Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Diagnostics (Basel) ; 11(2)2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33671952

ABSTRACT

Cytomegalovirus (CMV) infection is a major cause of morbidity and mortality following hematopoietic stem cell transplantation (HSCT). Measuring CMV-specific cellular immunity may improve the risk stratification and management of patients. IFN-γ ELISpot assays, based on the stimulation of peripheral blood mononuclear cells with CMV pp65 and IE-1 proteins or peptides, have been validated in clinical settings. However, it remains unclear to which extend the T-cell response to synthetic peptides reflect that mediated by full-length proteins processed by antigen-presenting cells. We compared the stimulating ability of pp65 and IE-1 proteins and corresponding overlapping peptides in 16 HSCT recipients using a standardized IFN-γ ELISpot assay. Paired qualitative test results showed an overall 74.4% concordance. Discordant results were mainly due to low-response tests, with one exception. One patient with early CMV reactivation and graft-versus-host disease, sustained CMV DNAemia and high CD8+ counts showed successive negative protein-based ELISpot results but a high and sustained response to IE-1 peptides. Our results suggest that the response to exogenous proteins, which involves their uptake and processing by antigen-presenting cells, more closely reflects the physiological response to CMV infection, while the response to exogenous peptides may lead to artificial in vitro T-cell responses, especially in strongly immunosuppressed patients.

2.
Haematologica ; 106(2): 363-374, 2021 02 01.
Article in English | MEDLINE | ID: mdl-31879324

ABSTRACT

Recurrence of cytomegalovirus reactivation remains a major cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation. Monitoring cytomegalovirus-specific cellular immunity using a standardized assay might improve the risk stratification of patients. A prospective multicenter study was conducted in 175 intermediate- and high-risk allogeneic hematopoietic stem cell transplant recipients under preemptive antiviral therapy. Cytomegalovirus-specific cellular immunity was measured using a standardized IFN-γ ELISpot assay (T-Track® CMV). Primary aim was to evaluate the suitability of measuring cytomegalovirus-specific immunity after end of treatment for a first cytomegalovirus reactivation to predict recurrent reactivation. 40/101 (39.6%) patients with a first cytomegalovirus reactivation experienced recurrent reactivations, mainly in the high-risk group (cytomegalovirus-seronegative donor/cytomegalovirus-seropositive recipient). The positive predictive value of T-Track® CMV (patients with a negative test after the first reactivation experienced at least one recurrent reactivation) was 84.2% in high-risk patients. Kaplan-Meier analysis revealed a higher probability of recurrent cytomegalovirus reactivation in high-risk patients with a negative test after the first reactivation (hazard ratio 2.73; p=0.007). Interestingly, a post-hoc analysis considering T-Track® CMV measurements at day 100 post-transplantation, a time point highly relevant for outpatient care, showed a positive predictive value of 90.0% in high-risk patients. Our results indicate that standardized cytomegalovirus-specific cellular immunity monitoring may allow improved risk stratification and management of recurrent cytomegalovirus reactivation after hematopoietic stem cell transplantation. This study was registered at www.clinicaltrials.gov as #NCT02156479.


Subject(s)
Cytomegalovirus Infections , Hematopoietic Stem Cell Transplantation , Cytomegalovirus , Cytomegalovirus Infections/diagnosis , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Prospective Studies , Risk Assessment , Virus Activation
3.
Front Immunol ; 9: 263, 2018.
Article in English | MEDLINE | ID: mdl-29535708

ABSTRACT

Resistant mouse strains mount a protective T cell-mediated immune response upon infection with Leishmania (L.) parasites. Healing correlates with a T helper (Th) cell-type 1 response characterized by a pronounced IFN-γ production, while susceptibility is associated with an IL-4-dependent Th2-type response. It has been shown that dermal dendritic cells are crucial for inducing protective Th1-mediated immunity. Additionally, there is growing evidence that C-type lectin receptor (CLR)-mediated signaling is involved in directing adaptive immunity against pathogens. However, little is known about the function of the CLR Dectin-1 in modulating Th1- or Th2-type immune responses by DC subsets in leishmaniasis. We characterized the expression of Dectin-1 on CD11c+ DCs in peripheral blood, at the site of infection, and skin-draining lymph nodes of L. major-infected C57BL/6 and BALB/c mice and in peripheral blood of patients suffering from cutaneous leishmaniasis (CL). Both mouse strains responded with an expansion of Dectin-1+ DCs within the analyzed tissues. In accordance with the experimental model, Dectin-1+ DCs expanded as well in the peripheral blood of CL patients. To study the role of Dectin-1+ DCs in adaptive immunity against L. major, we analyzed the T cell stimulating potential of bone marrow-derived dendritic cells (BMDCs) in the presence of the Dectin-1 agonist Curdlan. These experiments revealed that Curdlan induces the maturation of BMDCs and the expansion of Leishmania-specific CD4+ T cells. Based on these findings, we evaluated the impact of Curdlan/Dectin-1 interactions in experimental leishmaniasis and were able to demonstrate that the presence of Curdlan at the site of infection modulates the course of disease in BALB/c mice: wild-type BALB/c mice treated intradermally with Curdlan developed a protective immune response against L. major whereas Dectin-1-/- BALB/c mice still developed the fatal course of disease after Curdlan treatment. Furthermore, the vaccination of BALB/c mice with a combination of soluble L. major antigens and Curdlan was able to provide a partial protection from severe leishmaniasis. These findings indicate that the ligation of Dectin-1 on DCs acts as an important checkpoint in adaptive immunity against L. major and should therefore be considered in future whole-organism vaccination strategies.


