Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Biomark Insights ; 16: 11772719211047763, 2021.
Article in English | MEDLINE | ID: mdl-34658618

ABSTRACT

Companion diagnostics (CDx) hail promise of improving the drug development process and precision medicine. However, there are various challenges involved in the clinical development and regulation of CDx, which are considered high-risk in vitro diagnostic medical devices given the role they play in therapeutic decision-making and the complications they may introduce with respect to their sensitivity and specificity. The European Union (E.U.) is currently in the process of bringing into effect in vitro Diagnostic Medical Devices Regulation (IVDR). The new Regulation is introducing a wide range of stringent requirements for scientific validity, analytical and clinical performance, as well as on post-market surveillance activities throughout the lifetime of in vitro diagnostics (IVD). Compliance with General Safety and Performance Requirements (GSPRs) adopts a risk-based approach, which is also the case for the new classification system. This changing regulatory framework has an impact on all stakeholders involved in the IVD Industry, including Authorized Representatives, Distributors, Importers, Notified Bodies, and Reference Laboratories and is expected to have a significant effect on the development of new CDx.

2.
Oncogene ; 39(33): 5553-5569, 2020 08.
Article in English | MEDLINE | ID: mdl-32655131

ABSTRACT

Metastasis is the leading cause of cancer-related death. This multistage process involves contribution from both tumour cells and the tumour stroma to release metastatic cells into the circulation. Circulating tumour cells (CTCs) survive circulatory cytotoxicity, extravasate and colonise secondary sites effecting metastatic outcome. Reprogramming the transcriptomic landscape is a metastatic hallmark, but detecting underlying master regulators that drive pathological gene expression is a key challenge, especially in childhood cancer. Here we used whole tumour plus single-cell RNA-sequencing in primary bone cancer and CTCs to perform weighted gene co-expression network analysis to systematically detect coordinated changes in metastatic transcript expression. This approach with comparisons applied to data collected from cell line models, clinical samples and xenograft mouse models revealed mitogen-activated protein kinase 7/matrix metallopeptidase 9 (MAPK7/MMP9) signalling as a driver for primary bone cancer metastasis. RNA interference knockdown of MAPK7 reduces proliferation, colony formation, migration, tumour growth, macrophage residency/polarisation and lung metastasis. Parallel to these observations were reduction of activated interleukins IL1B, IL6, IL8 plus mesenchymal markers VIM and VEGF in response to MAPK7 loss. Our results implicate a newly discovered, multidimensional MAPK7/MMP9 signalling hub in primary bone cancer metastasis that is clinically actionable.


Subject(s)
Bone Neoplasms/complications , Mitogen-Activated Protein Kinase 7/metabolism , Animals , Bone Neoplasms/genetics , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Metastasis
3.
Immunohorizons ; 4(7): 382-391, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32631900

ABSTRACT

Hematopoietic progenitor kinase 1 (HPK1) is a negative regulator of TCR-initiated signal transduction. Both the HPK1-/- mice and the genetically engineered mice with a point mutation that disrupts the catalytic activity of HPK1 possess enhanced antitumor immunity, especially when these mice are treated with anti-PD-L1 immune checkpoint Ab. Because CD4+FOXP3+ regulatory T cells (Tregs) play an important role in suppressing tumor immunity, we investigated whether the loss of HPK1 expression could result in the reduction of Treg functions. We found that the number of HPK1-/- Tregs is elevated relative to the number found in wild-type C57/BL6 mice. However, HPK1-/- Tregs lack the ability to carry out effective inhibition of TCR-induced proliferative responses by effector T cells. Furthermore, HPK1-/- Tregs respond to TCR engagement with an elevated and sustained Erk MAPK and p65/RelA NF-κB phosphorylation in comparison with wild-type Tregs. Also, a multiplex cytokine analysis of HPK1-/- Tregs revealed that they demonstrate an aberrant cytokine expression profile when stimulated by anti-CD3ε and anti-CD28 crosslinking, including the uncharacteristic expression of IL-2 and antitumor proinflammatory cytokines and chemokines such as IFN-γ, CCL3, and CCL4. The aberrant HPK1-/- phenotype observed in these studies suggests that HPK1 may play an important role in maintaining Treg functions with wider implications for HPK1 as a novel immunotherapeutic target.


