Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360911

ABSTRACT

Pyrimethamine (Pyri) is being used in combination with other medications to treat serious parasitic infections of the body, brain, or eye and to also reduce toxoplasmosis infection in the patients with HIV infection. Additionally, Pyri can display significant anti-cancer potential in different tumor models, but the possible mode of its actions remains unclear. Hence, in this study, the possible anti-tumoral impact of Pyri on human chronic myeloid leukemia (CML) was deciphered. Pyri inhibited cell growth in various types of tumor cells and exhibited a marked inhibitory action on CML cells. In addition to apoptosis, Pyri also triggered sustained autophagy. Targeted inhibition of autophagy sensitized the tumor cells to Pyri-induced apoptotic cell death. Moreover, the activation of signal transducer and activator of transcription 5 (STAT5) and its downstream target gene Bcl-2 was attenuated by Pyri. Accordingly, small interfering RNA (siRNA)-mediated STAT5 knockdown augmented Pyri-induced autophagy and apoptosis and promoted the suppressive action of Pyri on cell viability. Moreover, ectopic overexpression of Bcl-2 protected the cells from Pyri-mediated autophagy and apoptosis. Overall, the data indicated that the attenuation of STAT5-Bcl-2 cascade by Pyri can regulate its growth inhibitory properties by simultaneously targeting both apoptosis and autophagy cell death mechanism(s).


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Pyrimethamine/pharmacology , Apoptosis/genetics , Autophagy/genetics , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Beclin-1/deficiency , Beclin-1/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Gene Knockdown Techniques , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , THP-1 Cells , Transfection , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
2.
Mol Cell Endocrinol ; 519: 111063, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33122102

ABSTRACT

Growth hormone (GH) promotes postnatal human growth primarily by regulating insulin-like growth factor (IGF)-I production through activation of the GH receptor (GHR)-JAK2-signal transducer and activator of transcription (STAT)-5B signaling pathway. Inactivating STAT5B mutations, both autosomal recessive (AR) and dominant-negative (DN), are causal of a spectrum of GH insensitivity (GHI) syndrome, IGF-I deficiency and postnatal growth failure. Only AR STAT5B defects, however, confer additional characteristics of immune dysfunction which can manifest as chronic, potentially fatal, pulmonary disease. Somatic activating STAT5B and JAK2 mutations are associated with a plethora of immune abnormalities but appear not to impact human linear growth. In this review, molecular defects associated with STAT5B deficiency is highlighted and insights towards understanding human growth and immunity is emphasized.


Subject(s)
Growth Disorders/metabolism , Growth Hormone/metabolism , Janus Kinase 2/metabolism , STAT5 Transcription Factor/metabolism , Body Height , Growth Disorders/immunology , Humans , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics
3.
J Clin Immunol ; 41(1): 136-146, 2021 01.
Article in English | MEDLINE | ID: mdl-33090292

ABSTRACT

Patients with rare homozygous mutations in signal transducer and activator of transcription 5B (STAT5B) develop immunodeficiency resulting in chronic eczema, chronic infections, autoimmunity, and chronic lung disease. STAT5B-deficient patients are typically diagnosed in the teenage years, limiting our understanding of the development of associated phenotypic immune abnormalities. We report the first detailed chronological account of post-natal immune dysfunction associated with STAT5B deficiency in humans. Annual immunophenotyping of three siblings carrying a novel homozygous nonsense mutation in STAT5B was carried out over 4 years between the ages of 7 months to 8 years. All three siblings demonstrated consistent B cell hyperactivity including elevated IgE levels and autoantibody production, associated with diagnoses of atopy and autoimmunity. Total T cell levels in each sibling remained normal, with regulatory T cells decreasing in the oldest sibling. Interestingly, a skewing toward memory T cells was identified, with the greatest changes in CD8+ effector memory T cells. These results suggest an importance of STAT5B in B cell function and naïve versus memory T cell survival. Progressive dysregulation of FOXP3+ regulatory T cells and CD8+ memory T cell subsets reveal a crucial role of STAT5B in T cell homeostasis. The early diagnosis and focused immune evaluations of these three young STAT5B-deficient siblings support an important role of STAT5B in adaptive immune development and function.


