Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 43(5): 780-805, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38316991

ABSTRACT

Inflammation is a common condition of prostate tissue, whose impact on carcinogenesis is highly debated. Microbial colonization is a well-documented cause of a small percentage of prostatitis cases, but it remains unclear what underlies the majority of sterile inflammation reported. Here, androgen- independent fluctuations of PSA expression in prostate cells have lead us to identify a prominent function of the Transient Receptor Potential Cation Channel Subfamily M Member 8 (TRPM8) gene in sterile inflammation. Prostate cells secret TRPM8 RNA into extracellular vesicles (EVs), which primes TLR3/NF-kB-mediated inflammatory signaling after EV endocytosis by epithelial cancer cells. Furthermore, prostate cancer xenografts expressing a translation-defective form of TRPM8 RNA contain less collagen type I in the extracellular matrix, significantly more infiltrating NK cells, and larger necrotic areas as compared to control xenografts. These findings imply sustained, androgen-independent expression of TRPM8 constitutes as a promoter of anticancer innate immunity, which may constitute a clinically relevant condition affecting prostate cancer prognosis.


Subject(s)
Prostatic Neoplasms , TRPM Cation Channels , Humans , Male , Androgens , Inflammation/genetics , Interferon Regulatory Factor-3 , Membrane Proteins , NF-kappa B/genetics , Prostatic Neoplasms/genetics , Toll-Like Receptor 3/genetics , TRPM Cation Channels/genetics , Animals
2.
Mol Cancer ; 23(1): 105, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755661

ABSTRACT

BACKGROUND: The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM. METHODS: After analyzing The Cancer Genome Atlas MM patients' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAFV600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo. RESULTS: Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi. CONCLUSIONS: Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner.


Subject(s)
Drug Resistance, Neoplasm , Mechanistic Target of Rapamycin Complex 2 , Melanoma , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Rapamycin-Insensitive Companion of mTOR Protein , Humans , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Melanoma/genetics , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Proto-Oncogene Proteins B-raf/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Drug Resistance, Neoplasm/genetics , Mice , Animals , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic , Mutation , Down-Regulation , Proteomics/methods
3.
Mol Cancer ; 22(1): 133, 2023 08 12.
Article in English | MEDLINE | ID: mdl-37573301

ABSTRACT

Prostate cancer (PCa) is a common and fatal type of cancer in men. Metastatic PCa (mPCa) is a major factor contributing to its lethality, although the mechanisms remain poorly understood. PTEN is one of the most frequently deleted genes in mPCa. Here we show a frequent genomic co-deletion of PTEN and STAT3 in liquid biopsies of patients with mPCa. Loss of Stat3 in a Pten-null mouse prostate model leads to a reduction of LKB1/pAMPK with simultaneous activation of mTOR/CREB, resulting in metastatic disease. However, constitutive activation of Stat3 led to high LKB1/pAMPK levels and suppressed mTORC1/CREB pathway, preventing mPCa development. Metformin, one of the most widely prescribed therapeutics against type 2 diabetes, inhibits mTORC1 in liver and requires LKB1 to mediate glucose homeostasis. We find that metformin treatment of STAT3/AR-expressing PCa xenografts resulted in significantly reduced tumor growth accompanied by diminished mTORC1/CREB, AR and PSA levels. PCa xenografts with deletion of STAT3/AR nearly completely abrogated mTORC1/CREB inhibition mediated by metformin. Moreover, metformin treatment of PCa patients with high Gleason grade and type 2 diabetes resulted in undetectable mTORC1 levels and upregulated STAT3 expression. Furthermore, PCa patients with high CREB expression have worse clinical outcomes and a significantly increased risk of PCa relapse and metastatic recurrence. In summary, we have shown that STAT3 controls mPCa via LKB1/pAMPK/mTORC1/CREB signaling, which we have identified as a promising novel downstream target for the treatment of lethal mPCa.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Prostatic Neoplasms , Animals , Humans , Male , Mice , AMP-Activated Protein Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Metformin/pharmacology , Neoplasm Recurrence, Local , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
4.
Bioessays ; 43(6): e2100078, 2021 06.
Article in English | MEDLINE | ID: mdl-33825205

ABSTRACT

In this report, we look at the challenges posed by the outbreak of COVID-19 and how the Executive Board of these two congresses succeeded in overcoming those challenges and holding two congresses. The approach for a large festival with different virtual setting components provided a suitable solution that led to exemplary achievements and created an appropriate model for future virtual or combined virtual and face-to-face events. These events proved that pandemic problems could not limit the organizers, pushing them to make better use of the facilities and turning this threat into an opportunity.