Subject(s)
NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Mice , NF-kappa B/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , T-Lymphocytes, Regulatory/cytology
4.
Clin Rev Allergy Immunol ; 53(2): 265-276, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28488248

ABSTRACT

Biologic TNFα inhibitors are a mainstay treatment option for patients with rheumatoid arthritis (RA) refractory to other treatment options. However, many patients either do not respond or relapse after initially responding to these agents. This study was carried out to identify biomarkers that can distinguish responder from non-responder patients before the initiation of treatment. The level of cytokines in plasma and those produced by ex vivo T cells, B cells and monocytes in 97 RA patients treated with biologic TNFα inhibitors was measured before treatment and after 1 and 3 months of treatment by multiplex analyses. The frequency of T cell subsets and intracellular cytokines were determined by flow cytometry. The results reveal that pre-treatment, T cells from patients who went on to respond to treatment with biologic anti-TNFα agents produced significantly more GM-CSF than non-responder patients. Furthermore, immune cells from responder patients produced higher levels of IL-1ß, TNFα and IL-6. Cytokine profiling in the blood of patients confirmed the association between high levels of GM-CSF and responsiveness to biologic anti-TNFα agents. Thus, high blood levels of GM-CSF pre-treatment had a positive predictive value of 87.5% (61.6 to 98.5% at 95% CI) in treated RA patients. The study also shows that cells from most anti-TNFα responder patients in the current cohort produced higher levels of GM-CSF and TNFα pre-treatment than non-responder patients. Findings from the current study and our previous observations that non-responsiveness to anti-TNFα is associated with high IL-17 levels suggest that the disease in responder and non-responder RA patients is likely to be driven/sustained by different inflammatory pathways. The use of biomarker signatures of distinct pro-inflammatory pathways could lead to evidence-based prescription of the most appropriate biological therapies for different RA patients.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , B-Lymphocytes/immunology , Biomarkers, Pharmacological/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-17/metabolism , T-Lymphocytes/immunology , Adult , Aged , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/diagnosis , Cells, Cultured , Female , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation , Male , Middle Aged , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
5.
PLoS Pathog ; 9(9): e1003627, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068934

ABSTRACT

Rheumatoid arthritis and periodontitis are two prevalent chronic inflammatory diseases in humans and are associated with each other both clinically and epidemiologically. Recent findings suggest a causative link between periodontal infection and rheumatoid arthritis via bacteria-dependent induction of a pathogenic autoimmune response to citrullinated epitopes. Here we showed that infection with viable periodontal pathogen Porphyromonas gingivalis strain W83 exacerbated collagen-induced arthritis (CIA) in a mouse model, as manifested by earlier onset, accelerated progression and enhanced severity of the disease, including significantly increased bone and cartilage destruction. The ability of P. gingivalis to augment CIA was dependent on the expression of a unique P. gingivalis peptidylarginine deiminase (PPAD), which converts arginine residues in proteins to citrulline. Infection with wild type P. gingivalis was responsible for significantly increased levels of autoantibodies to collagen type II and citrullinated epitopes as a PPAD-null mutant did not elicit similar host response. High level of citrullinated proteins was also detected at the site of infection with wild-type P. gingivalis. Together, these results suggest bacterial PAD as the mechanistic link between P. gingivalis periodontal infection and rheumatoid arthritis.


Subject(s)
Arthritis/microbiology , Bacterial Proteins/metabolism , Bacteroidaceae Infections/microbiology , Disease Models, Animal , Hydrolases/metabolism , Periodontitis/microbiology , Porphyromonas gingivalis/enzymology , Animals , Arthritis/immunology , Arthritis/pathology , Arthritis/physiopathology , Autoantibodies/analysis , Bacterial Proteins/genetics , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/pathology , Bacteroidaceae Infections/physiopathology , Bone Resorption/etiology , Citrulline/metabolism , Disease Progression , Gene Deletion , Hydrolases/genetics , Joints/immunology , Joints/metabolism , Joints/microbiology , Joints/pathology , Male , Mice, Inbred DBA , Neutrophil Infiltration , Periodontitis/immunology , Periodontitis/metabolism , Periodontitis/pathology , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/isolation & purification , Prevotella intermedia/enzymology , Prevotella intermedia/immunology , Prevotella intermedia/isolation & purification , Protein Processing, Post-Translational , Protein-Arginine Deiminases , Severity of Illness Index
6.
Arthritis Res Ther ; 14(3): R142, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22691272

