases of the Arf family and their guanine exchange factors (GEFs) and GTPase activating proteins (GAPs) as components of signalling cascades regulating PMN responses. GEFs and GAPs are multidomain proteins that control cellular events in time and space through interaction with other proteins and lipids inside the cells. The number of Arf GAPs identified in PMNs is expanding, and dissecting their functions will provide important insights into the role of these proteins in PMN physiology.


Subject(s)
GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Neutrophils/enzymology , Neutrophils/immunology , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , Animals , Cell Membrane/immunology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
17.
J Immunol ; 195(9): 4198-209, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26408663

ABSTRACT

Th17 cells are critical effectors in inflammation and tissue damage such as bone erosion, but the mechanisms regulating their activation in this process are not fully understood. In this study, we considered the cooperation between cytokine receptors and integrin pathways in Th17-osteoclast function. We found that human Th17 cells coexpress IL-7R and the collagen-binding integrin α2ß1 (CD49b), and IL-7 increases their adhesion to collagen via α2ß1 integrin. In addition, coengagement of the two receptors in human Th17 cells cooperatively enhanced their IL-17 production and their osteoclastogenic function. The functional cooperation between IL-7R and α2ß1 integrin involves activation of the JAK/PI3K/AKT (protein kinase B) and MAPK/ERK pathways. We also showed that IL-7-induced bone loss in vivo is associated with Th17 cell expansion. Moreover, blockade of α2ß1 integrin with a neutralizing mAb inhibited IL-7-induced bone loss and osteoclast numbers by reducing Th17 cell numbers in the bone marrow and reducing the production of IL-17 and the receptor activator of NF-κB ligand. Thus, the cooperation between IL-7R and α2ß1 integrin can represent an important pathogenic pathway in Th17-osteoclast function associated with inflammatory diseases.


Subject(s)
Bone Resorption/etiology , Integrin alpha2beta1/physiology , Receptors, Interleukin-7/physiology , Th17 Cells/physiology , Cell Adhesion , Cell Polarity , Collagen/pharmacology , Humans , Lymphocyte Activation , MAP Kinase Signaling System , Osteoclasts/physiology , Osteogenesis , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
18.
Semin Cancer Biol ; 35: 145-53, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26297892

ABSTRACT

Extracellular matrix, via its receptors the integrins, has emerged as a crucial factor in cancer development. The α2ß1 integrin is a major collagen receptor that is widely expressed and known to promote cell migration and control tissue homeostasis. Growing evidence suggests that it can be a key pathway in cancer. Recent studies have shown that α2ß1 integrin is a regulator of cancer metastasis either by promoting or inhibiting the dissemination process of cancer cells. The α2ß1 integrin signaling can also enhance tumor angiogenesis. Emerging evidence supports a role for α2ß1 integrin in cancer chemoresistance especially in hematological malignancies originating from the T cell lineage. In addition, α2ß1 integrin has been associated with cancer stem cells. In this review, we will discuss the complex role of α2ß1 integrin in these processes. Collagen is a major matrix protein of the tumor microenvironment and thus, understanding how α2ß1 integrin regulates cancer pathogenesis is likely to lead to new therapeutic approaches and agents for cancer treatment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Drug Resistance, Neoplasm/genetics , Integrin alpha2beta1/genetics , Integrin alpha2beta1/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Cell Movement , Cell Proliferation , Collagen/metabolism , Disease Progression , Extracellular Matrix/metabolism , Humans , Integrin alpha2beta1/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Signal Transduction
19.
Cell Signal ; 26(9): 2008-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24880062

ABSTRACT

Integrin-mediated attachment to extracellular matrix (ECM) is crucial for cancer progression. Malignant T cells such as acute lymphoblastic leukemia (T-ALL) express ß1 integrins, which mediate their interactions with ECM. However, the role of these interactions in T-ALL malignancy is still poorly explored. In the present study, we investigated the effect of collagen; an abundant ECM, on T-ALL survival and migration. We found that collagen through α2ß1 integrin promotes the survival of T-ALL cell lines in the absence of growth factors. T-ALL cell survival by collagen is associated with reduced caspase activation and maintenance of Mcl-1 levels. Collagen activated both ERK and p38 MAPKs but only MAPK/ERK was required for collagen-induced T-ALL survival. However, we found that α2ß1 integrin promoted T-ALL migration via both ERK and p38. Together these data indicate that α2ß1 integrin signaling can represent an important signaling pathway in T-ALL pathogenesis and suggest that its blockade could be beneficial in T-ALL treatment.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Integrin alpha2beta1/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Bone Marrow Cells/cytology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Survival/drug effects , Coculture Techniques , Collagen/pharmacology , Humans , Jurkat Cells , Mesenchymal Stem Cells/cytology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
20.
J Invest Dermatol ; 134(6): 1675-1685, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24499734

ABSTRACT

Tumor-produced extracellular matrix (ECM) proteins can be key elements in tumor growth and metastasis. Transforming growth factor beta-inducible (TGFBI) protein is a secreted ECM component that can have dual function in cancer, acting as tumor suppressor or promoter. Although TGFBI is expressed in human melanoma cells, the exact role it might have in melanoma metastasis remains elusive. Assessing the expression and secretion of TGFBI, we show that human metastatic melanomas express and secrete significantly higher amounts of TGFBI, compared with nevus lesions and primary melanoma tumors. Intravenous injection of highly metastatic human melanoma cells expressing shRNA that targets TGFBI assigns a critical role for TGFBI in the formation of melanoma distal metastases in nude mice. In vivo assays demonstrate that TGFBI silencing does not interfere with melanoma cells' dissemination to distal sites but rather with their proliferation and outgrowth within new microenvironment. In line, TGFBI silencing increases melanoma cells motility/invasion/extravasation in vitro but interferes with their progression through the cell cycle, drastically reducing their proliferation. Furthermore, we show that TGFBI is a regulator of cyclins and cyclin-dependent kinases in melanoma. Collectively, our data describe a mechanism of melanoma metastatic outgrowth via promotion of growth/survival by the ECM protein TGFBI.


Subject(s)
Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Skin Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Biopsy , Cell Adhesion , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Female , Gene Expression Profiling , Gene Silencing , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...