Subject(s)
Dendritic Cells/immunology , Lectins, C-Type/immunology , Leishmaniasis Vaccines/immunology , Leishmaniasis, Cutaneous/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Humans , Leishmania major/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
4.
Transpl Int ; 31(4): 436-450, 2018 04.
Article in English | MEDLINE | ID: mdl-29284181

ABSTRACT

Impaired cytomegalovirus (CMV)-specific cell-mediated immunity (CMV-CMI) is a major cause of CMV reactivation and associated complications in solid-organ transplantation. Reliably assessing CMV-CMI is desirable to individually adjust antiviral and immunosuppressive therapy. This study aimed to evaluate the suitability of T-Track® CMV, a novel IFN-γ ELISpot assay based on the stimulation of peripheral blood mononuclear cells with pp65 and IE-I CMV proteins, to monitor CMV-CMI following kidney transplantation. A prospective longitudinal multicenter study was conducted in 86 intermediate-risk renal transplant recipients. CMV-CMI, CMV viral load, and clinical complications were monitored over 6 months post-transplantation. Ninety-five percent and 88-92% ELISpot assays were positive pre- and post-transplantation, respectively. CMV-specific response was reduced following immunosuppressive treatment and increased in patients with graft rejection, indicating the ability of the ELISpot assay to monitor patients' immunosuppressive state. Interestingly, median pp65-specific response was ninefold higher in patients with self-clearing viral load compared to antivirally treated patients prior to first viral load detection (P < 0.001), suggesting that reactivity to pp65 represents a potential immunocompetence marker. Altogether, T-Track® CMV is a highly sensitive IFN-γ ELISpot assay, suitable for the immunomonitoring of CMV-seropositive renal transplant recipients, and with a potential use for the risk assessment of CMV-related clinical complications (ClinicalTrials.gov Identifier: NCT02083042).


Subject(s)
Cytomegalovirus Infections/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Immunity, Cellular , Phosphoproteins/immunology , Postoperative Complications/diagnosis , Viral Matrix Proteins/immunology , Adult , Aged , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Female , Graft Rejection/immunology , Graft Rejection/virology , Humans , Immunosuppressive Agents , Kidney Transplantation , Longitudinal Studies , Male , Middle Aged , Opportunistic Infections , Postoperative Complications/immunology , Prospective Studies , Young Adult
5.
BMC Immunol ; 18(1): 15, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28270092

ABSTRACT

BACKGROUND: Uncontrolled cytomegalovirus (CMV) replication in immunocompromised solid-organ transplant recipients is a clinically relevant issue and an indication of impaired CMV-specific cell-mediated immunity (CMI). Primary aim of this study was to assess the suitability of the immune monitoring tool T-Track® CMV to determine CMV-reactive CMI in a cohort of hemodialysis patients representative of patients eligible for renal transplantation. Positive and negative agreement of T-Track® CMV with CMV serology was examined in 124 hemodialysis patients, of whom 67 (54%) revealed a positive CMV serostatus. Secondary aim of the study was to evaluate T-Track® CMV performance against two unrelated CMV-specific CMI monitoring assays, QuantiFERON®-CMV and a cocktail of six class I iTAg™ MHC Tetramers. RESULTS: Positive T-Track® CMV results were obtained in 90% (60/67) of CMV-seropositive hemodialysis patients. In comparison, 73% (45/62) and 77% (40/52) positive agreement with CMV serology was achieved using QuantiFERON®-CMV and iTAg™ MHC Tetramer. Positive T-Track® CMV responses in CMV-seropositive patients were dominated by pp65-reactive cells (58/67 [87%]), while IE-1-responsive cells contributed to an improved (87% to 90%) positive agreement of T-Track® CMV with CMV serology. Interestingly, T-Track® CMV, QuantiFERON®-CMV and iTAg™ MHC Tetramers showed 79% (45/57), 87% (48/55) and 93% (42/45) negative agreement with serology, respectively, and a strong inter-assay variability. Notably, T-Track® CMV was able to detect IE-1-reactive cells in blood samples of patients with a negative CMV serology, suggesting either a previous exposure to CMV that yielded a cellular but no humoral immune response, or TCR cross-reactivity with foreign antigens, both suggesting a possible protective immunity against CMV in these patients. CONCLUSION: T-Track® CMV is a highly sensitive assay, enabling the functional assessment of CMV-responsive cells in hemodialysis patients prior to renal transplantation. T-Track® CMV thus represents a valuable immune monitoring tool to identify candidate transplant recipients potentially at increased risk for CMV-related clinical complications.