ABSTRACT

INTRODUCTION: Endosomal toll-like receptors (TLRs) have recently emerged as potential contributors to the inflammation observed in human and rodent models of rheumatoid arthritis (RA). This study aims to evaluate the role of endosomal TLRs and in particular TLR7 in the murine collagen induced arthritis (CIA) model. METHODS: CIA was induced by injection of collagen in complete Freund's adjuvant. To investigate the effect of endosomal TLRs in the CIA model, mianserin was administered daily from the day of disease onset. The specific role of TLR7 was examined by inducing CIA in TLR7-deficient mice. Disease progression was assessed by measuring clinical score, paw swelling, serum anti-collagen antibodies histological parameters, cytokine production and the percentage of T regulatory (Treg) cells. RESULTS: Therapeutic administration of mianserin to arthritic animals demonstrated a highly protective effect on paw swelling and joint destruction. TLR7-/- mice developed a mild arthritis, where the clinical score and paw swelling were significantly compromised in comparison to the control group. The amelioration of arthritis by mianserin and TLR7 deficiency both corresponded with a reduction in IL-17 responses, histological and clinical scores, and paw swelling. CONCLUSIONS: These data highlight the potential role for endosomal TLRs in the maintenance of inflammation in RA and support the concept of a role for TLR7 in experimental arthritis models. This study also illustrates the potential benefit that may be afforded by therapeutically inhibiting the endosomal TLRs in RA.


Subject(s)
Arthritis, Experimental/metabolism , Endosomes/metabolism , Membrane Glycoproteins/metabolism , Toll-Like Receptor 7/metabolism , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Male , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 7/deficiency
7.
Ann Rheum Dis ; 71(10): 1741-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22550316

ABSTRACT

OBJECTIVES: To establish if changes in Th1/Th17 cell populations previously reported in experimental arthritis occur in patients with rheumatoid arthritis (RA) treated with anti-tumour necrosis factor α (TNFα) agents, and whether the therapeutic response to anti-TNFα is compromised in patients and mice because of elevated Th17/IL-17 levels. Finally, to assess the efficacy of combined blockade of anti-TNFα and anti-IL-17 in experimental arthritis. METHODS: A longitudinal study of two independent cohorts (cohort 1, n=24; cohort 2, n=19) of patients with RA treated with anti-TNFα biological agents was carried out to assess their Th17/IL-17 levels before and after the start of anti-TNFα therapy. IL-12/23p40 production was assessed in plasma Peripheral blood lymphocytes (PBLs) and monocytes. Mice with collagen-induced arthritis (CIA) were treated with anti-TNFα alone, anti-IL17 alone or a combination of the two. Efficacy of treatment and response was assessed from changes in Disease Activity Score 28-erythrocyte sedimentation rate scores in patients, and in clinical scores and histological analysis in CIA. RESULTS: Significant increases in circulating Th17 cells were observed in patients after anti-TNFα therapy and this was accompanied by increased production of IL-12/23p40. There was an inverse relationship between baseline Th17 levels and the subsequent response of patients with RA to anti-TNFα therapy. In addition, PBLs from non-responder patients showed evidence of increased IL-17 production. Similarly, in anti-TNFα-treated mice, there was a strong correlation between IL-17 production and clinical score. Finally combined blockade of TNFα and IL-17 in CIA was more effective than monotherapy, particularly with respect to the duration of the therapeutic effect. CONCLUSIONS: These findings, which need to be confirmed in a larger cohort, suggest that a Th17-targeted therapeutic approach may be useful for anti-TNFα non-responder patients or as an adjunct to anti-TNFα therapy, provided that safety concerns can be addressed.


