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










Publication year range
1.
Oncotarget ; 7(37): 60332-60347, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27533087

ABSTRACT

Ephrin receptors (Ephs) are reported to control metastatic signaling of non-small cell lung cancer (NSCLC) and other tumors. Here we show for the first time that blocking expression of the Eph ligand Ephrin B3 inhibits NSCLC cell migration and invasion. We demonstrate that Ephrin B3 directly binds the EphAs EphA2, EphA3, EphA4, and EphA5. EphA2 Ser897 was previously shown to drive migration propensity of tumor cells and our study reveals that EphA2 stays phosphorylated on Ser897 in the Ephrin B3/EphA2 complex in NSCLC cells of different histology. Moreover, we report that within such Ephrin B3/EphA2 complex both Akt Ser 129 and p38MAPK are found indicating a potential to drive migration/proliferation. We also found the EMT marker E-cadherin expression to be maintained or increased upon Ephrin B3 blockade in NSCLC cells. Expression of Ephrin B3 was furthermore analyzed in a cohort of NSCLC stage IA-IB cases (n=200) alongside EphA2 and Ephrin A1. We found that Ephrin B3 was concomitantly expressed with EphA2 and Ephrin A1 with higher Ephrin B3 levels found in non-squamous than in squamous tumors, whereas EphA2 was higher expressed in well-differentiated than in low-differentiated tumors. In the entire NSCLC cohort, Ephrin B3 expression was not linked to patient survival, whereas a high EphA2 expression was associated with improved survival (p=0.03). In conclusion, we show that blocking Ephrin B3 expression inhibits NSCLC proliferation-, migration- and invasion capacity which calls for further studies on interference with Ephrin B3 as a possible therapeutic avenue in this tumor malignancy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Movement/genetics , Ephrin-B3/genetics , Lung Neoplasms/genetics , Receptors, Eph Family/genetics , A549 Cells , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Ephrin-A1/genetics , Ephrin-A1/metabolism , Ephrin-B3/metabolism , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Protein Binding , RNA Interference , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Receptors, Eph Family/metabolism
2.
Oncotarget ; 7(37): 59322-59335, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27528037

ABSTRACT

Ovarian cancer carries a significant mortality. Since symptoms tend to be minimal, the disease is often diagnosed when peritoneal metastases are already present. The standard of care in advanced ovarian cancer consists of platinum-based chemotherapy combined with cytoreductive surgery. Unfortunately, even after optimal cytoreduction and adjuvant chemotherapy, most patients with stage III disease will develop a recurrence. Intraperitoneal administration of chemotherapy is an alternative treatment for patients with localized disease. The pharmacological and physiochemical properties of melflufen, a peptidase potentiated alkylator, raised the hypothesis that this drug could be useful in ovarian cancer and particularily against peritoneal carcinomatosis. In this study the preclinical effects of melflufen were investigated in different ovarian cancer models. Melflufen was active against ovarian cancer cell lines, primary cultures of patient-derived ovarian cancer cells, and inhibited the growth of subcutaneous A2780 ovarian cancer xenografts alone and when combined with gemcitabine or liposomal doxorubicin when administered intravenously. In addition, an intra- and subperitoneal xenograft model showed activity of intraperitoneal administered melflufen for peritoneal carcinomatosis, with minimal side effects and modest systemic exposure. In conclusion, results from this study support further investigations of melflufen for the treatment of peritoneal carcinomatosis from ovarian cancer, both for intravenous and intraperitoneal administration.


Subject(s)
Melphalan/analogs & derivatives , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Phenylalanine/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cytoreduction Surgical Procedures , Disease-Free Survival , Drug Evaluation, Preclinical , Female , Humans , Hyperthermia, Induced , Injections, Intraperitoneal , Melphalan/therapeutic use , Mice , Mice, SCID , Neoplasm Recurrence, Local , Neoplasm Staging , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Phenylalanine/therapeutic use , Xenograft Model Antitumor Assays
3.
Mol Oncol ; 10(5): 719-34, 2016 05.
Article in English | MEDLINE | ID: mdl-26827254