Subject(s)
COVID-19 , Congresses as Topic/organization & administration , Genetics , Twins , Awards and Prizes , Biomedical Research , COVID-19/epidemiology , Humans , Iran
5.
Int J Mol Sci ; 21(24)2020 Dec 12.
Article in English | MEDLINE | ID: mdl-33322692

ABSTRACT

Biological systems respond to perturbations through the rewiring of molecular interactions, organised in gene regulatory networks (GRNs). Among these, the increasingly high availability of transcriptomic data makes gene co-expression networks the most exploited ones. Differential co-expression networks are useful tools to identify changes in response to an external perturbation, such as mutations predisposing to cancer development, and leading to changes in the activity of gene expression regulators or signalling. They can help explain the robustness of cancer cells to perturbations and identify promising candidates for targeted therapy, moreover providing higher specificity with respect to standard co-expression methods. Here, we comprehensively review the literature about the methods developed to assess differential co-expression and their applications to cancer biology. Via the comparison of normal and diseased conditions and of different tumour stages, studies based on these methods led to the definition of pathways involved in gene network reorganisation upon oncogenes' mutations and tumour progression, often converging on immune system signalling. A relevant implementation still lagging behind is the integration of different data types, which would greatly improve network interpretability. Most importantly, performance and predictivity evaluation of the large variety of mathematical models proposed would urgently require experimental validations and systematic comparisons. We believe that future work on differential gene co-expression networks, complemented with additional omics data and experimentally tested, will considerably improve our insights into the biology of tumours.


Subject(s)
Computational Biology/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks , Neoplasms/metabolism , Signal Transduction/genetics , Algorithms , Disease Progression , Gene Expression Profiling , Humans , Neoplasms/genetics , Transcriptome/genetics
6.
J Neurosci ; 37(16): 4255-4269, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28320842

ABSTRACT

After nerve injury, Schwann cells convert to a phenotype specialized to promote repair. But during the slow process of axonal regrowth, these repair Schwann cells gradually lose their regeneration-supportive features and eventually die. Although this is a key reason for the frequent regeneration failures in humans, the transcriptional mechanisms that control long-term survival and phenotype of repair cells have not been studied, and the molecular signaling underlying their decline is obscure. We show, in mice, that Schwann cell STAT3 has a dual role. It supports the long-term survival of repair Schwann cells and is required for the maintenance of repair Schwann cell properties. In contrast, STAT3 is less important for the initial generation of repair Schwann cells after injury. In repair Schwann cells, we find that Schwann cell STAT3 activation by Tyr705 phosphorylation is sustained during long-term denervation. STAT3 is required for maintaining autocrine Schwann cell survival signaling, and inactivation of Schwann cell STAT3 results in a striking loss of repair cells from chronically denervated distal stumps. STAT3 inactivation also results in abnormal morphology of repair cells and regeneration tracks, and failure to sustain expression of repair cell markers, including Shh, GDNF, and BDNF. Because Schwann cell development proceeds normally without STAT3, the function of this factor appears restricted to Schwann cells after injury. This identification of transcriptional mechanisms that support long-term survival and differentiation of repair cells will help identify, and eventually correct, the failures that lead to the deterioration of this important cell population.SIGNIFICANCE STATEMENT Although injured peripheral nerves contain repair Schwann cells that provide signals and spatial clues for promoting regeneration, the clinical outcome after nerve damage is frequently poor. A key reason for this is that, during the slow growth of axons through the proximal parts of injured nerves repair, Schwann cells gradually lose regeneration-supporting features and eventually die. Identification of signals that sustain repair cells is therefore an important goal. We have found that in mice the transcription factor STAT3 protects these cells from death and contributes to maintaining the molecular and morphological repair phenotype that promotes axonal regeneration. Defining the molecular mechanisms that maintain repair Schwann cells is an essential step toward developing therapeutic strategies that improve nerve regeneration and functional recovery.