Subject(s)
Adaptive Immunity/genetics , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , STAT5 Transcription Factor/deficiency , Siblings , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Cell Line , Consanguinity , Disease Management , Disease Susceptibility , Genetic Association Studies , Homozygote , Humans , Infant , Male , Memory T Cells/immunology , Memory T Cells/metabolism , Mutation , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Exome Sequencing
4.
A A Pract ; 13(7): 253-256, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31265442

ABSTRACT

This case demonstrates the airway management of a pediatric patient with short stature due to STAT5b deficiency, a rare genetic immunodeficiency associated with lung disease and endocrinopathy. The patient had recurrent pulmonary infections and pulmonary alveolar proteinosis (PAP) for which whole lung lavage (WLL) was recommended. Due to short stature and overall body habitus, the patient's airway would not accommodate a traditional double-lumen tube (DLT). Therefore, we placed 2 single-lumen breathing tubes: 1 endobronchial and 1 endotracheal, to mimic a DLT and facilitate WLL, demonstrating a viable option for lung isolation in the absence of purpose-built equipment.


Subject(s)
Bronchoalveolar Lavage/instrumentation , Lung/abnormalities , Pulmonary Alveolar Proteinosis/therapy , Adolescent , Dwarfism/complications , Dwarfism/metabolism , Dwarfism/pathology , Female , Humans , STAT5 Transcription Factor/deficiency , Treatment Outcome
5.
Skelet Muscle ; 9(1): 19, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31230596

ABSTRACT

BACKGROUND: Sexually dimorphic growth has been attributed to the growth hormone (GH)/insulin-like growth factor 1 (IGF1) axis, particularly GH-induced activation of the intracellular signal transducer and activator of transcription 5B (STAT5B), because deletion of STAT5B reduces body mass and the mass of skeletal muscles in male mice to that in female mice. However, it remains unclear why these effects are sex- and species-specific, because the loss of STAT5B retards growth in girls, but not in male mice. Our objectives were to determine whether sexually dimorphic growth of skeletal muscle persisted in STAT5B-/- mice and investigate the mechanisms by which STAT5B regulates sexually dimorphic growth. METHODS: Blood and skeletal muscle were harvested from male and female STAT5B-/- mice and their wild-type littermates from the onset of puberty to adulthood. RESULTS: Growth of the skeleton and skeletal muscles was retarded in both sexes of STAT5B-/- mice, but more so in males. Although reduced, sexually dimorphic growth of skeletal muscle persisted in STAT5B-/- mice with an oxidative shift in the composition of myofibres in both sexes. Concentrations of IGF1 in blood and skeletal muscle were reduced in male STAT5B-/- mice at all ages, but only in female STAT5B-/- mice at the onset of puberty. Expression of androgen receptor (AR) and oestrogen receptor alpha (ERα) mRNA and protein was reduced in skeletal muscles of male and female STAT5B-/- mice, respectively. Loss of STAT5B abolished the sexually dimorphic expression of myostatin protein and Igf1, Ar, Erα, suppressor of cytokine signalling 2 (Socs2), and cytokine-inducible SH2-containing protein (Cis) mRNA in skeletal muscle. CONCLUSIONS: STAT5B appears to mediate GH signalling in skeletal muscles of male mice at all ages, but only until puberty in female mice. STAT5B also appears to mediate the actions of androgens and oestrogens in both male and female mice, but sexually dimorphic growth persists in STAT5B-/- mice.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , STAT5 Transcription Factor/metabolism , Age Factors , Animals , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Growth Hormone/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development/genetics , Myostatin/genetics , Myostatin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Sex Characteristics , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
6.
FASEB J ; 33(5): 6412-6430, 2019 05.
Article in English | MEDLINE | ID: mdl-30779881