Subject(s)
Arthritis, Rheumatoid/immunology , Drug Resistance/immunology , Interleukin-17/immunology , Th17 Cells/immunology , Animals , Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/drug therapy , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Longitudinal Studies , Male , Mice , Middle Aged , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/antagonists & inhibitors
9.
J Immunol Methods ; 373(1-2): 229-39, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21910993

ABSTRACT

A cell-based assay has been developed for the quantification of the activity of TNFα antagonists based on human erythroleukemic K562 cells transfected with a NFκB regulated firefly luciferase reporter-gene construct. Both drug activity and anti-drug neutralizing antibodies can be quantified with a high degree of precision within 2h, and without interference from cytokines and other factors known to activate NFκB. The assay cells also contain the Renilla luciferase reporter gene under the control of a constitutive promoter that allows TNFα-induced firefly luciferase activity to be normalized relative to Renilla luciferase expression. Thus, results are independent of cell number or differences in cell viability, resulting in intra and inter assay coefficients of variation of 10% or less. Normalization of results relative to the expression of an internal standard also provides a means for correcting for serum matrix effects and allows residual drug levels or anti-drug neutralizing antibodies to be quantified even in serum samples with a relatively high degree of cytotoxicity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/analysis , Luciferases/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Crohn Disease/blood , Crohn Disease/drug therapy , Culture Media/chemistry , Culture Media/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Humans , Infliximab , K562 Cells , Luciferases/genetics , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Luciferases, Renilla/genetics , Luciferases, Renilla/metabolism , NF-kappa B/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Serum/chemistry , Serum/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
10.
Arthritis Rheum ; 63(12): 3818-23, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21953289

ABSTRACT

OBJECTIVE: To examine the hypothesis that the subset of rheumatoid arthritis (RA) characterized by antibodies to citrullinated α-enolase is mediated by Porphyromonas gingivalis enolase in the context of DR4 alleles. METHODS: Recombinant human α-enolase and P gingivalis enolase, either citrullinated or uncitrullinated, were used to immunize DR4-IE-transgenic mice and control mice (class II major histocompatibility complex-deficient [class II MHC(-/-)] and C57BL/6 wild-type mice). Arthritis was quantified by measurement of ankle swelling in the hind paws and histologic examination. Serum IgG reactivity with α-enolase and citrullinated α-enolase was assayed by Western blotting and enzyme-linked immunosorbent assay (ELISA). Antibodies to peptide 1 of citrullinated α-enolase (CEP-1) and its arginine-bearing control peptide, REP-1, were also assessed by ELISA. RESULTS: Significant hind-ankle swelling (≥0.3 mm) occurred in DR4-IE-transgenic mice immunized with citrullinated human α-enolase (9 of 12 mice), uncitrullinated human α-enolase (9 of 12 mice), citrullinated P gingivalis enolase (6 of 6 mice), and uncitrullinated P gingivalis enolase (6 of 6 mice). Swelling peaked on day 24. None of the control groups developed arthritis. The arthritic joints showed synovial hyperplasia and erosions, but there was a paucity of leukocyte infiltration. Antibodies to human α-enolase, both citrullinated and unmodified, and to CEP-1 and REP-1 were detectable in all immunized mice except the class II MHC(-/-) control mice. CONCLUSION: This is the first animal model that links an immune response to P gingivalis enolase to an important subset of RA, defined by antibodies to citrullinated α-enolase in the context of DR4. The fact that arthritis and anti-CEP-1 antibodies were induced independent of citrullination of the immunizing antigen suggests that the unmodified form of α-enolase may be important in initiating the corresponding subset of human RA.


Subject(s)
Arthritis, Experimental/immunology , Autoimmunity/drug effects , HLA-DR4 Antigen/genetics , Immunization , Phosphopyruvate Hydratase/pharmacology , Porphyromonas gingivalis/enzymology , Animals , Arthritis, Experimental/blood , Arthritis, Experimental/chemically induced , Autoimmunity/immunology , Disease Models, Animal , Histocompatibility Antigens Class II/genetics , Humans , Immunoglobulin G/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphopyruvate Hydratase/adverse effects , Phosphopyruvate Hydratase/immunology , Recombinant Proteins/pharmacology
11.
Pain ; 152(8): 1838-1845, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21507574