Subject(s)
Nerve Regeneration , Peripheral Nerve Injuries/metabolism , Phenotype , STAT3 Transcription Factor/genetics , Schwann Cells/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Female , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mice , STAT3 Transcription Factor/metabolism , Schwann Cells/cytology
7.
Hum Mol Genet ; 25(4): 740-54, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26685160

ABSTRACT

The congenital malformation split hand/foot (SHFM) is characterized by missing central fingers and dysmorphology or fusion of the remaining ones. Type-1 SHFM is linked to deletions/rearrangements of the DLX5-DLX6 locus and point mutations in the DLX5 gene. The ectrodactyly phenotype is reproduced in mice by the double knockout (DKO) of Dlx5 and Dlx6. During limb development, the apical ectodermal ridge (AER) is a key-signaling center responsible for early proximal-distal growth and patterning. In Dlx5;6 DKO hindlimbs, the central wedge of the AER loses multilayered organization and shows down-regulation of FGF8 and Dlx2. In search for the mechanism, we examined the non-canonical Wnt signaling, considering that Dwnt-5 is a target of distalless in Drosophila and the knockout of Wnt5, Ryk, Ror2 and Vangl2 in the mouse causes severe limb malformations. We found that in Dlx5;6 DKO limbs, the AER expresses lower levels of Wnt5a, shows scattered ß-catenin responsive cells and altered basolateral and planar cell polarity (PCP). The addition of Wnt5a to cultured embryonic limbs restored the expression of AER markers and its stratification. Conversely, the inhibition of the PCP molecule c-jun N-terminal kinase caused a loss of AER marker expression. In vitro, the addition of Wnt5a on mixed primary cultures of embryonic ectoderm and mesenchyme was able to confer re-polarization. We conclude that the Dlx-related ectrodactyly defect is associated with the loss of basoapical and PCP, due to reduced Wnt5a expression and that the restoration of the Wnt5a level is sufficient to partially reverts AER misorganization and dysmorphology.


Subject(s)
Homeodomain Proteins/genetics , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/pathology , Wnt-5a Protein/pharmacology , Animals , Cell Polarity/drug effects , Cell Polarity/physiology , Disease Models, Animal , Down-Regulation , Ectoderm/metabolism , Ectoderm/pathology , Homeodomain Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Limb Deformities, Congenital/drug therapy , Limb Deformities, Congenital/metabolism , Mesoderm/metabolism , Mice , Mice, Knockout , Trans-Activators/genetics , Wnt Signaling Pathway , Wnt-5a Protein/biosynthesis , Wnt-5a Protein/deficiency , Wnt-5a Protein/genetics , beta Catenin/metabolism
8.
Int J Mol Sci ; 19(9)2018 Sep 18.
Article in English | MEDLINE | ID: mdl-30231582

ABSTRACT

The transcription factor signal transducer and activator of transcription (STAT)3 mediates the functions of cytokines, growth factors, and oncogenes under both physiological and pathological conditions. Uncontrolled/constitutive STAT3 activity is often detected in tumors of different types, where its role is mostly that of an oncogene, contributing in multiple ways to tumor transformation, growth, and progression. For this reason, many laboratories and pharmaceutical companies are making efforts to develop specific inhibitors. However, STAT3 has also been shown to act as a tumor suppressor in a number of cases, suggesting that its activity is strongly context-specific. Here, we discuss the bases that can explain the multiple roles of this factor in both physiological and pathological contexts. In particular, we focus on the following four features: (i) the distinct properties of the STAT3α and ß isoforms; (ii) the multiple post-translational modifications (phosphorylation on tyrosine or serine, acetylation and methylation on different residues, and oxidation and glutathionylation) that can affect its activities downstream of multiple different signals; (iii) the non-canonical functions in the mitochondria, contributing to the maintenance of energy homeostasis under stress conditions; and (iv) the recently discovered functions in the endoplasmic reticulum, where STAT3 contributes to the regulation of calcium homeostasis, energy production, and apoptosis.