Subject(s)
Cytomegalovirus Infections/diagnosis , Cytomegalovirus/immunology , Immunocompromised Host , Kidney Failure, Chronic/diagnosis , Renal Dialysis , Adult , Aged , Aged, 80 and over , Antibodies, Viral/blood , Cells, Cultured , Cohort Studies , Cytomegalovirus Infections/immunology , Female , Humans , Immediate-Early Proteins/immunology , Immunity, Cellular , Immunoassay , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/therapy , Kidney Transplantation , Male , Middle Aged , Monitoring, Immunologic/methods , Observer Variation , Phosphoproteins/immunology , Sensitivity and Specificity , Viral Matrix Proteins/immunology , Waiting Lists
6.
BMC Immunol ; 18(1): 14, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28270111

ABSTRACT

BACKGROUND: In healthy individuals, Cytomegalovirus (CMV) infection is efficiently controlled by CMV-specific cell-mediated immunity (CMI). Functional impairment of CMI in immunocompromized individuals however can lead to uncontrolled CMV replication and severe clinical complications. Close monitoring of CMV-specific CMI is therefore clinically relevant and might allow a reliable prognosis of CMV disease as well as assist personalized therapeutic decisions. METHODS: Objective of this work was the optimization and technical validation of an IFN-γ ELISpot assay for a standardized, sensitive and reliable quantification of CMV-reactive effector cells. T-activated® immunodominant CMV IE-1 and pp65 proteins were used as stimulants. All basic assay parameters and reagents were tested and optimized to establish a user-friendly protocol and maximize the signal-to-noise ratio of the ELISpot assay. RESULTS: Optimized and standardized ELISpot revealed low intra-assay, inter-assay and inter-operator variability (coefficient of variation CV below 22%) and CV inter-site was lower than 40%. Good assay linearity was obtained between 6 × 104 and 2 × 105 PBMC per well upon stimulation with T-activated® IE-1 (R2 = 0.97) and pp65 (R2 = 0.99) antigens. Remarkably, stimulation of peripheral blood mononuclear cells (PBMC) with T-activated® IE-1 and pp65 proteins resulted in the activation of a broad range of CMV-reactive effector cells, including CD3+CD4+ (Th), CD3+CD8+ (CTL), CD3-CD56+ (NK) and CD3+CD56+ (NKT-like) cells. Accordingly, the optimized IFN-γ ELISpot assay revealed very high sensitivity (97%) in a cohort of 45 healthy donors, of which 32 were CMV IgG-seropositive. CONCLUSION: The combined use of T-activated® IE-1 and pp65 proteins for the stimulation of PBMC with the optimized IFN-γ ELISpot assay represents a highly standardized, valuable tool to monitor the functionality of CMV-specific CMI with great sensitivity and reliability.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/diagnosis , Cytomegalovirus/physiology , Enzyme-Linked Immunospot Assay/methods , Killer Cells, Natural/immunology , Natural Killer T-Cells/immunology , Adult , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Cytomegalovirus Infections/immunology , Cytotoxicity, Immunologic , Female , Humans , Immediate-Early Proteins/immunology , Immunity, Cellular , Interferon-gamma/metabolism , Killer Cells, Natural/virology , Male , Middle Aged , Monitoring, Immunologic , Natural Killer T-Cells/virology , Observer Variation , Phosphoproteins/immunology , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Viral Matrix Proteins/immunology , Young Adult
7.
J Med Virol ; 89(2): 324-331, 2017 02.
Article in English | MEDLINE | ID: mdl-27447923

ABSTRACT

Cytomegalovirus (CMV) is the most common congenital viral infection. Mother-to-child transmission can cause severe child disability. Intact CMV-specific cell-mediated immunity (CMI) was shown to prevent uncontrolled replication in healthy individuals. This study aimed to determine whether CMV-specific CMI is impaired in pregnant women, thus potentially increasing the overall risk for active CMV replication and transmission. CMV-specific CMI in peripheral blood of 60 pregnant women was determined using T-Track® CMV for detection of IE-1 and pp65-reactive effector cells by IFN-γ ELISpot, and compared to the CMV-IgG and -IgM serostatus. CMV-specific CMI was detected in 65% of CMV-seropositive pregnant women. Five percent of CMV-IgG seronegative women showed IE-1- but not pp65-reactive cells. The overall number of CMV-reactive cells in pregnant women was significantly lower compared to a matched non-pregnant control group (P < 0.001). No significant difference in CMV-specific CMI was detected in the course of the three trimesters of pregnancy of CMV-IgG seropositive women. Postpartum (median days postnatal = 123), IE-1- and pp65-specific CMI remained significantly lower than in the non-pregnant control group (P < 0.001 and 0.0032, respectively). Functional analysis of CMV-reactive immune cells using T-Track® CMV therefore suggests a systemic down-regulation of CMV-specific CMI in pregnant women. Further studies are needed to investigate whether this may be indicative of a higher susceptibility to CMV reactivation or transmission. J. Med. Virol. 89:324-331, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cytomegalovirus/immunology , Immune Tolerance , Immunity, Cellular , Adolescent , Adult , Antibodies, Viral/blood , Antigens, Viral/immunology , Enzyme-Linked Immunospot Assay , Female , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Middle Aged , Pregnancy , Young Adult
8.
Methods Mol Biol ; 1510: 257-276, 2017.
Article in English | MEDLINE | ID: mdl-27761827

ABSTRACT

Transcriptional activation by STAT5 is repressed by deacetylase inhibitors. Investigating the role of deacetylases (HDACs) in STAT5-mediated transcription implies the analysis of molecular events taking place at the chromatin level. We describe here two alternative methods of chromatin immunoprecipitation that allow the characterization of chromatin modifications ensuing STAT5 activation and its inhibition by deacetylase inhibitors, in particular changes in histone acetylation, in histone occupancy, and in the association/dissociation of transcription factors and other chromatin-associated factors.


Subject(s)
B-Lymphocytes/immunology , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , STAT5 Transcription Factor/genetics , Acetylation , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Line, Tumor , Chromatin/chemistry , Chromatin/immunology , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/immunology , Histone Deacetylases/immunology , Histones/genetics , Histones/immunology , Interleukin-3/pharmacology , Lymphocyte Activation , Mice , STAT5 Transcription Factor/immunology , Transcription Factors , Transcription, Genetic
9.
PLoS One ; 11(6): e0157430, 2016.
Article in English | MEDLINE | ID: mdl-27304884

ABSTRACT

Sulforaphane (SFN) and moringin (GMG-ITC) are edible isothiocyanates present as glucosinolate precursors in cruciferous vegetables and in the plant Moringa oleifera respectively, and recognized for their chemopreventive and medicinal properties. In contrast to the well-studied SFN, little is known about the molecular pathways targeted by GMG-ITC. We investigated the ability of GMG-ITC to inhibit essential signaling pathways that are frequently upregulated in cancer and immune disorders, such as JAK/STAT and NF-κB. We report for the first time that, similarly to SFN, GMG-ITC in the nanomolar range suppresses IL-3-induced expression of STAT5 target genes. GMG-ITC, like SFN, does not inhibit STAT5 phosphorylation, suggesting a downstream inhibitory event. Interestingly, treatment with GMG-ITC or SFN had a limited inhibitory effect on IFNα-induced STAT1 and STAT2 activity, indicating that both isothiocyanates differentially target JAK/STAT signaling pathways. Furthermore, we showed that GMG-ITC in the micromolar range is a more potent inhibitor of TNF-induced NF-κB activity than SFN. Finally, using a cellular system mimicking constitutive active STAT5-induced cell transformation, we demonstrated that SFN can reverse the survival and growth advantage mediated by oncogenic STAT5 and triggers cell death, therefore providing experimental evidence of a cancer chemopreventive activity of SFN. This work thus identified STAT5, and to a lesser extent STAT1/STAT2, as novel targets of moringin. It also contributes to a better understanding of the biological activities of the dietary isothiocyanates GMG-ITC and SFN and further supports their apparent beneficial role in the prevention of chronic illnesses such as cancer, inflammatory diseases and immune disorders.


Subject(s)
Isothiocyanates/pharmacology , Janus Kinases/metabolism , Moringa oleifera/chemistry , STAT Transcription Factors/metabolism , Seeds/chemistry , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Dose-Response Relationship, Drug , Gene Expression/drug effects , HeLa Cells , Humans , Interferon-alpha/pharmacology , Interleukin-3/pharmacology , Isothiocyanates/chemistry , Isothiocyanates/isolation & purification , Janus Kinases/genetics , Mice , Molecular Structure , Phosphorylation/drug effects , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics
10.
Biol Chem ; 397(11): 1187-1204, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27341558

ABSTRACT

The JAK/STAT pathway is an essential mediator of cytokine signaling, often upregulated in human diseases and therefore recognized as a relevant therapeutic target. We previously identified the synthetic chalcone α-bromo-2',3,4,4'-tetramethoxychalcone (α-Br-TMC) as a novel JAK2/STAT5 inhibitor. We also found that treatment with α-Br-TMC resulted in a downward shift of STAT5 proteins in SDS-PAGE, suggesting a post-translational modification that might affect STAT5 function. In the present study, we show that a single cysteine within STAT5 is responsible for the α-Br-TMC-induced protein shift, and that this modification does not alter STAT5 transcriptional activity. We also compared the inhibitory activity of α-Br-TMC to that of another synthetic chalcone, α-trifluoromethyl-2',3,4,4'-tetramethoxychalcone (α-CF3-TMC). We found that, like α-Br-TMC, α-CF3-TMC inhibits JAK2 and STAT5 phosphorylation in response to interleukin-3, however without altering STAT5 mobility in SDS-PAGE. Moreover, we demonstrate that both α-Br-TMC and α-CF3-TMC inhibit interferon-α-induced activation of STAT1 and STAT2, by inhibiting their phosphorylation and the expression of downstream interferon-stimulated genes. Together with the previous finding that α-Br-TMC and α-CF3-TMC inhibit the response to inflammation by inducing Nrf2 and blocking NF-κB activities, our data suggest that synthetic chalcones might be useful as anti-inflammatory, anti-cancer and immunomodulatory agents in the treatment of human diseases.


Subject(s)
Chalcones/pharmacology , Interferon-alpha/antagonists & inhibitors , Interleukin-3/antagonists & inhibitors , Janus Kinase 2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Phosphorylation/drug effects , STAT5 Transcription Factor/chemistry
11.
BMC Mol Biol ; 17: 10, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27074708

ABSTRACT

BACKGROUND: c-Myc has been proposed as a putative target gene of signal transducer and activator of transcription 5 (STAT5). No functional STAT5 binding site has been identified so far within the c-Myc gene locus, therefore a direct transcriptional regulation by STAT5 remains uncertain. c-Myc super-enhancer, located 1.7 Mb downstream of the c-Myc gene locus, was recently reported as essential for the regulation of c-Myc gene expression by hematopoietic transcription factors and bromodomain and extra-terminal (BET) proteins and for leukemia maintenance. c-Myc super-enhancer is composed of five regulatory regions (E1-E5) which recruit transcription and chromatin-associated factors, mediating chromatin looping and interaction with the c-Myc promoter. RESULTS: We now show that STAT5 strongly binds to c-Myc super-enhancer regions E3 and E4, both in normal and transformed Ba/F3 cells. We also found that the BET protein bromodomain-containing protein 2 (BRD2), a co-factor of STAT5, co-localizes with STAT5 at E3/E4 in Ba/F3 cells transformed by the constitutively active STAT5-1*6 mutant, but not in non-transformed Ba/F3 cells. BRD2 binding at E3/E4 coincides with c-Myc transcriptional activation and is lost upon treatment with deacetylase and BET inhibitors, both of which inhibit STAT5 transcriptional activity and c-Myc gene expression. CONCLUSIONS: Our data suggest that constitutive STAT5 binding to c-Myc super-enhancer might contribute to BRD2 maintenance and thus allow sustained expression of c-Myc in Ba/F3 cells transformed by STAT5-1*6.


Subject(s)
Genes, myc , Promoter Regions, Genetic , STAT5 Transcription Factor/metabolism , Animals , Base Sequence , Cell Line , Cell Line, Transformed , Gene Expression Regulation , Mice , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Receptors, Cell Surface/metabolism , Sequence Alignment , Transcription Factors
12.
Fitoterapia ; 106: 122-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26304764

ABSTRACT

The proazulene matricine (1) is present in chamomile flower heads and has been proven to exhibit strong in vivo anti-inflammatory activity. In contrast to other secondary metabolites in chamomile preparations like its degradation product chamazulene (2), no plausible targets have been found to explain this activity. Therefore we revisited 1 regarding its in vitro anti-inflammatory activity in cellular and molecular studies. Using ICAM-1 as a marker for NF-κB activation, it was shown that ICAM-1 protein expression induced by TNF-α and LPS, but not by IFN-γ, was remarkably inhibited by 1 in endothelial cells (HMEC-1). Inhibition was concentration-dependent in a micromolar range (10-75 µM) and did not involve cytotoxic effects. At 75 µM expression of the adhesion molecule ICAM-1 was down to 52.7 ± 3.3% and 20.4 ± 1.8% of control in TNF-α and LPS-stimulated HMEC-1, respectively. In contrast, 2 showed no activity. Quantitative RT-PCR experiments revealed that TNF-α-induced expression of the ICAM-1 gene was also reduced by 1 in a concentration-dependent manner, reaching 32.3 ± 6.2% of control at 100 µM matricine. Additional functional assays (NF-κB promotor activity and cytoplasm to nucleus translocation) confirmed the inhibitory effect of 1 on NF-κB signaling. Despite the fact that 1 lacks an α,ß-unsaturated carbonyl and is thus not able to act via a Michael reaction with electron rich SH groups of functional biological molecules, data gave strong evidence that 1 inhibits NF-κB transcriptional activity in endothelial cells by an hitherto unknown mechanism and this may contribute to its well-known anti-inflammatory activity in vivo.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Azulenes/pharmacology , Endothelial Cells/drug effects , Lactones/pharmacology , Sesquiterpenes/pharmacology , Cells, Cultured , Chamomile/chemistry , Flowers/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Sesquiterpenes, Guaiane , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
13.
Nucleic Acids Res ; 43(7): 3524-45, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25769527

ABSTRACT

Signal transducer and activator of transcription STAT5 is essential for the regulation of proliferation and survival genes. Its activity is tightly regulated through cytokine signaling and is often upregulated in cancer. We showed previously that the deacetylase inhibitor trichostatin A (TSA) inhibits STAT5-mediated transcription by preventing recruitment of the transcriptional machinery at a step following STAT5 binding to DNA. The mechanism and factors involved in this inhibition remain unknown. We now show that deacetylase inhibitors do not target STAT5 acetylation, as we initially hypothesized. Instead, they induce a rapid increase in global histone acetylation apparently resulting in the delocalization of the bromodomain and extra-terminal (BET) protein Brd2 and of the Brd2-associated factor TBP to hyperacetylated chromatin. Treatment with the BET inhibitor (+)-JQ1 inhibited expression of STAT5 target genes, supporting a role of BET proteins in the regulation of STAT5 activity. Accordingly, chromatin immunoprecipitation demonstrated that Brd2 is associated with the transcriptionally active STAT5 target gene Cis and is displaced upon TSA treatment. Our data therefore indicate that Brd2 is required for the proper recruitment of the transcriptional machinery at STAT5 target genes and that deacetylase inhibitors suppress STAT5-mediated transcription by interfering with Brd2 function.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , STAT5 Transcription Factor/physiology , Transcription, Genetic/drug effects , Acetylation , Amino Acid Sequence , Animals , Cell Line , Chromatin Immunoprecipitation , Electroporation , Histones/metabolism , Mice , Molecular Sequence Data , Polymerase Chain Reaction , STAT5 Transcription Factor/chemistry , Sequence Homology, Amino Acid , Transcription, Genetic/physiology
14.
Org Biomol Chem ; 13(10): 3040-7, 2015 Mar 14.
Article in English | MEDLINE | ID: mdl-25622264

ABSTRACT

Inflammatory signaling pathways orchestrate the cellular response to infection and injury. These pathways are known to be modulated by compounds that alkylate cysteinyl thiols. One class of phytochemicals with strong thiol alkylating activity is the chalcones. In this study we tested fourteen chalcone derivatives, α-X-substituted 2',3,4,4'-tetramethoxychalcones (α-X-TMCs, X = H, F, Cl, Br, I, CN, Me, p-NO2-C6H4, Ph, p-OMe-C6H4, NO2, CF3, COOEt, COOH), for their ability to modulate inflammatory responses, as monitored by their influence on heme oxygenase-1 (HO-1) activity, inducible nitric oxide synthase (iNOS) activity, and cytokine expression levels. We confirmed that the transcriptional activity of Nrf2 was activated by α-X-TMCs while for NF-κB it was inhibited. For most α-X-TMCs, anti-inflammatory activity was positively correlated with thiol alkylating activity, i.e. stronger electrophiles (X = CF3, Br and Cl) being more potent. Notably, this correlation did not hold true for the strongest electrophiles (X = CN and NO2) which were found to be ineffective as anti-inflammatory compounds. These results emphasize the idea that chemical fine-tuning of electrophilicity is needed to achieve and optimize desired therapeutic effects.


Subject(s)
Anti-Inflammatory Agents/chemistry , Chalcones/chemistry , Inflammation/metabolism , Animals , Cysteamine/chemistry , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Glutathione/chemistry , HeLa Cells , Heme Oxygenase-1/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipopolysaccharides/chemistry , Macrophages/metabolism , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Photochemical Processes , Sulfhydryl Compounds/chemistry , Transcription, Genetic
15.
PLoS One ; 9(6): e99391, 2014.
Article in English | MEDLINE | ID: mdl-24910998

ABSTRACT

Signal transducer and activator of transcription STAT5 is an essential mediator of cytokine, growth factor and hormone signaling. While its activity is tightly regulated in normal cells, its constitutive activation directly contributes to oncogenesis and is associated to a number of hematological and solid tumor cancers. We previously showed that deacetylase inhibitors can inhibit STAT5 transcriptional activity. We now investigated whether the dietary chemopreventive agent sulforaphane, known for its activity as deacetylase inhibitor, might also inhibit STAT5 activity and thus could act as a chemopreventive agent in STAT5-associated cancers. We describe here sulforaphane (SFN) as a novel STAT5 inhibitor. We showed that SFN, like the deacetylase inhibitor trichostatin A (TSA), can inhibit expression of STAT5 target genes in the B cell line Ba/F3, as well as in its transformed counterpart Ba/F3-1*6 and in the human leukemic cell line K562 both of which express a constitutively active form of STAT5. Similarly to TSA, SFN does not alter STAT5 initial activation by phosphorylation or binding to the promoter of specific target genes, in favor of a downstream transcriptional inhibitory effect. Chromatin immunoprecipitation assays revealed that, in contrast to TSA however, SFN only partially impaired the recruitment of RNA polymerase II at STAT5 target genes and did not alter histone H3 and H4 acetylation, suggesting an inhibitory mechanism distinct from that of TSA. Altogether, our data revealed that the natural compound sulforaphane can inhibit STAT5 downstream activity, and as such represents an attractive cancer chemoprotective agent targeting the STAT5 signaling pathway.


Subject(s)
Anticarcinogenic Agents/pharmacology , Biological Products/pharmacology , Isothiocyanates/pharmacology , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Acetylation , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Histones/metabolism , Humans , Interleukin-3/metabolism , Interleukin-3/pharmacology , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/metabolism , Sulfoxides , Transcriptional Activation/drug effects
16.
PLoS One ; 9(3): e90275, 2014.
Article in English | MEDLINE | ID: mdl-24595334

ABSTRACT

Signal transducer and activator of transcription STAT5 and its upstream activating kinase JAK2 are essential mediators of cytokine signaling. Their activity is normally tightly regulated and transient. However, constitutive activation of STAT5 is found in numerous cancers and a driving force for malignant transformation. We describe here the identification of the synthetic chalcone α-Br-2',3,4,4'-tetramethoxychalcone (α-Br-TMC) as a novel JAK/STAT inhibitor. Using the non-transformed IL-3-dependent B cell line Ba/F3 and its oncogenic derivative Ba/F3-1*6 expressing constitutively activated STAT5, we show that α-Br-TMC targets the JAK/STAT pathway at multiple levels, inhibiting both JAK2 and STAT5 phosphorylation. Moreover, α-Br-TMC alters the mobility of STAT5A/B proteins in SDS-PAGE, indicating a change in their post-translational modification state. These alterations correlate with a decreased association of STAT5 and RNA polymerase II with STAT5 target genes in chromatin immunoprecipitation assays. Interestingly, expression of STAT5 target genes such as Cis and c-Myc was differentially regulated by α-Br-TMC in normal and cancer cells. While both genes were inhibited in IL-3-stimulated Ba/F3 cells, expression of the oncogene c-Myc was down-regulated and that of the tumor suppressor gene Cis was up-regulated in transformed Ba/F3-1*6 cells. The synthetic chalcone α-Br-TMC might therefore represent a promising novel anticancer agent for therapeutic intervention in STAT5-associated malignancies.


Subject(s)
Chalcones/pharmacology , Janus Kinase 2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Base Sequence , Cell Line , Chromatin Immunoprecipitation , DNA Primers , Mice , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction
17.
J Am Soc Nephrol ; 24(11): 1830-48, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23990680

ABSTRACT

Mutations of the LMX1B gene cause nail-patella syndrome, a rare autosomal-dominant disorder affecting the development of the limbs, eyes, brain, and kidneys. The characterization of conventional Lmx1b knockout mice has shown that LMX1B regulates the development of podocyte foot processes and slit diaphragms, but studies using podocyte-specific Lmx1b knockout mice have yielded conflicting results regarding the importance of LMX1B for maintaining podocyte structures. In order to address this question, we generated inducible podocyte-specific Lmx1b knockout mice. One week of Lmx1b inactivation in adult mice resulted in proteinuria with only minimal foot process effacement. Notably, expression levels of slit diaphragm and basement membrane proteins remained stable at this time point, and basement membrane charge properties also did not change, suggesting that alternative mechanisms mediate the development of proteinuria in these mice. Cell biological and biophysical experiments with primary podocytes isolated after 1 week of Lmx1b inactivation indicated dysregulation of actin cytoskeleton organization, and time-resolved DNA microarray analysis identified the genes encoding actin cytoskeleton-associated proteins, including Abra and Arl4c, as putative LMX1B targets. Chromatin immunoprecipitation experiments in conditionally immortalized human podocytes and gel shift assays showed that LMX1B recognizes AT-rich binding sites (FLAT elements) in the promoter regions of ABRA and ARL4C, and knockdown experiments in zebrafish support a model in which LMX1B and ABRA act in a common pathway during pronephros development. Our report establishes the importance of LMX1B in fully differentiated podocytes and argues that LMX1B is essential for the maintenance of an appropriately structured actin cytoskeleton in podocytes.


Subject(s)
LIM-Homeodomain Proteins/physiology , Podocytes/cytology , Transcription Factors/physiology , Actins/physiology , Aging , Animals , Apoptosis , Cell Differentiation , Collagen Type IV/genetics , Intracellular Signaling Peptides and Proteins/genetics , LIM-Homeodomain Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Nail-Patella Syndrome/etiology , Oligonucleotide Array Sequence Analysis , Podocytes/chemistry , Podocytes/ultrastructure , Proteinuria/etiology , Transcription Factors/genetics , Zebrafish
18.
J Cell Biol ; 192(4): 631-45, 2011 Feb 21.
Article in English | MEDLINE | ID: mdl-21321097

ABSTRACT

Polycystin-2 (also called TRPP2), an integral membrane protein mutated in patients with cystic kidney disease, is located in the primary cilium where it is thought to transmit mechanical stimuli into the cell interior. After studying a series of polycystin-2 deletion mutants we identified two amino acids in loop 4 that were essential for the trafficking of polycystin-2 to the somatic (nonciliary) plasma membrane. However, polycystin-2 mutant proteins in which these two residues were replaced by alanine were still sorted into the cilium, thus indicating that the trafficking routes to the somatic and ciliary plasma membrane compartments are distinct. We also observed that the introduction of dominant-negative Sar1 mutant proteins and treatment of cells with brefeldin A prevented the transport into the ciliary plasma membrane compartment, whereas metabolic labeling experiments, light microscopical imaging, and high-resolution electron microscopy revealed that full-length polycystin-2 did not traverse the Golgi apparatus on its way to the cilium. These data argue that the transport of polycystin-2 to the ciliary and to the somatic plasma membrane compartments originates in a COPII-dependent fashion at the endoplasmic reticulum, that polycystin-2 reaches the cis side of the Golgi apparatus in either case, but that the trafficking to the somatic plasma membrane goes through the Golgi apparatus whereas transport vesicles to the cilium leave the Golgi apparatus at the cis compartment. Such an interpretation is supported by the finding that mycophenolic acid treatment resulted in the colocalization of polycystin-2 with GM130, a marker of the cis-Golgi apparatus. Remarkably, we also observed that wild-type Smoothened, an integral membrane protein involved in hedgehog signaling that under resting conditions resides in the somatic plasma membrane, passed through the Golgi apparatus, but the M2 mutant of Smoothened, which is constitutively located in the ciliary but not in the somatic plasma membrane, does not. Finally, a dominant-negative form of Rab8a, a BBSome-associated monomeric GTPase, prevented the delivery of polycystin-2 to the primary cilium whereas a dominant-negative form of Rab23 showed no inhibitory effect, which is consistent with the view that the ciliary trafficking of polycystin-2 is regulated by the BBSome.


Subject(s)
Cell Membrane/metabolism , TRPP Cation Channels/metabolism , Animals , COP-Coated Vesicles/physiology , COS Cells , Chlorocebus aethiops , Cilia/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , HeLa Cells , Humans , LLC-PK1 Cells , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Swine , TRPP Cation Channels/analysis , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/physiology
19.
Biochem J ; 433(2): 285-94, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21044049

ABSTRACT

PKD2 is one of the two genes mutated in ADPKD (autosomal-dominant polycystic kidney disease). The protein product of PKD2, polycystin-2, functions as a non-selective cation channel in the endoplasmic reticulum and possibly at the plasma membrane. Hydrophobicity plots and its assignment to the TRP (transient receptor potential) family of cation channels suggest that polycystin-2 contains six transmembrane domains and that both the N- and C-termini extend into the cytoplasm. However, no experimental evidence for this model has so far been provided. To determine the orientation of the different loops of polycystin-2, we truncated polycystin-2 within the predicted loops 1-5 and tagged the constructs at the C-terminus with an HA (haemagglutinin) epitope. After transient expression and selective membrane permeabilization, immunofluorescence staining for the HA epitope revealed that loops 1, 3 and 5 extend into the lumen of the endoplasmic reticulum or the extracellular space, whereas loops 2 and 4 extend into the cytoplasm. This approach also confirmed the cytoplasmic orientation of the N- and C-termini of polycystin-2. In accordance with the immunofluorescence data, protease protection assays from microsomal preparations yielded protected fragments when polycystin-2 was truncated in loops 1, 3 and 5, whereas no protected fragments could be detected when polycystin-2 was truncated in loops 2 and 4. The results of the present study therefore provide the first experimental evidence for the topological orientation of polycystin-2.


Subject(s)
Cell Membrane/chemistry , Intracellular Space/chemistry , TRPP Cation Channels/chemistry , Amino Acid Sequence , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Humans , Intracellular Space/metabolism , Molecular Sequence Data , TRPP Cation Channels/metabolism
20.
Exp Cell Res ; 315(1): 76-96, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18996370

ABSTRACT

LMX1B is a LIM-homeodomain transcription factor essential for development. Putative LMX1B target genes have been identified through mouse gene targeting studies, but their identity as direct LMX1B targets remains hypothetical. We describe here the first molecular characterization of LMX1B target gene regulation. Microarray analysis using a tetracycline-inducible LMX1B expression system in HeLa cells revealed that a subset of NF-kappaB target genes, including IL-6 and IL-8, are upregulated in LMX1B-expressing cells. Inhibition of NF-kappaB activity by short interfering RNA-mediated knock-down of p65 impairs, while activation of NF-kappaB activity by TNF-alpha synergizes induction of NF-kappaB target genes by LMX1B. Chromatin immunoprecipitation demonstrated that LMX1B binds to the proximal promoter of IL-6 and IL-8 in vivo, in the vicinity of the characterized kappaB site, and that LMX1B recruitment correlates with increased NF-kappaB DNA association. IL-6 promoter-reporter assays showed that the kappaB site and an adjacent putative LMX1B binding motif are both involved in LMX1B-mediated transcription. Expression of NF-kappaB target genes is affected in the kidney of Lmx1b(-/-) knock-out mice, thus supporting the biological relevance of our findings. Together, these data demonstrate for the first time that LMX1B directly regulates transcription of a subset of NF-kappaB target genes in cooperation with nuclear p50/p65 NF-kappaB.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/metabolism , NF-kappa B/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Conserved Sequence , HeLa Cells , Homeodomain Proteins/chemistry , Humans , Interferon-beta/pharmacology , Interleukin-6/genetics , Interleukin-8/genetics , Kidney/metabolism , LIM-Homeodomain Proteins , Mice , Mice, Knockout , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding/drug effects , Protein Structure, Tertiary , Sus scrofa , Transcription Factor RelA/metabolism , Transcription Factors/chemistry , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...