Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Clin Transl Oncol ; 24(1): 57-65, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34275119

ABSTRACT

PURPOSE: Ovarian cancer (OC) is a common malignancy, and IFN-γ, a multifunctional cytokine, is unveiled to impede the multiplication and enhance apoptosis in diverse tumor cells in previous research. Nonetheless, its function and mechanism in OC are blurred. METHODS: OC cell lines SKOV3 and OVCAR3 were dealt with different concentrations (0-40 ng/ml) of IFN-γ. CCK-8 experiment was utilized to examine cell multiplication; Flow cytometry was executed to detect apoptosis and cell cycle; Wound healing assay was utilized to detect cell migration; and Transwell experiment was implemented to examine cell invasion. qRT-PCR analysis was applied to detect STAT5, STAT3, JAK2 and JAK3 mRNA expression in OC cell lines. Western blot experiment was applied to detect the protein and phosphorylation levels of SOCS1, STAT5 and STAT3. RESULTS: IFN-γ suppressed OC cell multiplication in a concentration-dependent manner. Relative to the control group, IFN-γ restrained OC cell migration, invasion, enhanced apoptosis and prevented cell transformation from G0/G1 to S phase. Further analysis revealed that IFN-γ up-modulated SOCS1 expression and impeded STAT5 and STAT3 protein phosphorylation levels, and knockdown of SOCS1 partially counteracted the inhibitory effect of IFN-γ on STAT5 and STAT3 protein phosphorylation levels. CONCLUSION: IFN-γ represses OC progression by facilitating SOCS1 to suppress STAT3 and STAT5 protein phosphorylation.


Subject(s)
Interferon-gamma/physiology , Janus Kinase 2/physiology , Ovarian Neoplasms/pathology , STAT5 Transcription Factor/physiology , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/physiology , Disease Progression , Female , Humans , STAT3 Transcription Factor
2.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34967898

ABSTRACT

Leydig cells produce androgens that are essential for male sex differentiation and reproductive function. Leydig cell function is regulated by several hormones and signaling molecules, including growth hormone (GH). Although GH is known to upregulate Star gene expression in Leydig cells, its molecular mechanism of action remains unknown. The STAT5B transcription factor is a downstream effector of GH signaling in other systems. While STAT5B is present in both primary and Leydig cell lines, its function in these cells has yet to be ascertained. Here we report that treatment of MA-10 Leydig cells with GH or overexpression of STAT5B induces Star messenger RNA levels and increases steroid hormone output. The mouse Star promoter contains a consensus STAT5B element (TTCnnnGAA) at -756 bp to which STAT5B binds in vitro (electrophoretic mobility shift assay and supershift) and in vivo (chromatin immunoprecipitation) in a GH-induced manner. In functional promoter assays, STAT5B was found to activate a -980 bp mouse Star reporter. Mutating the -756 bp element prevented STAT5B binding but did not abrogate STAT5B-responsiveness. STAT5B was found to functionally cooperate with DNA-bound cJUN. The STAT5B/cJUN cooperation was only observed in Leydig cells and not in Sertoli or fibroblast cells, indicating that additional Leydig cell-enriched transcription factors are required. The STAT5B/cJUN cooperation was lost only when both STAT5B and cJUN elements were mutated. In addition to identifying the Star gene as a novel target for STAT5B in Leydig cells, our data provide important new insights into the mechanism of GH and STAT5B action in the regulation of Leydig cell function.


Subject(s)
Growth Hormone/pharmacology , Leydig Cells/metabolism , Phosphoproteins/genetics , Proto-Oncogene Proteins c-jun/physiology , STAT5 Transcription Factor/genetics , Animals , Base Sequence , Binding Sites , Cell Line , DNA/chemistry , DNA/metabolism , Gene Expression/drug effects , Leydig Cells/classification , Male , Mice , Phosphoproteins/analysis , Phosphoproteins/physiology , Promoter Regions, Genetic , RNA, Messenger/analysis , STAT5 Transcription Factor/analysis , STAT5 Transcription Factor/physiology , Up-Regulation/drug effects
3.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281163

ABSTRACT

Erythropoietin (EPO) acts on multiple tissues through its receptor EPOR, a member of a cytokine class I receptor superfamily with pleiotropic effects. The interaction of EPO and EPOR triggers the activation of several signaling pathways that induce erythropoiesis, including JAK2/STAT5, PI3K/AKT, and MAPK. The canonical EPOR/JAK2/STAT5 pathway is a known regulator of differentiation, proliferation, and cell survival of erythroid progenitors. In addition, its role in the protection of other cells, including cancer cells, is under intense investigation. The involvement of EPOR/JAK2/STAT5 in other processes such as mRNA splicing, cytoskeleton reorganization, and cell metabolism has been recently described. The transcriptomics, proteomics, and epigenetic studies reviewed in this article provide a detailed understanding of EPO signalization. Advances in this area of research may be useful for improving the efficacy of EPO therapy in hematologic disorders, as well as in cancer treatment.