Subject(s)
Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis , Cell Nucleus/genetics , Cell Nucleus/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Energy Metabolism , Humans , Mitochondria/genetics , Mitochondria/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oncogenes , Protein Processing, Post-Translational , STAT3 Transcription Factor/analysis , STAT3 Transcription Factor/genetics
9.
Int J Mol Sci ; 19(1)2018 Jan 06.
Article in English | MEDLINE | ID: mdl-29316631

ABSTRACT

Signal Transducer and Activator of Transcription (STAT)3 has recently emerged as a key player in the development and pathogenesis of psoriasis and psoriatic-like inflammatory conditions. Indeed, STAT3 hyperactivation has been reported in virtually every cell type involved in disease initiation and maintenance, and this factor mediates the signal of most cytokines that are involved in disease pathogenesis, including the central Interleukin (IL)-23/IL-17/IL-22 axis. Despite the recent availability of effective biological agents (monoclonal antibodies) against IL-17 and IL-23, which have radically changed the current standard of disease management, the possibility of targeting either STAT3 itself or, even better, the family of upstream activators Janus kinases (JAK1, 2, 3, and TYK2) offers additional therapeutic options. Due to the oral/topical administration modality of these small molecule drugs, their lower cost, and the reduced risk of eliciting adverse immune responses, these compounds are being actively scrutinized in clinical settings. Here, we summarize the main pathological features of psoriatic conditions that provide the rationale for targeting the JAK/STAT3 axis in disease treatment.


Subject(s)
Psoriasis/pathology , STAT3 Transcription Factor/metabolism , Humans , Interleukin-17/metabolism , Interleukin-23/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use , Psoriasis/drug therapy , Psoriasis/metabolism , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism
10.
Cytokine ; 98: 42-50, 2017 10.
Article in English | MEDLINE | ID: mdl-28579221

ABSTRACT

The transcription factor signal transducer and activator of transcription (STAT) 3 is activated downstream of cytokines, growth factors and oncogenes to mediate their functions under both physiological and pathological conditions. In particular, aberrant/unrestrained STAT3 activity is detected in a wide variety of tumors, driving multiple pro-oncogenic functions. For that, STAT3 is widely considered as an oncogene and is the object of intense translational studies. One of the distinctive features of this factor is however, its ability to elicit different and sometimes contrasting effects under different conditions. In particular, STAT3 activities have been shown to be either pro-oncogenic or tumor-suppressive according to the tumor aetiology/mutational landscape, suggesting that the molecular bases underlining its functions are still incompletely understood. Here we discuss some of the properties that may provide the bases to explain STAT3 heterogeneous functions, and in particular how post-translational modifications contribute shaping its sub-cellular localization and activities, the cross talk between these activities and cell metabolic conditions, and finally how its functions can control the behaviour of both tumor and tumor microenvironment cell populations.


Subject(s)
Neoplasms/physiopathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Animals , Autophagy , Cell Line, Tumor , Energy Metabolism , Humans , Mice , Protein Processing, Post-Translational , Signal Transduction , Tumor Microenvironment
11.
J Am Soc Nephrol ; 27(12): 3690-3705, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27153926