ABSTRACT

Growth hormone (GH) has an important function as an insulin antagonist with elevated insulin sensitivity evident in humans and mice lacking a functional GH receptor (GHR). We sought the molecular basis for this sensitivity by utilizing a panel of mice possessing specific deletions of GHR signaling pathways. Metabolic clamps and glucose homeostasis tests were undertaken in these obese adult C57BL/6 male mice, which indicated impaired hepatic gluconeogenesis. Insulin sensitivity and glucose disappearance rate were enhanced in muscle and adipose of mice lacking the ability to activate the signal transducer and activator of transcription (STAT)5 via the GHR (Ghr-391-/-) as for GHR-null (GHR-/-) mice. These changes were associated with a striking inhibition of hepatic glucose output associated with altered glycogen metabolism and elevated hepatic glycogen content during unfed state. The enhanced hepatic insulin sensitivity was associated with increased insulin receptor ß and insulin receptor substrate 1 activation along with activated downstream protein kinase B signaling cascades. Although phosphoenolpyruvate carboxykinase (Pck)-1 expression was unchanged, its inhibitory acetylation was elevated because of decreased sirtuin-2 expression, thereby promoting loss of PCK1. Loss of STAT5 signaling to defined chromatin immunoprecipitation targets would further increase lipogenesis, supporting hepatosteatosis while lowering glucose output. Finally, up-regulation of IL-15 expression in muscle, with increased secretion of adiponectin and fibroblast growth factor 1 from adipose tissue, is expected to promote insulin sensitivity.-Chhabra, Y., Nelson, C. N., Plescher, M., Barclay, J. L., Smith, A. G., Andrikopoulos, S., Mangiafico, S., Waxman, D. J., Brooks, A. J., Waters, M. J. Loss of growth hormone-mediated signal transducer and activator of transcription 5 (STAT5) signaling in mice results in insulin sensitivity with obesity.


Subject(s)
Carrier Proteins , Fatty Liver , Insulin Resistance/genetics , Liver , Obesity , STAT5 Transcription Factor/deficiency , Signal Transduction/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Glucose/genetics , Glucose/metabolism , Glycogen/genetics , Glycogen/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , STAT5 Transcription Factor/metabolism
8.
Endocrinology ; 159(3): 1377-1392, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29346554

ABSTRACT

Sex-specific temporal patterns of pituitary growth hormone (GH) secretion determine the sex-biased transcription of hundreds of genes in the liver and impart important sex differences in liver physiology, metabolism, and disease. Sex differences in hepatic gene expression vary widely, ranging from less than twofold to >1000-fold in the mouse. Here, we use small RNA sequencing to discover 24 sex-biased mouse liver microRNAs (miRNAs), and then investigate the roles of two of these miRNAs in GH-regulated liver sex differences. Studies in prepubertal and young adult mice, and in mice in which pituitary hormones are ablated or where sex-specific hepatic GH signaling is dysregulated, demonstrated that the male-biased miR-1948 and the female-biased miR-802 are both regulated by sex-specific pituitary GH secretory patterns, acquire sex specificity at puberty, and are dependent on the GH-activated transcription factor STAT5 for their sex-specific expression. Both miRNAs are within genomic regions characterized by sex-biased chromatin accessibility. miR-1948, an uncharacterized miRNA, has essential features for correct Drosha/Dicer processing, generates a bona fide mature miRNA with strong strand bias for the 5p arm, and is bound by Argonaute in liver tissue, as is miR-802. In vivo studies using inhibitory locked nucleic acid sequences revealed that miR-1948-5p preferentially represses female-biased messenger RNAs (mRNAs) and induces male-biased mRNAs in male liver; conversely, miR-802-5p preferentially represses male-biased mRNAs and increases levels of female-biased mRNAs in female liver. Cytochrome P450 mRNAs were strongly enriched as targets of both miRNAs. Thus, miR-1948-5p and miR-802-5p are functional components of the GH regulatory network that shapes sex-differential gene expression in mouse liver.


Subject(s)
Growth Hormone/pharmacology , Liver/metabolism , MicroRNAs/physiology , Sex Characteristics , Animals , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Growth Hormone/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Knockout , MicroRNAs/genetics , RNA, Messenger , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/physiology
9.
Cardiovasc Res ; 114(5): 679-689, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29365089