Subject(s)
Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/physiology , Animals , Cell Differentiation/drug effects , Epigenomics/methods , Erythropoiesis/drug effects , Erythropoietin/physiology , Humans , Janus Kinase 2/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/metabolism , Receptors, Erythropoietin/physiology , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcriptome/genetics
4.
Front Immunol ; 12: 642856, 2021.
Article in English | MEDLINE | ID: mdl-34054809

ABSTRACT

Invariant NKT (iNKT) cells are thymus-generated innate-like T cells, comprised of three distinct subsets with divergent effector functions. The molecular mechanism that drives the lineage trifurcation of immature iNKT cells into the NKT1, NKT2, and NKT17 subsets remains a controversial issue that remains to be resolved. Because cytokine receptor signaling is necessary for iNKT cell generation, cytokines are proposed to contribute to iNKT subset differentiation also. However, the precise roles and requirements of cytokines in these processes are not fully understood. Here, we show that IL-2Rß, a nonredundant component of the IL-15 receptor complex, plays a critical role in both the development and differentiation of thymic iNKT cells. While the induction of IL-2Rß expression on postselection thymocytes is necessary to drive the generation of iNKT cells, surprisingly, premature IL-2Rß expression on immature iNKT cells was detrimental to their development. Moreover, while IL-2Rß is necessary for NKT1 generation, paradoxically, we found that the increased abundance of IL-2Rß suppressed NKT1 generation without affecting NKT2 and NKT17 cell differentiation. Thus, the timing and abundance of IL-2Rß expression control iNKT lineage fate and development, thereby establishing cytokine receptor expression as a critical regulator of thymic iNKT cell differentiation.


Subject(s)
Interleukin-2 Receptor beta Subunit/physiology , Natural Killer T-Cells/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Interleukin-15/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/classification , Natural Killer T-Cells/cytology , STAT5 Transcription Factor/physiology
5.
J Invest Dermatol ; 141(10): 2449-2458, 2021 10.
Article in English | MEDLINE | ID: mdl-33862068

ABSTRACT

Staphylococcal enterotoxins are believed to fuel disease activity in cutaneous T-cell lymphoma. Recent data support this by showing that antibiotics inhibit malignant T cells in skin lesions in mycosis fungoides and Sézary syndrome, the most common forms of cutaneous T-cell lymphoma. Yet, it remains incompletely characterized how staphylococcal enterotoxins fuel disease activity. In this study, we show that staphylococcal enterotoxins induce the expression of the oncogenic microRNA miR-155 in primary malignant T cells. Thus, staphylococcal enterotoxins and Staphyloccocus aureus isolates from lesional skin of patients induce miR-155 expression at least partly through the IL-2Rg‒Jak‒signal transducer and activator of transcription 5 pathway, and the effect is augmented by the presence of nonmalignant T cells. Importantly, mycosis fungoides lesions harbor S. aureus, express Y-phosphorylated signal transducer and activator of transcription 5, and display enhanced miR-155 expression, when compared with nonlesional and healthy skin. Preliminary data show that aggressive antibiotic therapy is associated with decreased Y-phosphorylated signal transducer and activator of transcription 5 and miR-155 expression in lesional skin in two patients with Sézary syndrome. In conclusion, we show that S. aureus and its enterotoxins induce enhanced expression of oncogenic miR-155, providing mechanistic insight into the role of S. aureus in cutaneous T-cell lymphoma. Our findings support that environmental stimuli such as bacteria can fuel disease progression in cutaneous T-cell lymphoma.