ABSTRACT

In CKD, tubular cells may be involved in the induction of interstitial fibrosis, which in turn, leads to loss of renal function. However, the molecular mechanisms that link tubular cells to the interstitial compartment are not clear. Activation of the Stat3 transcription factor has been reported in tubular cells after renal damage, and Stat3 has been implicated in CKD progression. Here, we combined an experimental model of nephron reduction in mice from different genetic backgrounds and genetically modified animals with in silico and in vitro experiments to determine whether the selective activation of Stat3 in tubular cells is involved in the development of interstitial fibrosis. Nephron reduction caused Stat3 phosphorylation in tubular cells of lesion-prone mice but not in resistant mice. Furthermore, specific deletion of Stat3 in tubular cells significantly reduced the extent of interstitial fibrosis, which correlated with reduced fibroblast proliferation and matrix synthesis, after nephron reduction. Mechanistically, in vitro tubular Stat3 activation triggered the expression of a specific subset of paracrine profibrotic factors, including Lcn2, Pdgfb, and Timp1. Together, our results provide a molecular link between tubular and interstitial cells during CKD progression and identify Stat3 as a central regulator of this link and a promising therapeutic target.


Subject(s)
Cell Communication , Kidney Tubules/cytology , Renal Insufficiency, Chronic/physiopathology , STAT3 Transcription Factor/physiology , Animals , Female , Mice
12.
Blood ; 124(15): 2370-9, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25185262

ABSTRACT

The members of the signal transducer and activator of transcription (STAT) family of transcription factors modulate the development and function of natural killer (NK) cells. NK cell-mediated tumor surveillance is particularly important in the body's defense against hematological malignancies such as leukemia. STAT3 inhibitors are currently being developed, although their potential effects on NK cells are not clear. We have investigated the function of STAT3 in NK cells with Stat3(Δ/Δ)Ncr1-iCreTg mice, whose NK cells lack STAT3. In the absence of STAT3, NK cells develop normally and in normal numbers, but display alterations in the kinetics of interferon-γ (IFN-γ) production. We report that STAT3 directly binds the IFN-γ promoter. In various in vivo models of hematological diseases, loss of STAT3 in NK cells enhances tumor surveillance. The reduced tumor burden is paralleled by increased expression of the activating receptor DNAM-1 and the lytic enzymes perforin and granzyme B. Our findings imply that STAT3 inhibitors will stimulate the cytolytic activity of NK cells against leukemia, thereby providing an additional therapeutic benefit.


Subject(s)
Immunologic Surveillance , Killer Cells, Natural/metabolism , Neoplasms/immunology , Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antigens, Ly/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytokines/pharmacology , Disease Models, Animal , Granzymes/metabolism , Immunologic Surveillance/drug effects , Integrases/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Intestines/pathology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Kinetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/metabolism , Perforin/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Spleen/pathology
13.
NMR Biomed ; 29(4): 475-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26866929

ABSTRACT

Herein, a new relaxometric method for the assessment of intestinal permeability based on the oral administration of clinically approved gadolinium (Gd)-based MRI contrast agents (CAs) is proposed. The fast, easily performed and cheap measurement of the longitudinal water proton relaxation rate (R1) in urine reports the amount of paramagnetic probe that has escaped the gastrointestinal tract. The proposed method appears to be a compelling alternative to the available methods for the assessment of intestinal permeability. The method was tested on the murine model of dextran sulfate sodium (DSS)-induced colitis in comparison with healthy mice. Three CAs were tested, namely ProHance®, MultiHance® and Magnevist®. Urine was collected for 24 h after the oral ingestion of the Gd-containing CA at day 3-4 (severe damage stage) and day 8-9 (recovery stage) after treatment with DSS. The Gd content in urine measured by (1)H relaxometry was confirmed by inductively coupled plasma-mass spectrometry (ICP-MS). The extent of urinary excretion was given as a percentage of excreted Gd over the total ingested dose. The method was validated by comparing the results obtained with the established methodology based on the lactulose/mannitol and sucralose tests. For ProHance and Magnevist, the excreted amounts in the severe stage of damage were 2.5-3 times higher than in control mice. At the recovery stage, no significant differences were observed with respect to healthy mice. Overall, a very good correlation with the lactulose/mannitol and sucralose results was obtained. In the case of MultiHance, the percentage of excreted Gd complex was not significantly different from that of control mice in either the severe or recovery stages. The difference from ProHance and Magnevist was explained on the basis of the (known) partial biliary excretion of MultiHance in mice.