ABSTRACT

Interleukin-17 (IL-17) and tumour necrosis factor-α (TNF) are critical in the pathogenesis of arthritis but their relationship during inflammatory pain has received limited attention. We aimed to establish whether IL-17 can induce hyperalgesia in acute conditions, and investigated the role of TNF in mediating the pain response. Hyperalgesia was elicited in C57BL/6 mice by injection of recombinant IL-17, TNF or vehicle into the plantar tissue. Elevated pain was measured by the Hargreaves test for thermal hyperalgesia and Linton incapacitance tester for weight-bearing change. Cellular infiltration during hyperalgesia was determined by histological analysis and myeloperoxidase assay. IL-17 was found to induce hyperalgesia, but this was dependent on neutrophil migration and TNF binding to TNF receptor 1 (TNFR1). Because TNF-induced hyperalgesia was also dependent on neutrophil migration, the relationship between the resident fibroblasts, the cytokines and the migrating neutrophils was further investigated. By means of an air pouch model of cell migration, it was established that IL-17-induced neutrophil infiltration was dependent of TNF/TNFR1 as this interaction was required for the induction of the chemokine keratinocyte chemoattractant. These findings suggest that IL-17 causes acute hyperalgesia indirectly by inducing TNF from resident cells. The subsequent production of keratinocyte chemoattractant then triggers neutrophil chemotaxis to the plantar tissue, releasing algesic mediators locally to sensitise the nerve.


Subject(s)
Hyperalgesia/chemically induced , Interleukin-17/adverse effects , Neutrophil Infiltration/drug effects , Pain Threshold/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Enzyme-Linked Immunosorbent Assay , Hyperalgesia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor, Type I/deficiency , Time Factors , Tumor Necrosis Factor-alpha/adverse effects
12.
FEBS Lett ; 585(23): 3649-59, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21515267

ABSTRACT

The development of an immune response to self antigens drives naive T cells to differentiate into subsets of CD8(+) and CD4(+) effector cells including T(H)1, T(H)2, cells and the more recently described T(H)17, and regulatory T cells (T(reg)). Rheumatoid arthritis is an autoimmune disease that engages an uncontrolled influx of inflammatory cells to the joints, eventually leading to joint damage. The role that effector T cells play in the local or systemic maintenance of, or protection against, inflammation and subsequent joint damage is now becoming better understoodthrough the use of animal models. In this review, we will explore the different animal models of RA, and their contribution to elucidating the role that effector T cells play in the regulation, induction, and maintenance of inflammatory joint disease. This understanding will aid in the design of more effective therapeutic strategies for rheumatoid arthritis and other autoimmune disorders.


Subject(s)
Arthritis, Rheumatoid/immunology , Disease Models, Animal , T-Lymphocytes/immunology , Animals , Arthritis, Rheumatoid/pathology , Cytokines/immunology , Humans
13.
Nat Immunol ; 12(3): 231-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21240265

ABSTRACT

Polymorphisms in the gene encoding the transcription factor IRF5 that lead to higher mRNA expression are associated with many autoimmune diseases. Here we show that IRF5 expression in macrophages was reversibly induced by inflammatory stimuli and contributed to the plasticity of macrophage polarization. High expression of IRF5 was characteristic of M1 macrophages, in which it directly activated transcription of the genes encoding interleukin 12 subunit p40 (IL-12p40), IL-12p35 and IL-23p19 and repressed the gene encoding IL-10. Consequently, those macrophages set up the environment for a potent T helper type 1 (T(H)1)-T(H)17 response. Global gene expression analysis demonstrated that exogenous IRF5 upregulated or downregulated expression of established phenotypic markers of M1 or M2 macrophages, respectively. Our data suggest a critical role for IRF5 in M1 macrophage polarization and define a previously unknown function for IRF5 as a transcriptional repressor.


Subject(s)
Interferon Regulatory Factors/immunology , Macrophages/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Flow Cytometry , Humans , Immunoblotting , Interferon Regulatory Factors/genetics , Mice , Mice, Knockout , Microarray Analysis
14.
Cancer Immunol Immunother ; 59(3): 419-29, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19787351

ABSTRACT

Lung cancer is the leading cause of cancer-related mortality in the world, resulting in over a million deaths each year. Non-small cell lung cancers (NSCLCs) are characterized by a poor immunogenic response, which may be the result of immunosuppressive factors such as prostaglandin E2 (PGE(2)) present in the tumor environment. The effect of PGE(2) in the suppression of anti-tumor immunity and its promotion of tumor survival has been established for over three decades, but with limited mechanistic understanding. We have previously reported that PGE(2) activates hematopoietic progenitor kinase 1 (HPK1), a hematopoietic-specific kinase known to negatively regulate T-cell receptor signaling. Here, we report that mice genetically lacking HPK1 resist the growth of PGE(2)-producing Lewis lung carcinoma (LLC). The presence of tumor-infiltrating lymphocytes (TILs) and T-cell transfer into T cell-deficient mice revealed that tumor rejection is T cell mediated. Further analysis demonstrated that this may be significantly due to the ability of HPK1 (-/-) T cells to withstand PGE(2)-mediated suppression of T-cell proliferation, IL-2 production, and apoptosis. We conclude that PGE(2) utilizes HPK1 to suppress T cell-mediated anti-tumor responses.


Subject(s)
Carcinoma, Lewis Lung/immunology , Dinoprostone/physiology , Protein Serine-Threonine Kinases/physiology , T-Lymphocytes/immunology , Tumor Escape , Animals , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Serine-Threonine Kinases/genetics
15.
J Immunol ; 182(10): 6187-94, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414772

ABSTRACT

Hematopoietic progenitor kinase 1 (HPK1) is a hematopoietic cell-restricted member of the Ste20 kinases that acts as a negative regulator of T cell functions through the AP-1, NFAT, and NFkappaB pathways. Using HPK1-deficient (HPK1(-/-)) mice, we report in this study a novel role for HPK1 in dendritic cells (DCs). Specifically, we observed that matured HPK1(-/-) bone marrow-derived DCs (BMDCs) are superior to their wild-type (WT) counterpart in stimulating T cell proliferation in vivo and in vitro. Several characteristics of HPK1(-/-) BMDCs may account for this enhanced activity: Matured HPK1(-/-) BMDCs express higher levels of costimulatory molecules CD80, CD86, and I-A(b) as well as produce more proinflammatory cytokines IL-12, IL-1beta, TNF-alpha, and IL-6 than their WT littermates. The role of HPK1 as a proapoptotic molecule was assessed post activation with LPS, and results indicated that HPK1(-/-) BMDCs are significantly resistant to LPS-induced apoptosis. Our results led us to investigate the role of HPK1(-/-) BMDCs in tumor immunotherapy. Using a s.c. murine model of Lewis Lung Carcinoma, we found that HPK1(-/-) BMDCs eliminate established s.c. Lewis Lung Carcinoma more efficiently than their WT counterpart. Our data reveal a novel role for HPK1 as a negative regulator of DC functions, identifying its potential as a molecular target for DC-based immunotherapy against cancers.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Protein Serine-Threonine Kinases/immunology , Animals , Apoptosis/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Carcinoma, Lewis Lung/immunology , Cell Differentiation/immunology , Cytokines/biosynthesis , Dendritic Cells/cytology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , T-Lymphocytes/immunology
16.
J Exp Med ; 205(11): 2491-7, 2008 Oct 27.
Article in English | MEDLINE | ID: mdl-18936235

ABSTRACT

IL-17 is implicated in the pathogenesis of rheumatoid arthritis (RA) and has previously been shown to be induced by tumor necrosis factor (TNF) in vitro. The aim of this study was to assess the impact of TNF inhibition on IL-17 production in collagen-induced arthritis, a model of RA. TNF blockade using TNFR-Fc fusion protein or anti-TNF monoclonal antibody reduced arthritis severity but, unexpectedly, expanded populations of Th1 and Th17 cells, which were shown by adoptive transfer to be pathogenic. Th1 and Th17 cell populations were also expanded in collagen-immunized TNFR p55(-/-) but not p75(-/-) mice. The expression of IL-12/IL-23 p40 was up-regulated in lymph nodes (LN) from p55(-/-) mice, and the expansion of Th1/Th17 cells was abrogated by blockade of p40. Treatment of macrophages with rTNF also inhibited p40 production in vitro. These findings indicate that at least one of the ways in which TNF regulates Th1/Th17 responses in arthritis is by down-regulating the expression of p40. Finally, although TNF blockade increased numbers of Th1 and Th17 cells in LN, it inhibited their accumulation in the joint, thereby providing an explanation for the paradox that anti-TNF therapy ameliorates arthritis despite increasing numbers of pathogenic T cells.


Subject(s)
Arthritis, Experimental/immunology , Immunoglobulin G/pharmacology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adoptive Transfer , Animals , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Etanercept , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Receptors, Tumor Necrosis Factor , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
17.
J Biol Chem ; 282(48): 34693-9, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17895239

ABSTRACT

Hematopoietic progenitor kinase 1 (HPK1) is a hematopoietic cell-restricted member of the Ste20 serine/threonine kinase super family. We recently reported that the immunosuppressive eicosanoid, prostaglandin E(2) (PGE(2)), is capable of activating HPK1 in T cells. In this report, we demonstrate that unlike the TCR-induced activation of HPK1 kinase activity, the induction of HPK1 catalytic activity by PGE(2) does not require the presence of phosphotyrosine-based signaling molecules such as Lck, ZAP-70, SLP-76, and Lat. Nor does the PGE(2)-induced HPK1 activation require the intermolecular interaction between its proline-rich regions and the SH3 domain-containing adaptor proteins, as required by the signaling from the TCR to HPK1. Instead, our study reveals that PGE(2) signal to HPK1 via a 3' -5 '-cyclic adenosine monophosphate-regulated, PKA-dependent pathway. Consistent with this observation, changing the serine 171 residue that forms the optimal PKA phosphorylation site within the "activation loop" of HPK1 to alanine completely prevents this mutant from responding to PGE(2)-generated stimulation signals. Moreover, the inability of HPK1 to respond to PGE(2) stimulation in PKA-deficient S49 cells further supports the importance of PKA in this signaling pathway. We speculate that this unique signaling pathway enables PGE(2) signals to engage a proven negative regulator of TCR signal transduction pathway and uses it to inhibit T cell activation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/physiology , Gene Expression Regulation, Enzymologic , Protein Serine-Threonine Kinases/biosynthesis , Amino Acid Sequence , Catalysis , Dinoprostone/metabolism , Enzyme Activation , Humans , Immunosuppressive Agents/pharmacology , Jurkat Cells , Molecular Sequence Data , Phosphorylation , Proline/chemistry , Sequence Homology, Amino Acid , Signal Transduction
18.
Cancer Res ; 65(8): 3249-56, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15833857

ABSTRACT

The INK4 family of proteins negatively regulates cell cycle progression at the G(1)-S transition by inhibiting cyclin-dependent kinases. Two of these cell cycle inhibitors, p16(INK4A) and p15(INK4B), have tumor suppressor activities and are inactivated in human cancer. Interestingly, both INK4 genes express alternative splicing variants. In addition to p16(INK4A), the INK4A locus encodes a splice variant, termed p12--specifically expressed in human pancreas--and ARF, a protein encoded by an alternative reading frame that acts as a tumor suppressor through the p53 pathway. Similarly, the human INK4B locus encodes the p15(INK4B) tumor suppressor and one alternatively spliced form, termed as p10. We show here that p10, which arises from the use of an alternative splice donor site within intron 1, is conserved in the mouse genome and is widely expressed in mouse tissues. Similarly to mouse p15(INK4B), p10 expression is also induced by oncogenic insults and transforming growth factor-beta treatment and acts as a cell cycle inhibitor. Importantly, we show that mouse p10 is able to induce cell cycle arrest in a p53-dependent manner. We also show that mouse p10 is able to inhibit foci formation and anchorage-independent growth in wild-type mouse embryonic fibroblasts, and that these antitransforming properties of mouse p10 are also p53-dependent. These results indicate that the INK4B locus, similarly to INK4A-ARF, harbors two different splicing variants that can be involved in the regulation of both the p53 and retinoblastoma pathways, the two major molecular pathways in tumor suppression.


Subject(s)
Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , Tumor Suppressor Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle/genetics , Cell Cycle Proteins/biosynthesis , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p15 , Genes, ras/genetics , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Isoforms , Retinoblastoma Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...