ABSTRACT

Aims: To study the protective effects of late remote ischaemic preconditioning (RIPC) against myocardial ischaemia/reperfusion (I/R) injury and determine whether Stat5 is involved in this protection by using cardiomyocyte-specific Stat5 knockout mice (Stat5-cKO). Methods and results: Mice were exposed to lower limb RIPC or sham ischaemia. After 24 h, the left anterior descending artery (LAD) was ligated for 30 min, then reperfused for 180 min. The myocardial infarct size (IS), apoptotic rate of cardiomyocytes, and serum myocardial enzymes were measured to evaluate for cardioprotective effects. Heart tissues were harvested to determine the cardiomyocytes' anti-apoptotic and survival signaling. When compared with the Stat5fl/fl mice without RIPC, Stat5fl/fl mice with RIPC (Stat5fl/fl+RIPC + I/R) displayed a decreased myocardial IS/LV (16 ± 1.5 vs. 30.1 ± 3.1%, P < 0.01; IS/ area at risk (AAR), 42.2 ± 3.5 vs. 69.2 ± 4.9%, P < 0.01), a reduced cardiomyocyte apoptotic rate (2.1 ± 0.37 vs. 5.5 ± 0.53%, P < 0.01), and lower creatine kinase (CK), lactate dehydrogenase (LDH), and creatine kinase-MB (CK-MB) levels. To the contrary, the Stat5-cKO mice (Stat5fl/fl; Tnnt2Cremice with Doxycycline treatment for 7 days) did not exhibit any effect of RIPC-induced cardioprotection. Activation of STAT5 protein was significantly higher in the Stat5fl/fl+RIPC + I/R group than in the Stat5fl/fl+I/R group, while there was no significant difference between the Stat5-cKO + RIPC + I/R and the Stat5-cKO + I/R group. Further analyses with heart tissues detected decreased protein expressions of cytochrome c (Cyt c) and cleaved Caspase-3 in the Stat5fl/fl+RIPC + I/R mice, along with increased anti-apoptotic molecules, including B-cell lymphoma-extra large (Bcl-xL) and B-cell lymphoma-2 (Bcl-2); such changes were not noted in the Stat5-cKO + RIPC + I/R mice. Additionally, RIPC increased cardiac hypoxia inducible factor-1 (HIF-1α) and interleukin-10 (IL10) protein levels and caused activation of AKT, phosphatidylinositol 3 kinase (PI3K), and vascular endothelial growth factor in the heart of the Stat5fl/fl mice. However, these changes were completely inhibited by the absence of Stat5. Conclusions: These results suggest that RIPC-induced late cardioprotection against myocardial I/R injury is Stat5-dependent and is correlated with the activation of anti-apoptotic signaling and cardiomyocyte-survival signaling.


Subject(s)
Apoptosis , Femoral Artery/surgery , Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , STAT5 Transcription Factor/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Creatine Kinase, MB Form/blood , Cytochromes c/metabolism , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-10/metabolism , L-Lactate Dehydrogenase/blood , Ligation , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
10.
PLoS One ; 12(8): e0181948, 2017.
Article in English | MEDLINE | ID: mdl-28806763

ABSTRACT

Acupuncture is reported to be effective in treating obesity related illnesses, but its mechanism is still unclear. To investigate this mechanism we applied electro-acupuncture (EA) in a mouse model of obesity and used RNA-seq to identify molecular consequences. Deletion of the transcription factor STAT5 from neurons (Stat5NKO) led to obesity. Acupuncture, in turn, reduced body weight and the ratio of epididymal white adipose tissue (Epi-WAT) to body weight, and it also decreased plasma concentrations of glucose, triglyceride, and cholesterol. In addition, EA increased cold endurance of Stat5NKO obese mice. EA reversed altered gene expressions in the hypothalamus and Epi-WAT, especially in the hypothalamus in Stat5NKO obese mice. This study provides, for the first time, insight into genomic networks of obesity and their modulation by electro-acupuncture, which in turn reveals potential mechanisms that explain acupuncture-induced weight-loss.


Subject(s)
Electroacupuncture , Genome , STAT5 Transcription Factor/deficiency , Adaptation, Physiological , Adipose Tissue, White/metabolism , Animals , Cold Temperature , Down-Regulation/genetics , Energy Metabolism/genetics , Epididymis/metabolism , Gene Expression Profiling , Hypothalamus/metabolism , Male , Mice, Knockout , Mice, Obese , Molecular Sequence Annotation , Phenotype , Reproducibility of Results , STAT5 Transcription Factor/metabolism , Sequence Analysis, RNA , Up-Regulation/genetics
11.
Eur J Nutr ; 56(2): 569-579, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26582580

ABSTRACT

PURPOSE: Growth hormone (GH) controls liver metabolism through the transcription factor signal transducer and activator of transcription 5 (STAT5). However, it remains to be fully understood to what extent other GH/STAT5 target tissues contribute to lipid and glucose metabolism. This question was now addressed in muscle-specific STAT5 knockout (STAT5 MKO) mice model. METHODS: Changes in lipid and glucose metabolism were investigated at physiological and molecular levels in muscle and liver tissues of STAT5 MKO mice under normal diet or high-fat diet (HFD) conditions. RESULTS: STAT5 MKO mice exhibited an increased intramyocellular lipid (IMCL) accumulation in the quadriceps in HFD group. Decreased lipolytic hormone-sensitive lipase transcript levels may contribute to the increased IMCL accumulation in STAT5 MKO mice. STAT5 MKO induced hepatic lipid accumulation without deregulated STAT5 signaling. The upregulation of lipoprotein lipase and Cd36 mRNA levels, an increased trend of very low-density lipoprotein receptor mRNA levels, and elevated circulating concentrations of free fatty acid, triglyceride, and total cholesterol support the increase in hepatic lipid accumulation. CONCLUSIONS: STAT5 MKO in conjunction with a HFD deregulated both lipid and glucose metabolism in skeletal muscle, and this deregulation induced hepatic fat accumulation via increased circulating glucose, FFA, and TG concentrations. Our study emphasizes that muscle-specific STAT5 signaling is important for balancing lipid and glucose metabolism in peripheral tissues, including muscle and liver and that the deregulation of local STAT5 signaling augments HFD-induced lipid accumulation in both muscle and liver.


Subject(s)
Diet, High-Fat , Lipid Metabolism/physiology , Liver/metabolism , Muscle, Skeletal/metabolism , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/physiology , Animals , CD36 Antigens/genetics , Glucose/metabolism , Lipoprotein Lipase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , Receptors, LDL/genetics , Signal Transduction/physiology , Up-Regulation/physiology
12.
Cancer Discov ; 6(4): 414-29, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26873347

ABSTRACT

UNLABELLED: Natural killer (NK) cells are tightly regulated by the JAK-STAT signaling pathway and cannot survive in the absence of STAT5. We now report that STAT5-deficient NK cells can be rescued by overexpression of BCL2. Our experiments define STAT5 as a master regulator of NK-cell proliferation and lytic functions. Although NK cells are generally responsible for killing tumor cells, the rescued STAT5-deficient NK cells promote tumor formation by producing enhanced levels of the angiogenic factor VEGFA. The importance of VEGFA produced by NK cells was verified by experiments with a conditional knockout of VEGFA in NK cells. We show that STAT5 normally represses the transcription of VEGFA in NK cells, in both mice and humans. These findings reveal that STAT5-directed therapies may have negative effects: In addition to impairing NK-cell-mediated tumor surveillance, they may even promote tumor growth by enhancing angiogenesis. SIGNIFICANCE: The importance of the immune system in effective cancer treatment is widely recognized. We show that the new signal interceptors targeting the JAK-STAT5 pathway may have dangerous side effects that must be taken into account in clinical trials: inhibiting JAK-STAT5 has the potential to promote tumor growth by enhancing NK-cell-mediated angiogenesis.


Subject(s)
Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Immunologic Surveillance , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neoplasms/immunology , Neoplasms/metabolism , STAT5 Transcription Factor/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival/genetics , Cytotoxicity, Immunologic , Disease Models, Animal , Gene Expression , Gene Knockout Techniques , Humans , Killer Cells, Natural/cytology , Lymphocyte Activation , Mice , Mice, Transgenic , Neoplasms/mortality , Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT5 Transcription Factor/deficiency , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
13.
Oncotarget ; 6(30): 28961-72, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26338970

ABSTRACT

Despite being an attractive molecular target for both lymphoid and myeloid leukemias characterized by activated tyrosine kinases, the molecular and physiological consequences of reduced signal transducer and activator of transcription-5 (Stat5) during leukemogenesis are not well known. Stat5 is a critical regulator of mouse hematopoietic stem cell (HSC) self-renewal and is essential for normal lymphocyte development. We report that pan-hematopoietic deletion in viable adult Vav1-Cre conditional knockout mice as well as Stat5ab(null/null) fetal liver transplant chimeras generated HSCs with reduced expression of quiescence regulating genes (Tie2, Mpl, Slamf1, Spi1, Cited2) and increased expression of B-cell development genes (Satb1, Dntt, Btla, Flk2). Using a classical murine B-cell acute lymphoblastic leukemia (B-ALL) model, we demonstrate that these HSCs were also poised to produce a burst of B-cell precursors upon expression of Bcl-2 combined with oncogenic Myc. This strong selective advantage for leukemic transformation in the background of Stat5 deficient hematopoiesis was permissive for faster initiation of Myc-induced transformation to B-ALL. However, once established, the B-ALL progression in secondary transplant recipients was Stat5-independent. Overall, these studies suggest that Stat5 can play multiple important roles that not only preserve the HSC compartment but can limit accumulation of potential pre-leukemic lymphoid populations.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Leukemia, B-Cell/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , STAT5 Transcription Factor/deficiency , Animals , Bone Marrow Transplantation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Progression , Gene Expression Regulation, Leukemic , Genetic Predisposition to Disease , Hematopoietic Stem Cells/pathology , Leukemia, B-Cell/genetics , Leukemia, B-Cell/pathology , Liver/embryology , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , STAT5 Transcription Factor/genetics , Signal Transduction , Time Factors
14.
J Clin Immunol ; 35(3): 264-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25753012

ABSTRACT

UNLABELLED: The signal transducer and activator of transcription (STAT) family of proteins regulate gene transcription in response to a variety of cytokines. STAT5B, in particular, plays an important role in T cells, where it is a key mediator of interleukin-2 (IL-2) induced responses. STAT5B deficiency causes a rare autosomal recessive disorder reported in only a handful of individuals. There are currently ten published cases of STAT5B deficiency, four of which are Argentinians. AIM: This is a report of more than 10 years follow up of the clinical and immunological features of three Argentinian STAT5B deficient patients. CONCLUSION: More than a decade of follow-up demonstrates that STAT5B deficiency is associated with various clinical pathologies that cause significant morbidity. Early diagnosis is critical for the prevention and improvement of clinical outcomes for STAT5B deficient patients.


Subject(s)
Immunologic Deficiency Syndromes/immunology , STAT5 Transcription Factor/deficiency , Adult , Argentina , Autoantibodies/blood , B-Lymphocytes/immunology , Female , Humans , Immunoglobulins/blood , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/genetics , Lymphocyte Count , Mutation , STAT5 Transcription Factor/genetics , T-Lymphocytes/immunology , Young Adult
15.
Mol Cell Biol ; 34(7): 1363-77, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469394

ABSTRACT

Stat5 (signal transducer and activator of transcription 5) is an essential mediator of cytokine receptor signaling and plays important roles in the proliferation of alveolar progenitors and the survival of functionally differentiated epithelial cells in the mammary gland. A deregulated expression and activation of Stat5 leads to precocious alveolar development in the absence of pregnancy hormones, impaired mammary gland remodeling following the cessation of lactation, and mammary tumor formation. We reported previously that Stat5 induces the transcription of the Akt1 gene from a novel promoter. In this report, we provide experimental evidence that Akt1 is an essential mediator for the biological function of Stat5 as a survival factor. Additionally, Stat5 controls the expression of the regulatory and catalytic subunits of the phosphatidylinositol 3-kinase (PI3K) (p85α and p110α), thereby greatly augmenting signaling through the prosurvival PI3K/Akt pathway. In agreement with this model, we observed that the constitutive activation of Stat5 cooperates with the loss of function of the tumor suppressor PTEN by accelerating the formation of preneoplastic lesions and mammary tumors. The mammary gland-specific ablation of Stat5 is sufficient to prevent mammary carcinogenesis in a genuine mouse model for Cowden syndrome. Therefore, targeting the Jak2/Stat5 pathway might be a suitable strategy to prevent breast cancer in patients that carry a mutant PTEN allele.


Subject(s)
Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , Animals , Cell Line , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Female , Gene Knockout Techniques , Hamartoma Syndrome, Multiple/etiology , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/metabolism , Humans , Janus Kinase 2/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Precancerous Conditions/etiology , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Signal Transduction
16.
Arq Bras Endocrinol Metabol ; 57(5): 333-8, 2013 Jul.
Article in Portuguese | MEDLINE | ID: mdl-23896798

ABSTRACT

A new presentation of growth hormone insensitivity (GHI) caused by homozygous mutations in STAT5B (signal transducer and activator of transcription 5B) gene has been characterized in the last years. Its particularity is the association with severe immune dysfunction, especially with lymphocytic interstitial pneumonitis. This may mislead physicians into considering short stature as secondary to chronic immunological disease and consequently into underdiagnosing this form of GHI. The objective of this review is to propagate current knowledge about this rare pathology, facilitating the diagnosis of patients with GHI due to STAT5B mutations in endocrinology and other specialties clinics.


Subject(s)
Human Growth Hormone/genetics , Immune System Diseases/genetics , Laron Syndrome/genetics , Mutation , Rare Diseases/genetics , STAT5 Transcription Factor/deficiency , Humans , Immune System Diseases/immunology , Interleukins/metabolism , Laron Syndrome/therapy , Rare Diseases/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction
17.
J Immunol ; 190(7): 3390-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23440411

ABSTRACT

IL-2 signals during the primary response to infection are essential in shaping CD8(+) T cell fate decisions. How CD8(+) T cells integrate IL-2 signals in the development of functional memory is not well understood. Because IL-2 induces potent activation of the STAT5 transcription factor, we tested the role of STAT5 in CD8(+) memory T cell differentiation and function using a model system in which STAT5 activity is inducibly abrogated upon CD8(+) T cell activation. We report that STAT5 activity is broadly important for the expansion and effector function of all effector CTL subsets. After pathogen clearance, STAT5 was required for the survival of effector phenotype memory CTLs during the contraction phase. However, despite its role in supporting full primary CD8(+) T cell expansion, and unlike IL-2, STAT5 activity is not required for the development of memory CD8(+) T cells capable of robust secondary expansion upon rechallenge. Our findings highlight differential requirements for survival signals between primary and secondary effector CTL, and demonstrate that IL-2-dependent programming of memory CD8(+) T cells capable of secondary expansion and secondary effector differentiation is largely STAT5 independent.


Subject(s)
Immunologic Memory , STAT5 Transcription Factor/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Arenaviridae Infections/genetics , Arenaviridae Infections/immunology , Arenaviridae Infections/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Gene Expression , Immunologic Memory/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Knockout , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes, Cytotoxic/cytology
18.
Curr Opin Rheumatol ; 24(5): 515-21, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22820513

ABSTRACT

PURPOSE OF REVIEW: To provide an overview of the mechanisms of autoimmunity associated with primary immunodeficiencies. RECENT FINDINGS: Over the past several years, new concepts of the relationship between primary immunodeficiencies and autoimmunity have developed that promise to illuminate the mechanisms by which alterations in the same gene may alternately, or sometimes concomitantly, lead to increased susceptibility to infection and loss of self-tolerance. A common pathway in the process leading to autoimmunity involves gene defects that permit effector T-cell development in the absence of sufficient regulatory T-cell function. Conversely, gene defects that primarily lead to autoimmunity may impair host defense by neutralizing key elements of immunity. The production of neutralizing antibodies against cytokines comprises a newly recognized mechanism in which autoimmunity may lead to immunodeficiency. SUMMARY: Autoimmunity has long been known to be a part of the presenting symptoms and clinical course of many primary immunodeficiencies. This review will provide an overview of the new concepts regarding the complex relationship between the genetic immune deficiencies and autoimmunity. The mechanisms by which immunodeficiency may lead to autoimmunity or, in some instances, by which autoimmunity produces immunodeficiency can provide important insights into the underlying pathogenic processes and ultimately better diagnosis and treatment for the patient.


Subject(s)
Autoimmunity , Immunologic Deficiency Syndromes/immunology , Autoantibodies/metabolism , Autoimmunity/genetics , Cytokines/antagonists & inhibitors , Cytokines/immunology , DiGeorge Syndrome/immunology , Forkhead Transcription Factors/deficiency , Humans , Immunologic Deficiency Syndromes/genetics , Interleukin-2 Receptor alpha Subunit/deficiency , Interleukin-2 Receptor alpha Subunit/immunology , Mutation , STAT5 Transcription Factor/deficiency , Syndrome , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Wiskott-Aldrich Syndrome/immunology
19.
J Biol Chem ; 287(14): 11234-9, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22318729

ABSTRACT

Recent work has identified a new subset of CD4(+) T cells named as Tfh cells that are localized in germinal centers and critical in germinal center formation. Tfh cell differentiation is regulated by IL-6 and IL-21, possibly via STAT3 factor, and B cell lymphoma 6 (Bcl6) is specifically expressed in Tfh cells and required for their lineage specification. In the current study, we characterized the role of STAT5 in Tfh cell development. We found that a constitutively active form of STAT5 effectively inhibited Tfh differentiation by suppressing the expression of Tfh-associated factors (CXC motif) receptor 5 (CXCR5), musculoaponeurotic fibrosarcoma (c-Maf), Bcl6, basic leucine zipper transcription factor ATF-like (Batf), and IL-21, and STAT5 deficiency greatly enhanced Tfh gene expression. Importantly, STAT5 regulated the expression of Tfh cell suppressor factor B lymphocyte-induced maturation protein 1 (Blimp-1); STAT5 deficiency impaired Blimp-1 expression and resulted in elevated expression of Tfh-specific genes. Similarly, inhibition of IL-2 potentiated Tfh generation, associated with dampened Blimp-1 expression; Blimp-1 overexpression inhibited Tfh gene expression in Stat5-deficient T cells, suggesting that the IL-2/STAT5 axis functions to regulate Blimp-1 expression. In vivo, deletion of STAT5 in CD4(+) T cells resulted in enhanced development of Tfh cells and germinal center B cells and led to an impairment of B cell tolerance in a well defined mouse tolerance model. Taken together, this study demonstrates that STAT5 controls Tfh differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , STAT5 Transcription Factor/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Mice , Positive Regulatory Domain I-Binding Factor 1 , STAT5 Transcription Factor/deficiency , Transcription Factors/genetics , Up-Regulation
20.
Cell Mol Life Sci ; 69(1): 49-58, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21984607

ABSTRACT

We summarize the clinical presentation and molecular basis of a unique group of congenital immunodeficiency disorders in which defects in immune tolerance mechanisms result in severe autoimmunity. Patients with severe, familial forms of multi-organ autoimmunity have been recognized and clinically described for more than 40 years (Clin Exp Immunol 1: 119-128, 1966; Clin Exp Immunol 2: 19-30, 1967). Some are characterized primarily by autoimmunity and others by autoimmunity combined with susceptibility to specific infectious organisms. The first mechanistic understanding of these disorders began to emerge approximately 10 years ago with the initial identification of causative genes. As a result, our understanding of how immune tolerance is established and maintained in humans has expanded dramatically. Data generated over the last 3-4 years including identification of additional gene defects and functional characterization of each identified gene product in human and animal models have added clarity. This, in turn, has improved our ability to diagnose and effectively treat these severe, life-threatening disorders. Inherited disorders characterized by immune dysregulation have dramatically expanded our understanding of immune tolerance mechanisms in humans. Recognition and diagnosis of these disorders in the clinic allows timely initiation of life-saving therapies that may prevent death or irreversible damage to vital organs.


Subject(s)
Autoimmunity , Genetic Diseases, X-Linked , Immunologic Deficiency Syndromes , Polyendocrinopathies, Autoimmune , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Diagnostic Techniques and Procedures , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/therapy , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/therapy , Interleukin-10/deficiency , Interleukin-2 Receptor alpha Subunit/deficiency , Male , Mice , Polyendocrinopathies, Autoimmune/diagnosis , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/therapy , Receptors, Interleukin-10/deficiency , Repressor Proteins/deficiency , STAT5 Transcription Factor/deficiency , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Ubiquitin-Protein Ligases/deficiency , AIRE Protein
SELECTION OF CITATIONS
SEARCH DETAIL