Subject(s)
Contrast Media/administration & dosage , Gadolinium/administration & dosage , Intestines/pathology , Magnetic Resonance Imaging/methods , Administration, Oral , Animals , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Mice, Inbred BALB C , Permeability , Reproducibility of Results
14.
J Immunol ; 192(1): 447-58, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24319264

ABSTRACT

We have previously characterized mouse CMV (MCMV)-encoded immune-evasive IFN signaling inhibition and identified the viral protein pM27 as inducer of proteasomal degradation of STAT2. Extending our analysis to STAT1 and STAT3, we found that MCMV infection neither destabilizes STAT1 protein nor prevents STAT1 tyrosine Y701 phosphorylation, nuclear translocation, or the capability to bind γ-activated sequence DNA-enhancer elements. Unexpectedly, the analysis of STAT3 revealed an induction of STAT3 Y705 phosphorylation by MCMV. In parallel, we found decreasing STAT3 protein amounts upon MCMV infection, although STAT3 expression normally is positive autoregulative. STAT3 phosphorylation depended on the duration of MCMV infection, the infectious dose, and MCMV gene expression but was independent of IFNAR1, IL-10, IL-6, and JAK2. Although STAT3 phosphorylation did not require MCMV immediate early 1, pM27, and late gene expression, it was restricted to MCMV-infected cells and not transmitted to bystander cells. Despite intact STAT1 Y701 phosphorylation, IFN-γ-induced target gene transcription (e.g., IRF1 and suppressor of cytokine signaling [SOCS] 1) was strongly impaired. Likewise, the induction of STAT3 target genes (e.g., SOCS3) by IL-6 was also abolished, indicating that MCMV antagonizes STAT1 and STAT3 despite the occurrence of tyrosine phosphorylation. Consistent with the lack of SOCS1 induction, STAT1 phosphorylation was prolonged upon IFN-γ treatment. We conclude that the inhibition of canonical STAT1 and STAT3 target gene expression abrogates their intrinsic negative feedback loops, leading to accumulation of phospho-tyrosine-STAT3 and prolonged STAT1 phosphorylation. These findings challenge the generalization of tyrosine-phosphorylated STATs necessarily being transcriptional active and document antagonistic effects of MCMV on STAT1/3-dependent target gene expression.


Subject(s)
Janus Kinases/metabolism , Muromegalovirus/physiology , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Cell Line , Cell Nucleus/metabolism , Cytokine Receptor gp130/metabolism , Gene Expression Regulation , Gene Expression Regulation, Viral , Immediate-Early Proteins/metabolism , Interferon-gamma/pharmacology , Interleukin-10/metabolism , Interleukin-6/metabolism , Janus Kinase 2/metabolism , Mice , Phosphorylation/drug effects , Protein Binding , Protein Transport , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism
15.
Exp Dermatol ; 24(1): 29-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25382846

ABSTRACT

STAT3, a pleiotropic transcription factor acting downstream of cytokines and growth factors, is known to enhance proliferation, migration, invasion and aerobic glycolysis in tumors upon aberrant activation. In the murine epidermis, STAT3 is necessary for experimentally induced carcinogenesis. Skin tumorigenesis is conversely enhanced by overexpression in keratinocytes of the constitutively active STAT3C mutant, which also induces robust, psoriasis-like epidermal hyperplasia. We show here that STAT3C expression at physiological levels in knock-in mice leads to mild epidermal hyperplasia and attenuated expression of terminal differentiation markers. Altered differentiation is confirmed in isolated primary epidermal keratinocytes in vitro, correlating with enhanced proliferative and clonogenic potential, attenuated senescence and, strikingly, high-frequency spontaneous immortalization. These results suggest that moderate levels of continuous STAT3 activation, which closely resemble those triggered by chronic inflammation or persistent growth factor stimulation, may establish a preneoplastic state in part by promoting the escape of epidermal progenitor cells from differentiation and senescence checkpoints.