Subject(s)
Enterotoxins/toxicity , Lymphoma, T-Cell, Cutaneous/etiology , MicroRNAs/physiology , STAT5 Transcription Factor/physiology , Skin Neoplasms/etiology , Staphylococcus aureus/pathogenicity , Anti-Bacterial Agents/pharmacology , Cell Line, Tumor , Humans
6.
FASEB J ; 35(3): e21283, 2021 03.
Article in English | MEDLINE | ID: mdl-33617050

ABSTRACT

The pathological characteristics of osteoarthritis are cartilage matrix degradation, chondrocytes apoptosis, and low-grade inflammation of the joint. Recent studies have shown that blood vessels grow from the subchondral bone to the articular cartilage. However, the relationship among inflammation, angiogenesis, and chondrocyte apoptosis is still unclear. We found that chondrocytes could secrete chemokines and VEGF to promote the migration of vascular endothelial cells in response to TNF-α stimulation. The invasion of blood vessels leads to increased oxygen tension in the local environment, which increased the expression of SETD7 in chondrocytes by activating the JAK-STAT5 pathway. The bond of phosphorylated STAT5 and the specific locus in the promoter of SETD7 directly increased the transcription of SETD7. On the one hand, SETD7-regulated chemokine expression by forming a positive loop; on the other hand, SETD7-mediated chondrocyte apoptosis by inhibiting the nuclear localization of HIF-1α. In this study, we discovered a novel function of chondrocytes as mediators of inflammation and angiogenesis. Our study demonstrates that SETD7 is a potential molecular target to prevent OA development and progression.


Subject(s)
Apoptosis , Cartilage, Articular/pathology , Chondrocytes/pathology , Histone-Lysine N-Methyltransferase/physiology , Osteoarthritis/pathology , Animals , Cell Movement , Cells, Cultured , Chondrocytes/physiology , Endothelial Cells/physiology , Female , Histone-Lysine N-Methyltransferase/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Neovascularization, Pathologic/etiology , Osteoarthritis/etiology , Proto-Oncogene Proteins c-akt/physiology , STAT5 Transcription Factor/physiology
7.
STAR Protoc ; 1(3): 100195, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377089

ABSTRACT

Assessing IL-2-induced phosopho-STAT5 (pSTAT5) content can reveal the cytokine responsiveness of individual T cells. Identifying distinct T cell subsets by nuclear transcription factors, such as Foxp3, and concurrently quantifying intracellular pSTAT5, however, has been technically challenging. Conventional Foxp3 staining buffers quench pSTAT5 signals, while commonly used pSTAT5 staining protocols fail to detect Foxp3. The current protocol resolves these issues by describing a procedure to assess IL-2-induced pSTAT5 contents in Foxp3+ CD4 Treg cells using multiparameter flow cytometry. For complete details on the use and execution of this protocol, please refer to Waickman et al. (2020).


Subject(s)
Flow Cytometry/methods , STAT5 Transcription Factor/metabolism , Forkhead Transcription Factors/immunology , Humans , Interleukin-2/immunology , Phosphorylation , STAT5 Transcription Factor/physiology , Staining and Labeling/methods , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
8.
Front Endocrinol (Lausanne) ; 11: 572981, 2020.
Article in English | MEDLINE | ID: mdl-33133019

ABSTRACT

Glucocorticoids (GCs) and the glucocorticoid receptor (GR) are important regulators of development, inflammation, stress response and metabolism, demonstrated in various diseases including Addison's disease, Cushing's syndrome and by the many side effects of prolonged clinical administration of GCs. These conditions include severe metabolic challenges in key metabolic organs like the liver. In the liver, GR is known to regulate the transcription of key enzymes in glucose and lipid metabolism and contribute to the regulation of circadian-expressed genes. Insights to the modes of GR regulation and the underlying functional mechanisms are key for understanding diseases and for the development of improved clinical uses of GCs. The activity and function of GR is regulated at numerous levels including ligand availability, interaction with heat shock protein (HSP) complexes, expression of GR isoforms and posttranslational modifications. Moreover, recent genomics studies show functional interaction with multiple transcription factors (TF) and coregulators in complex transcriptional networks controlling cell type-specific gene expression by GCs. In this review we describe the different regulatory steps important for GR activity and discuss how different TF interaction partners of GR selectively control hepatic gene transcription and metabolism.