ABSTRACT

BACKGROUND: Chemotherapy options in advanced urothelial carcinoma (UC) remain limited. Here we evaluated the peptide-based alkylating agent melphalan-flufenamide (mel-flufen) for UC. METHODS: UC cell lines J82, RT4, TCCsup and 5637 were treated with mel-flufen, alone or combined with cisplatin, gemcitabine, dasatinib or bestatin. Cell viability (MTT assay), intracellular drug accumulation (liquid chromatography) apoptosis induction (apoptotic cell nuclei morphology, western blot analysis of PARP-1/caspase-9 cleavage and Bak/Bax activation) were evaluated. Kinome alterations were characterized by PathScan array and phospho-Src validated by western blotting. Aminopeptidase N (ANPEP) expression was evaluated in UC clinical specimens in relation to patient outcome. RESULTS: In J82, RT4, TCCsup and 5637 UC cells, mel-flufen amplified the intracellular loading of melphalan in part via aminopeptidase N (ANPEP), resulting in increased cytotoxicity compared to melphalan alone. Mel-flufen induced apoptosis seen as activation of Bak/Bax, cleavage of caspase-9/PARP-1 and induction of apoptotic cell nuclei morphology. Combining mel-flufen with cisplatin or gemcitabine in J82 cells resulted in additive cytotoxic effects and for gemcitabine also increased apoptosis induction. Profiling of mel-flufen-induced kinome alterations in J82 cells revealed that mel-flufen alone did not inhibit Src phosphorylation. Accordingly, the Src inhibitor dasatinib sensitized for mel-flufen cytotoxicity. Immunohistochemical analysis of the putative mel-flufen biomarker ANPEP demonstrated prominent expression levels in tumours from 82 of 83 cystectomy patients. Significantly longer median overall survival was found in patients with high ANPEP expression (P = 0.02). CONCLUSION: Mel-flufen alone or in combination with cisplatin, gemcitabine or Src inhibition holds promise as a novel treatment for UC.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dasatinib/pharmacology , Melphalan/analogs & derivatives , Phenylalanine/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Urologic Neoplasms/drug therapy , src-Family Kinases/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Humans , Melphalan/pharmacology , Phenylalanine/pharmacology , Urologic Neoplasms/pathology , Urothelium/drug effects , Urothelium/pathology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
4.
PLoS One ; 9(11): e112242, 2014.
Article in English | MEDLINE | ID: mdl-25393309

ABSTRACT

The infusion of ex vivo-expanded autologous T regulatory (Treg) cells is potentially an effective immunotherapeutic strategy against graft-versus-host disease (GvHD) and several autoimmune diseases, such as type 1 diabetes (T1D). However, in vitro differentiation of antigen-specific T cells into functional and stable Treg (iTreg) cells has proved challenging. As insulin is the major autoantigen leading to T1D, we tested the capacity of insulin-specific T-cell receptor (TCR) transgenic CD4(+) T cells of the BDC12-4.1 clone to convert into Foxp3(+) iTreg cells. We found that in vitro polarization toward Foxp3(+) iTreg was effective with a majority (>70%) of expanded cells expressing Foxp3. However, adoptive transfer of Foxp3(+) BDC12-4.1 cells did not prevent diabetes onset in immunocompetent NOD mice. Thus, in vitro polarization of insulin-specific BDC12-4.1 TCR transgenic CD4(+) T cells toward Foxp3+ cells did not provide dominant tolerance in recipient mice. These results highlight the disconnect between an in vitro acquired Foxp3(+) cell phenotype and its associated in vivo regulatory potential.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/metabolism , Insulin/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Diabetes Mellitus, Type 1/prevention & control , Forkhead Transcription Factors/genetics , Gene Expression , Mice , Mice, Inbred NOD , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics
5.
Int J Radiat Biol ; 90(8): 718-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24646078

ABSTRACT

PURPOSE: We aimed to analyze the activation status of commonly deregulated receptor tyrosine kinases (RTK) in human non-small cell lung cancer (NSCLC) tumor initiating cells (TIC) previously demonstrated to be refractory to ionizing radiation and chemotherapy. MATERIALS AND METHODS: Phosphorylated RTK and important signaling nodes were assayed using PathScan RTK Signaling Antibody Array Kit in NSCLC TIC and bulk cells 4 h post-irradiation (IR) and validated by Western blot. The effect of mitogen- activated protein kinase kinase (MEK) inhibition combined with IR was analyzed using clonogenic assay. RESULTS: H125 TIC displayed decreased basal phosphorylation of insulin-like growth factor 1 receptor (IGF-1R) and signal transducer and activator of transcription 1 (STAT1) (Tyr701) as compared to bulk cells. Total IGF-1R levels were significantly lower in NSCLC TIC as compared to bulk cells. A higher degree of extracellular signal-regulated kinase (ERK) phosphorylation was evident in TIC and concordantly MEK inhibition reduced TIC viability. Moreover, MEK inhibition also decreased clonogenicity upon IR suggesting that MEK and downstream signaling impart on TIC radiation response. CONCLUSIONS: We demonstrate reduced basal phosphorylation of several signaling pathways including lower total IGF-1R levels in NSCLC TIC which is of potential concern for RTK inhibitor use. Importantly, MEK inhibition decreased cell viability of NSCLC TIC alone or in combination with IR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Molecular Targeted Therapy , Neoplastic Stem Cells/pathology , Phosphoproteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Mitogen-Activated Protein Kinases/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Phenotype , Phosphoproteins/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , STAT1 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Diabetes ; 61(5): 1169-79, 2012 May.
Article in English | MEDLINE | ID: mdl-22403296

ABSTRACT

In diabetic patients and susceptible mice, insulin is a targeted autoantigen. Insulin B chain 9-23 (B:9-23) autoreactive CD4 T cells are key for initiating autoimmune diabetes in NOD mice; however, little is known regarding their origin and function. To this end, B:9-23-specific, BDC12-4.1 T-cell receptor (TCR) transgenic (Tg) mice were studied, of which, despite expressing a single TCR on the recombination activating gene-deficient background, only a fraction develops diabetes in an asynchronous manner. BDC12-4.1 CD4 T cells convert into effector (Teff) and Foxp3(+)-expressing adaptive regulatory T cells (aTregs) soon after leaving the thymus as a result of antigen recognition and homeostatic proliferation. The generation of aTreg causes the heterogeneous diabetes onset, since crossing onto the scurfy (Foxp3) mutation, BDC12-4.1 TCR Tg mice develop accelerated and fully penetrant diabetes. Similarly, adoptive transfer and bone marrow transplantation experiments showed differential diabetes kinetics based on Foxp3(+) aTreg's presence in the BDC12-4.1 donors. A single-specificity, insulin-reactive TCR escapes thymic deletion and simultaneously converts into aTreg and Teff, establishing an equilibrium that determines diabetes penetrance. These results are of particular importance for understanding disease pathogenesis. They suggest that once central tolerance is bypassed, autoreactive cells arriving in the periphery do not by default follow solely a pathogenic fate upon activation.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/physiology , Cell Differentiation/physiology , Diabetes Mellitus/prevention & control , Insulin/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Major Histocompatibility Complex , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Mutation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/physiology
7.
J Immunol ; 187(6): 2915-22, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21832162

ABSTRACT

Recent studies have shown that IL-17 can contribute beneficially to pathogen defense but also that excessive IL-17 levels are associated with chronic inflammation and autoimmune disorders. To date, the role of IL-17 in viral infections and type 1 diabetes is ambiguous. In this study, we used IL-17A enhanced green fluorescent protein bicistronic reporter mouse strains to analyze in situ production of IL-17A. Upon Klebsiella pneumoniae bacterial infection, CD4(+) and γδ T cells produce IL-17A. In contrast, CD4(+) or CD8(+) T cells do not produce IL-17A in response to acute or protracted viral infection with lymphocytic choriomeningitis virus or during autoimmune diabetes development in the CD8-driven lymphocytic choriomeningitis virus-induced model of type 1 diabetes. We conclude that viral elimination and type 1 diabetes can occur in the absence of detectable IL-17A production, suggesting IL-17A is not essential in these settings.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Interleukin-17/immunology , Virus Diseases/immunology , Animals , CD4-Positive T-Lymphocytes , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Gene Knock-In Techniques , Genes, Reporter , Green Fluorescent Proteins/genetics , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL
8.
PLoS One ; 6(1): e14502, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21253594

ABSTRACT

BACKGROUND: Memory CD8 T cells form an essential part of protective immunity against viral infections. Antigenic load, costimulation, CD4-help, cytokines and chemokines fluctuate during the course of an antiviral immune response thus affecting CD8 T cell activation and memory conversion. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, naïve TCR transgenic LCMV-specific P14 CD8 T cells engaged at a late stage during the acute antiviral LCMV response showed reduced expansion kinetics but greater memory conversion in the spleen. Such late activated cells displayed a memory precursor effector phenotype already at the peak of the systemic antiviral response, suggesting that the environment determined their fate during antigen encounter. In the spleen, the majority of late transferred cells exhibited a central memory phenotype compared to the effector memory displayed by the early transferred cells. Increasing the inflammatory response by exogenous administration of IFNγ, PolyI:C or CpG did not affect memory conversion in the late transferred group, suggesting that the diverging antigen load early versus later during acute infection had determined their fate. In agreement, reduction in the LCMV antigenic load after ribavirin treatment enhanced the contribution of early transferred cells to the long lasting memory pool. CONCLUSIONS/SIGNIFICANCE: Our results show that naïve CD8 cells, exposed to reduced duration or concentration of antigen during viral infection convert into memory more efficiently, an observation that could have significant implications for vaccine design.


Subject(s)
Antigens, Viral/analysis , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Virus Diseases/immunology , Animals , Antigens, Viral/immunology , Cells, Cultured , Immunity , Inflammation , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , T-Cell Antigen Receptor Specificity/immunology , Time Factors
9.
Viral Immunol ; 23(3): 285-94, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20565292

ABSTRACT

Impaired antiviral CD8 and CD4 T-cell responses are often associated with chronic viral infections. Cell-intrinsic as well as cell-extrinsic mechanisms are thought to dampen such responses, for example programmed death 1 receptor (PD-1) expression on T cells, and interleukin (IL)-10 production primarily by dendritic cells (DCs), have been shown to support viral persistence by suppressing immune responses. Here we demonstrate that CD103, an alpha E integrin necessary for T-cell homing and retention in the gut and other epithelia expressed by the majority of naïve CD8(+), and CD4(+)CD25(+) T cells and some DC subsets, is unnecessary for controlling T-cell responses during chronic lymphocytic choriomeningitis virus clone 13 (LCMV cl13) infection. T-cell analysis following viral infection showed that the primary as well as the memory CD8(+) and CD4(+) T-cell responses among CD103-sufficient and CD103-deficient mice were identical. In addition, no rescue of cytokine production by virus-specific T cells or alterations in viral titers in the absence of intrinsic CD103 expression was observed. Interestingly, CD103 levels on the effector CD8(+) T cells became reduced soon after virus infection, with a small proportion of cells co-expressing PD-1 and CD103. In contrast, although no substantial differences in the frequency and number of the CD4(+)CD25(+) cell population were seen, CD103 expression increased significantly over time in this population, correlating with viral persistence. Thus, a lack of CD103 expression does not affect functional impairment of effector T-cell responses during chronic viral infection.


Subject(s)
Antigens, CD/immunology , Integrin alpha Chains/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunologic Memory , Integrin alpha Chains/deficiency , Mice , Mice, Knockout
10.
J Autoimmun ; 34(4): 445-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20004555

ABSTRACT

Redirection of immune responses by manipulation of antigen-presenting cells is an emerging strategy for immunosuppressive treatment of autoimmune diseases. In vivo expansion of dendritic cells (DC) by Fms-like tyrosine kinase-3 (Flt3)-Ligand (FL) treatment was shown to delay diabetes onset in the NOD model of autoimmune diabetes. However, we show here that Flt3 stimulation actually accelerates autoimmunity when autoreactive CD8 T cells are detectable in blood prior to treatment. With autoreactive CD8 cells present, the capacity of FL to expand DCs and induce Treg remained intact, but both numbers and the functional response of islet-specific CD8s were boosted. Also, the inhibitory receptor PD-1 on (autoreactive) CD8 T cells and its ligand PD-L1 on Treg were no longer upregulated. These data highlight the need to pre-screen for T cell autoreactivity prior to generalized DC expansion and illustrate how accelerated disease can occur when the intended initiation of regulatory mechanisms is impaired later in diabetogenesis.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/drug therapy , Membrane Proteins/therapeutic use , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Dendritic Cells/cytology , Humans , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/cytology , Treatment Outcome
11.
J Autoimmun ; 32(1): 70-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19162441

ABSTRACT

Recent studies suggest a beneficial role for blocking CD103 signaling in preventing islet allograft rejection and thus Type 1 diabetes (T1D) in non-obese diabetic (NOD) mice. However, antibody blockade approaches generally raise anti-microbial safety issues, necessitating additional studies to address the possible adverse effects of antibody therapy. Here we report that CD103 had no significant impact on the development of primary and memory CD8(+) or CD4(+) responses after acute lymphocytic choriomeningitis virus (LCMV) infection. In addition, CD103 was found to be dispensable for T1D progression in a rapid, CD8-mediated virally-induced T1D model (the rat insulin promoter [RIP]-LCMV), suggesting that its previous efficacy in the NOD mouse model may not be related to its effect on the generation, memory conversion and/or effector function of CD8(+) or CD4(+) T cells. While the data does not preclude a role for CD103 in T1D in its entirety, the current study does provide much evidence to suggest that CD103 blockade may prove to be a safe intervention for autoimmunity and allo-transplantation. While in cases of rapid microbial (CD8)-driven T1D CD103 antibody blockade may not limit disease progression or severity, in mucosally-driven cases of T1D anti-CD103 antibody treatment may provide a new and safe therapeutic avenue.


Subject(s)
Antigens, CD/physiology , Diabetes Mellitus, Type 1/immunology , Integrin alpha Chains/physiology , Virus Diseases/immunology , Animals , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/etiology , Epitopes, T-Lymphocyte/immunology , Female , Insulin/genetics , Insulin/metabolism , Interferon-gamma/metabolism , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pancreas/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Virus Diseases/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...