Subject(s)
Cell Differentiation , Cellular Senescence , Epidermal Cells , Keratinocytes/metabolism , STAT3 Transcription Factor/metabolism , Animals , Animals, Newborn , Cell Movement , Cell Proliferation , Glycolysis , Hyperplasia/metabolism , Keratinocytes/cytology , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Skin/metabolism , Skin Aging , Stem Cells/cytology , beta-Galactosidase/metabolism
16.
Int J Cancer ; 135(9): 1997-2003, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24500994

ABSTRACT

The transcription factor Signal Transducer and Activator of Transcription (STAT)3 has been considered as a potential anticancer target since its first description as an oncogene in 1999, recently leading to STAT3 inhibitors been brought to clinical trial for the treatment of solid tumors. However, the past 14 years of intense basic research have uncovered novel STAT3-mediated pathways that could affect the outcome of the designed therapies while at the same time help designing function-specific inhibitors. Particularly intriguing are the recent findings that suggest profound implications of STAT3 with the regulation of cellular metabolism in both canonical, that is transcriptional, and non-canonical ways. Here, after a short description of the main known features of STAT3 signaling and function, we review the recent literature on the role of STAT3 in regulating cellular metabolism and discuss the potential consequences on the therapeutic approaches currently under clinical experimentation.


Subject(s)
Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology , STAT3 Transcription Factor/metabolism
17.
Cancer Immunol Res ; 12(4): 440-452, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38331413

ABSTRACT

Tumor neoantigens (nAg) represent a promising target for cancer immunotherapy. The identification of nAgs that can generate T-cell responses and have therapeutic activity has been challenging. Here, we sought to unravel the features of nAgs required to induce tumor rejection. We selected clinically validated Great Ape-derived adenoviral vectors (GAd) as a nAg delivery system for differing numbers and combinations of nAgs. We assessed their immunogenicity and efficacy in murine models of low to high disease burden, comparing multi-epitope versus mono-epitope vaccines. We demonstrated that the breadth of immune response is critical for vaccine efficacy and having multiple immunogenic nAgs encoded in a single vaccine improves efficacy. The contribution of each single neoantigen was examined, leading to the identification of 2 nAgs able to induce CD8+ T cell-mediated tumor rejection. They were both active as individual nAgs in a setting of prophylactic vaccination, although to different extents. However, the efficacy of these single nAgs was lost in a setting of therapeutic vaccination in tumor-bearing mice. The presence of CD4+ T-cell help restored the efficacy for only the most expressed of the two nAgs, demonstrating a key role for CD4+ T cells in sustaining CD8+ T-cell responses and the necessity of an efficient recognition of the targeted epitopes on cancer cells by CD8+ T cells for an effective antitumor response. This study provides insight into understanding the determinants of nAgs relevant for effective treatment and highlights features that could contribute to more effective antitumor vaccines. See related Spotlight by Slingluff Jr, p. 382.


Subject(s)
Cancer Vaccines , Neoplasms , Mice , Animals , Tumor Burden , CD8-Positive T-Lymphocytes , CD4-Positive T-Lymphocytes , Epitopes , Antigens, Neoplasm
18.
Cell Death Dis ; 15(5): 369, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806478

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is frequently overexpressed in patients with acute myeloid leukemia (AML). STAT3 exists in two distinct alternatively spliced isoforms, the full-length isoform STAT3α and the C-terminally truncated isoform STAT3ß. While STAT3α is predominantly described as an oncogenic driver, STAT3ß has been suggested to act as a tumor suppressor. To elucidate the role of STAT3ß in AML, we established a mouse model of STAT3ß-deficient, MLL-AF9-driven AML. STAT3ß deficiency significantly shortened survival of leukemic mice confirming its role as a tumor suppressor. Furthermore, RNA sequencing revealed enhanced STAT1 expression and interferon (IFN) signaling upon loss of STAT3ß. Accordingly, STAT3ß-deficient leukemia cells displayed enhanced sensitivity to blockade of IFN signaling through both an IFNAR1 blocking antibody and the JAK1/2 inhibitor Ruxolitinib. Analysis of human AML patient samples confirmed that elevated expression of IFN-inducible genes correlated with poor overall survival and low STAT3ß expression. Together, our data corroborate the tumor suppressive role of STAT3ß in a mouse model in vivo. Moreover, they provide evidence that its tumor suppressive function is linked to repression of the STAT1-mediated IFN response. These findings suggest that the STAT3ß/α mRNA ratio is a significant prognostic marker in AML and holds crucial information for targeted treatment approaches. Patients displaying a low STAT3ß/α mRNA ratio and unfavorable prognosis could benefit from therapeutic interventions directed at STAT1/IFN signaling.