Subject(s)
Gene Regulatory Networks , Liver/metabolism , Receptors, Glucocorticoid/physiology , Transcription Factors/physiology , Active Transport, Cell Nucleus , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic/physiology , Forkhead Box Protein O1/physiology , Humans , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , STAT5 Transcription Factor/physiology , Signal Transduction/physiology
9.
Int J Mol Sci ; 21(18)2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32899608

ABSTRACT

Tolerogenic dendritic cells (toDCs) are crucial to controlling the development of autoreactive T cell responses and the prevention of autoimmunity. We have reported that NOD.CD11cStat5b-CA transgenic mice expressing a constitutively active (CA) form of Stat5b under the control of a CD11c promoter are protected from diabetes and that Stat5b-CA-expressing DCs are tolerogenic and halt ongoing diabetes in NOD mice. However, the molecular mechanisms by which Stat5b-CA modulates DC tolerogenic function are not fully understood. Here, we used bone marrow-derived DCs (BMDCs) from NOD.CD11cStat5b-CA transgenic mice (Stat5b-CA.BMDCs) and found that Stat5b-CA.BMDCs displayed high levels of MHC class II, CD80, CD86, PD-L1, and PD-L2 and produced elevated amounts of TGFß but low amounts of TNFα and IL-23. Stat5b-CA.BMDCs upregulated Irf4 and downregulated Irf8 genes and protein expression and promoted CD11c+CD11b+ DC2 subset differentiation. Interestingly, we found that the histone methyltransferase Ezh2 and Stat5b-CA bound gamma-interferon activated site (GAS) sequences in the Irf8 enhancer IRF8 transcription, whereas Stat5b but not Ezh2 bound GAS sequences in the Irf4 promoter to enhance IRF4 transcription. Injection of Stat5b-CA.BMDCs into prediabetic NOD mice halted progression of islet inflammation and protected against diabetes. Importantly, inhibition of Ezh2 in tolerogenic Stat5b-CA.BMDCs reduced their ability to prevent diabetes development in NOD recipient mice. Taken together, our data suggest that the active form of Stat5b induces tolerogenic DC function by modulating IRF4 and IRF8 expression through recruitment of Ezh2 and highlight the fundamental role of Ezh2 in Stat5b-mediated induction of tolerogenic DC function.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Interferon Regulatory Factors/metabolism , STAT5 Transcription Factor/metabolism , Adoptive Transfer , Animals , Autoimmunity/immunology , Bone Marrow/drug effects , CD11c Antigen/metabolism , Cell Differentiation/drug effects , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/metabolism , Enhancer of Zeste Homolog 2 Protein/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , STAT5 Transcription Factor/physiology , T-Lymphocytes, Regulatory/immunology
10.
Blood ; 136(6): 657-668, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32530039

ABSTRACT

Cytokine storm syndromes (CSS) are severe hyperinflammatory conditions characterized by excessive immune system activation leading to organ damage and death. Hemophagocytic lymphohistiocytosis (HLH), a disease often associated with inherited defects in cell-mediated cytotoxicity, serves as a prototypical CSS for which the 5-year survival is only 60%. Frontline therapy for HLH consists of the glucocorticoid dexamethasone (DEX) and the chemotherapeutic agent etoposide. Many patients, however, are refractory to this treatment or relapse after an initial response. Notably, many cytokines that are elevated in HLH activate the JAK/STAT pathway, and the JAK1/2 inhibitor ruxolitinib (RUX) has shown efficacy in murine HLH models and humans with refractory disease. We recently reported that cytokine-induced JAK/STAT signaling mediates DEX resistance in T cell acute lymphoblastic leukemia (T-ALL) cells, and that this could be effectively reversed by RUX. On the basis of these findings, we hypothesized that cytokine-mediated JAK/STAT signaling might similarly contribute to DEX resistance in HLH, and that RUX treatment would overcome this phenomenon. Using ex vivo assays, a murine model of HLH, and primary patient samples, we demonstrate that the hypercytokinemia of HLH reduces the apoptotic potential of CD8 T cells leading to relative DEX resistance. Upon exposure to RUX, this apoptotic potential is restored, thereby sensitizing CD8 T cells to DEX-induced apoptosis in vitro and significantly reducing tissue immunopathology and HLH disease manifestations in vivo. Our findings provide rationale for combining DEX and RUX to enhance the lymphotoxic effects of DEX and thus improve the outcomes for patients with HLH and related CSS.


Subject(s)
Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cytokine Release Syndrome/drug therapy , Dexamethasone/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Lymphohistiocytosis, Hemophagocytic/drug therapy , Pyrazoles/therapeutic use , Signal Transduction/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/physiopathology , Cytokines/physiology , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Disease Models, Animal , Drug Resistance/drug effects , Drug Therapy, Combination , Humans , Interleukin-2/pharmacology , Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/pharmacology , Janus Kinases , Lymphocytic Choriomeningitis/complications , Lymphocytic Choriomeningitis/physiopathology , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/enzymology , Lymphohistiocytosis, Hemophagocytic/immunology , Mice , Mice, Inbred C57BL , Nitriles , Perforin/deficiency , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidines , STAT5 Transcription Factor/physiology , Specific Pathogen-Free Organisms
11.
Mol Metab ; 40: 101026, 2020 10.
Article in English | MEDLINE | ID: mdl-32473405

ABSTRACT

OBJECTIVE: Increasing energy expenditure through activation of brown adipose tissue (BAT) thermogenesis is an attractive approach to counteract obesity. It is therefore essential to understand the molecular mechanisms that control BAT functions. Until now several members of the Janus kinase (JAK) - signal transducer and activator of transcription (STAT) pathway have been implicated as being relevant for BAT physiology. However, whether the STAT family member STAT5 is important for the thermogenic property of adipose tissues is unknown. Therefore, we have investigated the role of STAT5 in thermogenic fat in this paper. METHODS: We performed metabolic and molecular analyses using mice that harbor an adipocyte-specific deletion of Stat5a/b alleles. RESULTS: We found that STAT5 is necessary for acute cold-induced temperature maintenance and the induction of lipid mobilization in BAT following ß3-adrenergic stimulation. Moreover, mitochondrial respiration of primary differentiated brown adipocytes lacking STAT5 was diminished. Increased sensitivity to cold stress upon STAT5 deficiency was associated with reduced expression of thermogenic markers including uncoupling protein 1 (UCP1), while decreased stimulated lipolysis was linked to decreased protein kinase A (PKA) activity. Additionally, brown remodeling of white adipose tissue was diminished following chronic ß3-adrenergic stimulation, which was accompanied by a decrease in mitochondrial performance. CONCLUSION: We conclude that STAT5 is essential for the functionality and the ß-adrenergic responsiveness of thermogenic adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , STAT5 Transcription Factor/metabolism , Thermogenesis/physiology , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Cold-Shock Response/physiology , Energy Metabolism , Female , Lipid Metabolism/physiology , Lipids/physiology , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Obesity/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta/physiology , STAT5 Transcription Factor/physiology
12.
Diabetes ; 69(7): 1463-1475, 2020 07.
Article in English | MEDLINE | ID: mdl-32332156

ABSTRACT

Diabetes occurs due to a loss of functional ß-cells, resulting from ß-cell death and dysfunction. Lactogens protect rodent and human ß-cells in vitro and in vivo against triggers of ß-cell cytotoxicity relevant to diabetes, many of which converge onto a common pathway of endoplasmic reticulum (ER) stress. However, whether lactogens modulate the ER stress pathway is unknown. This study examines whether lactogens can protect ß-cells against ER stress and mitigate diabetes incidence in Akita (Ak) mice, a rodent model of ER stress-induced diabetes, akin to neonatal diabetes in humans. We show that lactogens protect INS-1 cells, primary rodent and human ß-cells in vitro against two distinct ER stressors, tunicamycin and thapsigargin, through activation of the JAK2/STAT5 pathway. Lactogens mitigate expression of proapoptotic molecules in the ER stress pathway that are induced by chronic ER stress in INS-1 cells and rodent islets. Transgenic expression of placental lactogen in ß-cells of Ak mice drastically reduces the severe hyperglycemia, diabetes incidence, hypoinsulinemia, ß-cell death, and loss of ß-cell mass observed in Ak littermates. These are the first studies in any cell type demonstrating that lactogens modulate the ER stress pathway, causing enhanced ß-cell survival and reduced diabetes incidence in the face of chronic ER stress.


Subject(s)
Diabetes Mellitus/prevention & control , Endoplasmic Reticulum Stress/drug effects , Insulin-Secreting Cells/drug effects , Placental Lactogen/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Female , Glucose/metabolism , Humans , Insulin/blood , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Janus Kinase 2/physiology , Male , Mice , Mice, Inbred C57BL , Prolactin/pharmacology , STAT5 Transcription Factor/physiology , Signal Transduction/drug effects
13.
Front Immunol ; 11: 307, 2020.
Article in English | MEDLINE | ID: mdl-32256487

ABSTRACT

SHIP-1 is an inositol phosphatase that hydrolyzes phosphatidylinositol 3-kinase (PI3K) products and negatively regulates protein kinase B (Akt) activity, thereby modulating a variety of cellular processes in mammals. However, the role of SHIP-1 in bacterial-induced sepsis is largely unknown. Here, we show that SHIP-1 regulates inflammatory responses during Gram-negative bacterium Pseudomonas aeruginosa infection. We found that infected-SHIP-1-/- mice exhibited decreased survival rates, increased inflammatory responses, and susceptibility owing to elevated expression of PI3K than wild-type (WT) mice. Inhibiting SHIP-1 via siRNA silencing resulted in lipid raft aggregates, aggravated oxidative damage, and bacterial burden in macrophages after PAO1 infection. Mechanistically, SHIP-1 deficiency augmented phosphorylation of PI3K and nuclear transcription of signal transducer and activator of transcription 5 (STAT5) to induce the expression of Trib1, which is critical for differentiation of M2 but not M1 macrophages. These findings reveal a previously unrecognized role of SHIP-1 in inflammatory responses and macrophage homeostasis during P. aeruginosa infection through a PI3K/Akt-STAT5-Trib1 axis.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Macrophages/physiology , Phagocytosis , Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , Pseudomonas Infections/immunology , STAT5 Transcription Factor/physiology , Animals , Cell Polarity , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/physiology
14.
Theranostics ; 10(7): 3351-3365, 2020.
Article in English | MEDLINE | ID: mdl-32194873

ABSTRACT

Rationale: Glioma is the most common primary malignant brain tumor in adults. Chemoresistance of temozolomide (TMZ), the first-line chemotherapeutic agent, is a major issue in the management of patients with glioma. Alterations of alpha thalassemia/mental retardation syndrome X-linked (ATRX) gene constitute one of the most prevalent genetic abnormalities in gliomas. Therefore, elucidation of the role of ATRX contributing to TMZ resistance in glioma is urgently needed. Methods: We performed the bioinformatics analysis of gene expression, and DNA methylation profiling, as well as RNA and ChIP-seq data sets. CRISPR-Cas9 gene editing system was used to achieve the ATRX knockout in TMZ resistant cells. In vitro and in vivo experiments were carried out to investigate the role of ATRX contributing to TMZ resistance in glioma. Results: We found that ATRX expression was upregulated via DNA demethylation mediated by STAT5b/TET2 complex and strengthened DNA damage repair by stabilizing PARP1 protein in TMZ resistant cells. ATRX elicited PARP1 stabilization by the down-regulating of FADD expression via the H3K27me3 enrichment, which was dependent on ATRX/EZH2 complex in TMZ resistant cells. Magnetic resonance imaging (MRI) revealed that the PARP inhibitor together with TMZ inhibited glioma growth in ATRX wild type TMZ resistant intracranial xenograft models. Conclusions: The present study further illustrated the novel mechanism of the ATRX/PARP1 axis contributing to TMZ resistance. Our results provided substantial new evidence that PARP inhibitor might be a potential adjuvant agent in overcoming ATRX mediated TMZ resistance in glioma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , DNA Methylation , Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/physiology , Fas-Associated Death Domain Protein/physiology , Gene Expression Regulation, Neoplastic/genetics , Glioma/drug therapy , Neoplasm Proteins/physiology , Poly (ADP-Ribose) Polymerase-1/physiology , Temozolomide/pharmacology , X-linked Nuclear Protein/physiology , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , CRISPR-Cas Systems , DNA Damage , DNA Repair , DNA, Neoplasm/genetics , DNA-Binding Proteins/physiology , Dioxygenases , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Editing , Gene Knockout Techniques , Glioma/genetics , Glioma/metabolism , Histone Code , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic , Proto-Oncogene Proteins/physiology , STAT5 Transcription Factor/physiology , Temozolomide/therapeutic use , Tumor Stem Cell Assay , Up-Regulation , X-linked Nuclear Protein/antagonists & inhibitors , X-linked Nuclear Protein/genetics , Xenograft Model Antitumor Assays
15.
J Neurosci ; 40(11): 2189-2199, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32019828

ABSTRACT

The interaction between the immune system and the nervous system has been at the center of multiple research studies in recent years. Whereas the role played by cytokines as neuronal mediators is no longer contested, the mechanisms by which cytokines modulate pain processing remain to be elucidated. In this study, we have analyzed the involvement of granulocyte-macrophage colony stimulating factor (GM-CSF) in nociceptor activation in male and female mice. Previous studies have suggested GM-CSF might directly activate neurons. However, here we established the absence of a functional GM-CSF receptor in murine nociceptors, and suggest an indirect mechanism of action, via immune cells. We report that GM-CSF applied directly to magnetically purified nociceptors does not induce any transcriptional changes in nociceptive genes. In contrast, conditioned medium from GM-CSF-treated murine macrophages was able to drive nociceptor transcription. We also found that conditioned medium from nociceptors treated with the well established pain mediator, nerve growth factor, could also modify macrophage gene transcription, providing further evidence for a bidirectional crosstalk.SIGNIFICANCE STATEMENT The interaction of the immune system and the nervous system is known to play an important role in the development and maintenance of chronic pain disorders. Elucidating the mechanisms of these interactions is an important step toward understanding, and therefore treating, chronic pain disorders. This study provides evidence for a two-way crosstalk between macrophages and nociceptors in the peripheral nervous system, which may contribute to the sensitization of nociceptors by cytokines in pain development.


Subject(s)
Chronic Pain/physiopathology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Nociceptors/drug effects , Animals , Calcium Signaling/drug effects , Cell Communication , Cells, Cultured , Chronic Pain/chemically induced , Culture Media, Conditioned/pharmacology , Female , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Inflammation/chemically induced , Inflammation/physiopathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factor/pharmacology , Nociceptors/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , STAT5 Transcription Factor/physiology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Transcription, Genetic/drug effects
16.
Mol Cell Endocrinol ; 498: 110574, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31494175

ABSTRACT

Several hypothalamic neuronal populations are directly responsive to growth hormone (GH) and central GH action regulates glucose and energy homeostasis. However, the potential role of GH signaling in proopiomelanocortin (POMC) neurons has not been studied yet. Thus, we investigated whether POMC neurons are responsive to GH and if ablation of GH receptor (GHR) or STAT5 in POMC cells leads to metabolic imbalances. Approximately 60% of POMC neurons of the arcuate nucleus exhibited STAT5 phosphorylation after intracerebroventricular GH injection. Ablation of GHR or STAT5 in POMC cells did not affect energy or glucose homeostasis. However, glucoprivic hyperphagia was blunted in male and female GHR knockout mice, and in male POMC-specific STAT5 knockout mice. Additionally, the absence of GHR in POMC neurons decreased glycemia during prolonged food restriction in male mice. Thus, GH action in POMC neurons regulates glucoprivic hyperphagia as well as blood glucose levels during prolonged food restriction.


Subject(s)
Carrier Proteins/physiology , Glucose/metabolism , Hyperphagia/pathology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , STAT5 Transcription Factor/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Female , Hyperphagia/metabolism , Male , Mice , Mice, Knockout
17.
Leukemia ; 33(8): 1881-1894, 2019 08.
Article in English | MEDLINE | ID: mdl-30816328

ABSTRACT

In more than 30% of B-cell precursor acute lymphoblastic leukaemia (B-ALL), chromosome 21 sequence is overrepresented through aneuploidy or structural rearrangements, exemplified by intrachromosomal amplification of chromosome 21 (iAMP21). Although frequent, the mechanisms by which these abnormalities promote B-ALL remain obscure. Intriguingly, we found copy number neutral loss of heterozygosity (CN-LOH) of 12q was recurrent in iAMP21-ALL, but never observed in B-ALL without some form of chromosome 21 gain. As a consequence of CN-LOH 12q, mutations or deletions of the adaptor protein, SH2B3, were converted to homozygosity. In patients without CN-LOH 12q, bi-allelic abnormalities of SH2B3 occurred, but only in iAMP21-ALL, giving an overall incidence of 18% in this sub-type. Review of published data confirmed a tight association between overrepresentation of chromosome 21 and both CN-LOH 12q and SH2B3 abnormalities in B-ALL. Despite relatively small patient numbers, preliminary analysis linked 12q abnormalities to poor outcome in iAMP21-ALL (p = 0.03). Homology modelling of a leukaemia-associated SH2 domain mutation and in vitro analysis of patient-derived xenograft cells implicated the JAK/STAT pathway as one likely target for SH2B3 tumour suppressor activity in iAMP21-ALL.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , Loss of Heterozygosity , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proteins/genetics , Adaptor Proteins, Signal Transducing , Humans , Interleukin-7/pharmacology , Intracellular Signaling Peptides and Proteins , Mutation , STAT5 Transcription Factor/physiology
18.
Leuk Res ; 79: 6-16, 2019 04.
Article in English | MEDLINE | ID: mdl-30784762

ABSTRACT

The BCR-ABL fusion gene (BCR-ABL) has different subtypes such as p210 and p190 with p190 appear to lead to a worse prognosis. To explore the mechanism of difference in pathogenesis and prognosis in different BCR-ABL subtype-related leukemia, expression profile microarray analysis was conducted between p190 and p210 patients and verified by RT-PCR. The p21-activated kinase (PAK1) gene was chosen and regulation of the PAK1-STAT5 biological axis and its influence on proliferation and apoptosis in leukemia cells were also analyzed. The results showed that PAK1 might be an important molecular mechanism of the pathogenic difference between different BCR-ABL subtypes. In P210 (+) chronic myelogenous leukemia (CML), down-regulated PAK1 gene expressions may lead to the suppression of cell proliferation and promotion of apoptosis through phosphorylation of STAT5, with a reverse effect in P190 (+) acute lymphoblastic leukemia(ALL), especially acute B lymphoblastic leukemia (B-ALL). Additionally, in P210 (+) CML, down-regulated PAK1 expression may enhance the effect of TKI, whereas the reverse is true in P190 (+) B-ALL, demonstrating that PAK1 might also be an important therapeutic target between different BCR-ABL subtypes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Fusion Proteins, bcr-abl/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma , STAT5 Transcription Factor/physiology , Tumor Suppressor Proteins/physiology , p21-Activated Kinases/physiology , Fusion Proteins, bcr-abl/classification , Gene Expression Profiling , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Microarray Analysis , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Signal Transduction/genetics , Tumor Cells, Cultured , p21-Activated Kinases/genetics
19.
Oncogene ; 38(24): 4657-4668, 2019 06.
Article in English | MEDLINE | ID: mdl-30783189

ABSTRACT

The signal transducer and activator of transcription (STAT) are transcription factors that work via JAK/STAT pathway regulating the expression of genes involved in cell survival, proliferation, differentiation, development, immune response, and, among other essential biological functions, hematopoiesis. JAK/STAT signaling is strictly regulated under normal physiological conditions. However, a large group of diverse diseases has been associated to an aberrant regulation of STAT factors. Erroneous modulation of the pathway leads to constitutive STAT activation, thereby driving proliferation, inflammation, and an uncontrolled immune response. Deregulated STAT5 activation has been found in the development of many hematopoietic tumors, including chronic and acute leukemias, polycythemia vera, and lymphoma. Mutations in the kinases that phosphorylate STAT5, and/or overexpression of the upstream receptor-associated tyrosine kinases have been suggested as the main drivers of constitutive STAT5 activation. Hyper-activated STAT5 leads to the aberrant expression of its target genes including antiapoptotic, proliferative, and pro-inflammatory genes, favouring tumorigenesis. In this review, we intent to discuss the biology of JAK/STAT pathway, with particular focus on STAT5 and its crucial role in the development and progression of hematologic malignancies. Furthermore, we provide a synopsis of potential therapeutic strategies based on STAT5 activity inhibition that may represent an excellent opportunity for drug development in oncohematology.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Development , Hematologic Neoplasms/drug therapy , Medical Oncology , STAT5 Transcription Factor/physiology , Tumor Suppressor Proteins/physiology , Animals , Antineoplastic Agents/chemistry , Drug Development/trends , Hematopoiesis/drug effects , Hematopoiesis/genetics , Humans , Janus Kinases/physiology , Medical Oncology/methods , Medical Oncology/trends , STAT Transcription Factors/physiology , Signal Transduction
20.
Cancer Cell ; 34(2): 271-285.e7, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30107177

ABSTRACT

The NUP214-ABL1 fusion is a constitutively activated tyrosine kinase that is significantly associated with overexpression of the TLX1 and TLX3 transcription factors in T cell acute lymphoblastic leukemia (T-ALL). Here we show that NUP214-ABL1 cooperates with TLX1 in driving T-ALL development using a transgenic mouse model and human T-ALL cells. Using integrated ChIP-sequencing, ATAC-sequencing, and RNA-sequencing data, we demonstrate that TLX1 and STAT5, the downstream effector of NUP214-ABL1, co-bind poised enhancer regions, and cooperatively activate the expression of key proto-oncogenes such as MYC and BCL2. Inhibition of STAT5, downregulation of TLX1 or MYC, or interference with enhancer function through BET-inhibitor treatment leads to reduction of target gene expression and induction of leukemia cell death.


Subject(s)
Enhancer Elements, Genetic , Homeodomain Proteins/physiology , Nuclear Pore Complex Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins/physiology , STAT5 Transcription Factor/physiology , Animals , Gene Fusion , Homeodomain Proteins/genetics , Humans , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/physiology , STAT5 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...