Subject(s)
Leukemia, Myeloid, Acute , STAT3 Transcription Factor , Animals , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Humans , STAT3 Transcription Factor/metabolism , Mice , Signal Transduction , Interferons/metabolism , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Cell Line, Tumor , Nitriles , Pyrazoles , Pyrimidines
19.
Hum Mutat ; 34(5): 735-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23420607

ABSTRACT

Polymorphisms in regulatory DNA regions are believed to play an important role in determining phenotype, including disease, and in providing raw material for evolution. We devised a new pipeline for the systematic identification of functional variation in human regulatory sequences. The algorithm is based on the identification of SNPs leading to significant changes in both the affinity of a regulatory region for transcription factors (TFs) and the expression in vivo of the regulated gene. We tested the algorithm by identifying SNPs leading to altered regulation by STAT3 in human promoters and introns, and experimentally validated the top-scoring ones, showing that most of the SNPs identified by the algorithm indeed correspond to differential binding of STAT3 and differential induction of the target gene upon stimulation with IL6. Using the same computational approach, we compiled a database of thousands of predicted functional regulatory SNPs for hundreds of human TFs, which we provide as online Supporting Information. We discuss possible applications to the interpretation of noncoding SNPs associated with human diseases. The method we propose and the database of predicted functional cis-regulatory polymorphisms will be useful in future studies of regulatory variation and in particular to interpret the results of past and future genome-wide association studies.


Subject(s)
Genome, Human , Polymorphism, Single Nucleotide , Regulatory Sequences, Nucleic Acid , Algorithms , Cell Line , Databases, Protein , Humans , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/genetics
20.
J Cell Sci ; 124(Pt 6): 900-9, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21325026

ABSTRACT

STAT3 is an important transcription factor involved in immunity and cancer. In response to cytokine stimulation, STAT3 becomes phosphorylated on a single tyrosine residue. Tyrosine-phosphorylated STAT3 accumulates in the nucleus, binds to specific DNA response elements and induces gene expression. Unphosphorylated, latent STAT3 shuttles constitutively between cytoplasm and nucleus. We analysed the importance of previously identified putative nuclear localization sequences (NLS) and nuclear export sequences (NES) for nucleocytoplasmic shuttling of latent STAT3 using STAT3-deficient cells reconstituted with fluorescently labelled STAT3 mutants. Mutation of a putative NLS or NES sequence did not impair nucleocytoplasmic shuttling of latent STAT3. We were also interested in the structural requirements for dimerization of unphosphorylated STAT3 and its relevance for nucleocytoplasmic shuttling. By native gel electrophoresis and dual-focus fluorescence correlation spectroscopy (2f-FCS) we identified the N-terminal domain (amino acids 1-125) to be essential for formation of unphosphorylated STAT3 dimers but not for assembly of tyrosine-phosphorylated STAT3 dimers. In resting cells, the monomeric N-terminal deletion mutant (STAT3-ΔNT) shuttles faster between the cytoplasm and nucleus than the wild-type STAT3, indicating that dimer formation is not required for nucleocytoplasmic shuttling of latent STAT3. STAT3-ΔNT becomes phosphorylated and dimerizes in response to interleukin-6 stimulation but, surprisingly, does not accumulate in the nucleus. These results highlight the importance of the N-terminal domain in the formation of unphosphorylated STAT3 dimers and nuclear accumulation of STAT3 upon phosphorylation.


Subject(s)
Cell Nucleus/metabolism , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/genetics , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , Dimerization , Mice , Phosphorylation , Protein Structure, Tertiary , STAT3 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL