Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507993

ABSTRACT

Interleukin 6 (IL-6) is known to regulate the CD4 T cell function by inducing gene expression of a number of cytokines through activation of Stat3 transcription factor. Here, we reveal that IL-6 strengthens the mechanics of CD4 T cells. The presence of IL-6 during activation of mouse and human CD4 T cells enhances their motility (random walk and exploratory spread), resulting in an increase in travel distance and higher velocity. This is an intrinsic effect of IL-6 on CD4 T-cell fitness that involves an increase in mitochondrial Ca2+ Although Stat3 transcriptional activity is dispensable for this process, IL-6 uses mitochondrial Stat3 to enhance mitochondrial Ca2+-mediated motility of CD4 T cells. Thus, through a noncanonical pathway, IL-6 can improve competitive fitness of CD4 T cells by facilitating cell motility. These results could lead to alternative therapeutic strategies for inflammatory diseases in which IL-6 plays a pathogenic role.


Subject(s)
Cell Movement/physiology , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cytokines/metabolism , Female , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects
2.
Immunity ; 36(4): 553-5, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22520849

ABSTRACT

STAT proteins bind DNA as dimers to regulate gene expression. Cooperative recruitment of pairs of dimers (tetramers) to adjacent DNA sites has also been documented. In this issue, Lin et al. (2012) examined tetramer function in vivo and showed that STAT5 tetramers function primarily as transcriptional activators.

3.
Immunity ; 36(2): 166-74, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22365663

ABSTRACT

Interferons (IFNs) were discovered as cytokines induced during and protecting from viral infection. They have been documented to play essential roles in numerous physiological processes beyond antiviral and antimicrobial defense, including immunomodulation, cell cycle regulation, cell survival, and cell differentiation. Recent data have also uncovered a potentially darker side to IFN, including roles in inflammatory diseases, such as autoimmunity and diabetes. IFN can have effects in the absence of acute infection, highlighting a physiologic role for constitutive IFN. Type I IFNs are constitutively produced at vanishingly low quantities and yet exert profound effects, mediated in part through modulation of signaling intermediates required for responses to diverse cytokines. We review evidence for a yin-yang of IFN function through its role in modulating crosstalk between multiple cytokines by both feedforward and feedback regulation of common signaling intermediates and postulate a homeostatic role for IFN through tonic signaling in the absence of acute infection.


Subject(s)
Interferon Type I/physiology , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Antiviral Agents/immunology , Antiviral Agents/metabolism , Autoimmunity , Cytokines/physiology , Feedback, Physiological , Homeostasis , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/metabolism
4.
Immunity ; 34(1): 39-49, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21215659

ABSTRACT

Signal transducer and activator of transcription (STAT) family members direct the differentiation of T helper cells, with specific STAT proteins promoting distinct effector subsets. STAT6 is required for the development of T helper 2 (Th2) cells, whereas STAT3 promotes differentiation of Th17 and follicular helper T cell subsets. We demonstrated that STAT3 was also activated during Th2 cell development and was required for the expression of Th2 cell-associated cytokines and transcription factors. STAT3 bound directly to Th2 cell-associated gene loci and was required for the ability of STAT6 to bind target genes. In vivo, STAT3 deficiency in T cells eliminated the allergic inflammation in mice sensitized and challenged with ovalbumin or transgenic for constitutively active STAT6. Thus, STAT3 cooperates with STAT6 in promoting Th2 cell development. These results demonstrate that differentiating T helper cells integrate multiple STAT protein signals during Th2 cell development.


Subject(s)
Hypersensitivity/immunology , STAT3 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Animals , Cell Differentiation/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Hypersensitivity/genetics , Hypersensitivity/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding/immunology , Receptor Cross-Talk/immunology , STAT3 Transcription Factor/genetics , STAT6 Transcription Factor/genetics , Signal Transduction/immunology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Transcriptional Activation
5.
Proc Natl Acad Sci U S A ; 114(25): E4924-E4933, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28584133

ABSTRACT

In addition to its canonical role in nuclear transcription, signal transducer and activator of transcription 3 (STAT3) is emerging as an important regulator of mitochondrial function. Here, we demonstrate that a novel inhibitor that binds with high affinity to the STAT3 SH2 domain triggers a complex cascade of events initiated by interference with mitochondrial STAT3 (mSTAT3). The mSTAT3-drug interaction leads to mitochondrial dysfunction, accumulation of proteotoxic STAT3 aggregates, and cell death. The cytotoxic effects depend directly on the drug's ability to interfere with mSTAT3 and mitochondrial function, as demonstrated by site-directed mutagenesis and use of STAT3 knockout and mitochondria-depleted cells. Importantly, the lethal consequences of mSTAT3 inhibition are enhanced by glucose starvation and by increased reliance of cancer cells and tumor-initiating cells on mitochondria, resulting in potent activity in cell cultures and tumor xenografts in mice. These findings can be exploited for eliciting synthetic lethality in metabolically stressed cancer cells using high-affinity STAT3 inhibitors. Thus, this study provides insights on the role of mSTAT3 in cancer cells and a conceptual framework for developing more effective cancer therapies.


Subject(s)
Mitochondria/genetics , Neoplasms/genetics , STAT3 Transcription Factor/genetics , Synthetic Lethal Mutations/genetics , src Homology Domains/genetics , Animals , Cell Death/genetics , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude
6.
Immunity ; 33(1): 1-2, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20643331

ABSTRACT

Induction of gene expression involves deployment of transcription factors. In this issue of Immunity, Farlik et al. (2010) provide a view in which cooperation between transcription factors NF-kappaB and ISGF3 divides the task of transcription by recruiting and activating distinct components of the transcriptional machinery.

7.
PLoS Pathog ; 12(3): e1005489, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26939124

ABSTRACT

Sensing invading pathogens early in infection is critical for establishing host defense. Two cytosolic RIG-like RNA helicases, RIG-I and MDA5, are key to type I interferon (IFN) induction in response to viral infection. Mounting evidence suggests that another viral RNA sensor, protein kinase R (PKR), may also be critical for IFN induction during infection, although its exact contribution and mechanism of action are not completely understood. Using PKR-deficient cells, we found that PKR was required for type I IFN induction in response to infection by vaccinia virus lacking the PKR antagonist E3L (VVΔE3L), but not by Sendai virus or influenza A virus lacking the IFN-antagonist NS1 (FluΔNS1). IFN induction required the catalytic activity of PKR, but not the phosphorylation of its principal substrate, eIF2α, or the resulting inhibition of host translation. In the absence of PKR, IRF3 nuclear translocation was impaired in response to MDA5 activators, VVΔE3L and encephalomyocarditis virus, but not during infection with a RIG-I-activating virus. Interestingly, PKR interacted with both RIG-I and MDA5; however, PKR was only required for MDA5-mediated, but not RIG-I-mediated, IFN production. Using an artificially activated form of PKR, we showed that PKR activity alone was sufficient for IFN induction. This effect required MAVS and correlated with IRF3 activation, but no longer required MDA5. Nonetheless, PKR activation during viral infection was enhanced by MDA5, as virus-stimulated catalytic activity was impaired in MDA5-null cells. Taken together, our data describe a critical and non-redundant role for PKR following MDA5, but not RIG-I, activation to mediate MAVS-dependent induction of type I IFN through a kinase-dependent mechanism.


Subject(s)
DEAD-box RNA Helicases/metabolism , Encephalomyocarditis virus/immunology , Vaccinia virus/immunology , Vaccinia/immunology , eIF-2 Kinase/metabolism , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Encephalomyocarditis virus/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Genes, Reporter , Humans , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1 , Mutation , Phosphorylation , RNA, Viral/genetics , Receptors, Immunologic , Signal Transduction , Vaccinia/virology , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , eIF-2 Kinase/genetics
8.
Immunity ; 31(6): 941-52, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20064451

ABSTRACT

Interleukin-21 (IL-21) is a pleiotropic cytokine that induces expression of transcription factor BLIMP1 (encoded by Prdm1), which regulates plasma cell differentiation and T cell homeostasis. We identified an IL-21 response element downstream of Prdm1 that binds the transcription factors STAT3 and IRF4, which are required for optimal Prdm1 expression. Genome-wide ChIP-Seq mapping of STAT3- and IRF4-binding sites showed that most regions with IL-21-induced STAT3 binding also bound IRF4 in vivo and furthermore revealed that the noncanonical TTCnnnTAA GAS motif critical in Prdm1 was broadly used for STAT3 binding. Comparing genome-wide expression array data to binding sites revealed that most IL-21-regulated genes were associated with combined STAT3-IRF4 sites rather than pure STAT3 sites. Correspondingly, ChIP-Seq analysis of Irf4(-/-) T cells showed greatly diminished STAT3 binding after IL-21 treatment, and Irf4(-/-) mice showed impaired IL-21-induced Tfh cell differentiation in vivo. These results reveal broad cooperative gene regulation by STAT3 and IRF4.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Interleukins/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factors/genetics , Animals , B-Lymphocytes/immunology , Base Sequence , Binding Sites , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Genome-Wide Association Study , Interferon Regulatory Factors/genetics , Introns , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Positive Regulatory Domain I-Binding Factor 1 , STAT3 Transcription Factor/genetics
9.
Blood ; 124(14): 2252-61, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25150294

ABSTRACT

Juvenile myelomonocytic leukemia, acute myeloid leukemia (AML), and other myeloproliferative neoplasms (MPNs) are genetically heterogeneous but frequently display activating mutations in Ras GTPases and activation of signal transducer and activator of transcription 3 (STAT3). Altered STAT3 activity is observed in up to 50% of AML correlating with poor prognosis. Activated STAT proteins, classically associated with tyrosine phosphorylation, support tumor development as transcription factors, but alternative STAT functions independent of tyrosine phosphorylation have been documented, including roles for serine-phosphorylated STAT3 in mitochondria supporting transformation by oncogenic Ras. We examined requirements for STAT3 in experimental murine K-Ras-dependent hematopoietic neoplasia. We show that STAT3 is phosphorylated on S727 but not Y705 in diseased animals. Moreover, a mouse with a point mutation abrogating STAT3 S727 phosphorylation displayed delayed onset and decreased disease severity with significantly extended survival. Activated K-Ras required STAT3 for cytokine-independent growth of myeloid progenitors in vitro, and mitochondrially restricted STAT3 and STAT3-Y705F, both transcriptionally inert mutants, supported factor-independent growth. STAT3 was dispensable for growth of normal or K-Ras-mutant myeloid progenitors in response to cytokines. However, abrogation of STAT3-S727 phosphorylation impaired factor-independent malignant growth. These data document that serine-phosphorylated mitochondrial STAT3 supports neoplastic hematopoietic cell growth induced by K-Ras.


Subject(s)
Gene Expression Regulation, Neoplastic , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , STAT3 Transcription Factor/metabolism , Serine/chemistry , Alleles , Animals , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloproliferative Disorders/metabolism , Phosphorylation , Proto-Oncogene Proteins p21(ras)/genetics , Stem Cells/cytology , Tyrosine/chemistry , ras GTPase-Activating Proteins/metabolism
10.
Nature ; 467(7312): 214-7, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20829794

ABSTRACT

Dendritic cells serve a key function in host defence, linking innate detection of microbes to activation of pathogen-specific adaptive immune responses. Whether there is cell-intrinsic recognition of human immunodeficiency virus (HIV) by host innate pattern-recognition receptors and subsequent coupling to antiviral T-cell responses is not yet known. Dendritic cells are largely resistant to infection with HIV-1, but facilitate infection of co-cultured T-helper cells through a process of trans-enhancement. Here we show that, when dendritic cell resistance to infection is circumvented, HIV-1 induces dendritic cell maturation, an antiviral type I interferon response and activation of T cells. This innate response is dependent on the interaction of newly synthesized HIV-1 capsid with cellular cyclophilin A (CYPA) and the subsequent activation of the transcription factor IRF3. Because the peptidylprolyl isomerase CYPA also interacts with HIV-1 capsid to promote infectivity, our results indicate that capsid conformation has evolved under opposing selective pressures for infectivity versus furtiveness. Thus, a cell-intrinsic sensor for HIV-1 exists in dendritic cells and mediates an antiviral immune response, but it is not typically engaged owing to the absence of dendritic cell infection. The virulence of HIV-1 may be related to evasion of this response, the manipulation of which may be necessary to generate an effective HIV-1 vaccine.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/immunology , HIV-1/immunology , Immunity, Innate , Capsid Proteins/immunology , Cell Line , Cyclophilin A/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , HIV Infections/virology , HIV-1/physiology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Lymphocyte Activation , Monocytes/cytology , T-Lymphocytes/immunology
11.
Cancer Cell ; 10(1): 77-87, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16843267

ABSTRACT

The tumor suppressor STAT1 is considered a key regulator of the surveillance of developing tumors. Here, we describe an unexpected tumor-promoting role for STAT1 in leukemia. STAT1(-/-) mice are partially protected from leukemia development, and STAT1(-/-) tumor cells induce leukemia in RAG2(-/-) and immunocompetent mice with increased latency. The low MHC class I protein levels of STAT1(-/-) tumor cells enable efficient NK cell lysis and account for the enhanced tumor clearance. Strikingly, STAT1(-/-) tumor cells acquire increased MHC class I expression upon leukemia progression. These findings define STAT1 as a tumor promoter in leukemia development. Furthermore, we describe the upregulation of MHC class I expression as a general mechanism that allows for the escape of hematopoietic malignancies from immune surveillance.


Subject(s)
Leukemia, Experimental/pathology , STAT1 Transcription Factor/physiology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Genotype , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins v-abl/genetics , Oncogene Proteins v-abl/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phenotype , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Stem Cells/metabolism , Stem Cells/pathology , Survival Analysis
12.
Mitochondrial Commun ; 2: 1-13, 2024.
Article in English | MEDLINE | ID: mdl-38500969

ABSTRACT

Signal transducer and activator of transcription (STAT) 3 has been found within mitochondria in addition to its canonical role of shuttling between cytoplasm and nucleus during cytokine signaling. Mitochondrial STAT3 has been implicated in modulation of cellular metabolism, largely through effects on the respiratory electron transport chain. However, the structural requirements underlying mitochondrial targeting and function have remained unclear. Here, we show that mitochondrial STAT3 partitions between mitochondrial compartments defined by differential detergent solubility, suggesting that mitochondrial STAT3 is membrane associated. The majority of STAT3 was found in an SDS soluble fraction copurifying with respiratory chain proteins, including numerous components of the complex I NADH dehydrogenase, while a minor component was found with proteins of the mitochondrial translation machinery. Mitochondrial targeting of STAT3 required the amino-terminal domain, and an internal linker domain motif also directed mitochondrial translocation. However, neither the phosphorylation of serine 727 nor the presence of mitochondrial DNA was required for the mitochondrial localization of STAT3. Two cysteine residues in the STAT3 SH2 domain, which have been previously suggested to be targets for protein palmitoylation, were also not required for mitochondrial translocation, but were required for its function as an enhancer of complex I activity. These structural determinants of STAT3 mitochondrial targeting and function provide potential therapeutic targets for disrupting the activity of mitochondrial STAT3 in diseases such as cancer.

13.
Blood ; 117(23): 6198-201, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21505191

ABSTRACT

The contribution of specific cell types to the production of cytokines that regulate hematopoiesis is still not well defined. We have previously identified T cell-dependent regulation of hematopoietic progenitor cell (HPC) numbers and cycling. In this report, we demonstrated that HPC activity is decreased in mice with STAT3-deficient T cells, a phenotype that is not because of decreased expression of IL-17 or RORγt. STAT3 expression in T cells was required for IL-21 production by multiple T helper subsets, and neutralization of IL-21 resulted in decreased HPC activity identical to that in mice with STAT3-deficient T cells. Importantly, injection of IL-21 rescued HPC activity in mice with STAT3-deficient T cells. Thus, STAT3-dependent IL-21 production in T cells is required for HPC homeostasis.


Subject(s)
Gene Expression Regulation/immunology , Hematopoietic Stem Cells/immunology , Homeostasis/immunology , Interleukins/immunology , STAT3 Transcription Factor/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/biosynthesis , Interleukins/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism
14.
PLoS Biol ; 8(4): e1000361, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20436908

ABSTRACT

Autocrine priming of cells by small quantities of constitutively produced type I interferon (IFN) is a well-known phenomenon. In the absence of type I IFN priming, cells display attenuated responses to other cytokines, such as anti-viral protection in response to IFNgamma. This phenomenon was proposed to be because IFNalpha/beta receptor1 (IFNAR1) is a component of the IFNgamma receptor (IFNGR), but our new data are more consistent with a previously proposed model indicating that regulated expression of STAT1 may also play a critical role in the priming process. Initially, we noticed that DNA binding activity of STAT1 was attenuated in c-Jun(-/-) fibroblasts because they expressed lower levels of STAT1 than wild-type cells. However, expression of STAT1 was rescued by culturing c-Jun(-/-) fibroblasts in media conditioned by wild-type fibroblasts suggesting they secreted a STAT1-inducing factor. The STAT1-inducing factor in fibroblast-conditioned media was IFNbeta, as it was inhibited by antibodies to IFNAR1, or when IFNbeta expression was knocked down in wild-type cells. IFNAR1(-/-) fibroblasts, which cannot respond to this priming, also expressed reduced levels of STAT1, which correlated with their poor responses to IFNgamma. The lack of priming in IFNAR1(-/-) fibroblasts was compensated by over-expression of STAT1, which rescued molecular responses to IFNgamma and restored the ability of IFNgamma to induce protective anti-viral immunity. This study provides a comprehensive description of the molecular events involved in priming by type I IFN. Adding to the previous working model that proposed an interaction between type I and II IFN receptors, our work and that of others demonstrates that type I IFN primes IFNgamma-mediated immune responses by regulating expression of STAT1. This may also explain how type I IFN can additionally prime cells to respond to a range of other cytokines that use STAT1 (e.g., IL-6, M-CSF, IL-10) and suggests a potential mechanism for the changing levels of STAT1 expression observed during viral infection.


Subject(s)
Interferon Type I/metabolism , Interferon-gamma/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Interferon Type I/genetics , Interferon-gamma/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/genetics , Spleen/cytology
15.
J Immunol ; 187(5): 2578-85, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21810606

ABSTRACT

Type I IFNs are crucial cytokines of innate immunity for combating viral infections. Signaling through type I IFN receptors triggers the activation of STAT proteins, including STAT1, STAT2, and STAT3. Although an essential role of STAT1 and STAT2 for type I IFN-induced antiviral response has been well established by studies of gene-targeted mice and human mutations, the role of STAT3 for this response remains unclear. Using gain-of-function and loss-of-function approaches, we demonstrated that STAT3 negatively regulates type I IFN-mediated response. STAT3 knockdown or knockout cells displayed enhanced gene expression and antiviral activity in response to IFN-α/ß. Restoration of STAT3 to STAT3KO cells resulted in attenuation of the response. Upon viral infection, increased type I IFN production in STAT3KO cells resulted in enhanced STAT activation and ISG expression. One mechanism for the enhanced IFN production and response in the absence of STAT3 might operate through an MDA5-dependent manner. STAT3 also appeared to suppress IFN response directly in a manner dependent on its N-terminal domain and independent of its function as a transcriptional factor. Taken together, these results define STAT3 as a negative regulator of type I IFN response and provide a therapeutic target for viral infections.


Subject(s)
Interferon Type I/immunology , Lentivirus Infections/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Gene Expression Regulation/immunology , Lentivirus/immunology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Am J Physiol Cell Physiol ; 302(5): C804-20, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22159083

ABSTRACT

We report unexpected nongenomic functions of signal transducer and activator of transcription (STAT) 5 species in the cytoplasm aimed at preserving the structure and function of the Golgi apparatus and rough endoplasmic reticulum (ER) in vascular cells. Immunoimaging and green fluorescent protein-tagged-STAT5a protein localization studies showed the constitutive association of nonphosphorylated STAT5a, and to a lesser extent STAT5b, with the Golgi apparatus and of STAT5a with centrosomes in human pulmonary arterial endothelial and smooth muscle cells. Acute knockdown of STAT5a/b species using small interfering RNAs (siRNAs), including in the presence of an mRNA synthesis inhibitor (5,6-dichloro-1-ß-d-ribofuranosylbenzimidazole), produced a dramatic phenotype within 1 day, consisting of dilatation and fragmentation of Golgi cisternae, a marked tubule-to-cyst change in the ER, increased accumulation of reticulon-4 (RTN4)/Nogo-B and atlastin-3 (ATL3) at cyst-zone boundaries, cystic separation of the outer and inner nuclear membranes, accompanied by scalloped/lunate distortion of the nucleus, with accumulation of RTN4 on convex sides of distorted nuclei. These cells showed inhibition of vesicular stomatitis virus G protein glycoprotein trafficking, mitochondrial fragmentation, and reduced mitochondrial function. STAT5a/b(-/-) mouse embryo fibroblasts also showed altered ER/Golgi dynamics. RTN4 knockdown using siRNA did not affect development of the cystic phenotype; ATL3 siRNA led to effacement of cyst-zone boundaries. In magnetic-bead cross-immunopanning assays, ATL3 bound both STAT5a and STAT5b. Remarkably, this novel cystic ER/lunate nucleus phenotype was characteristic of vascular cells in arterial lesions of idiopathic pulmonary hypertension, an unrelentingly fatal human disease. These data provide evidence of a STAT-family protein regulating the structure of a cytoplasmic organelle and implicate this mechanism in the pathogenesis of a human disease.


Subject(s)
Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cattle , Cell Nucleus/metabolism , Cells, Cultured , Centrosome/metabolism , Centrosome/ultrastructure , Cytoplasm/metabolism , Dichlororibofuranosylbenzimidazole/chemistry , Endothelial Cells , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/metabolism , Membrane Glycoproteins , Mice , Microscopy, Electron , Myelin Proteins/metabolism , Myocytes, Smooth Muscle , Nogo Proteins , Protein Transport , Pulmonary Artery/cytology , RNA, Small Interfering , STAT5 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Viral Envelope Proteins
17.
Breast Cancer Res ; 14(1): R16, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22264274

ABSTRACT

INTRODUCTION: Although breast cancers expressing estrogen receptor-α (ERα) and progesterone receptors (PR) are the most common form of mammary malignancy in humans, it has been difficult to develop a suitable mouse model showing similar steroid hormone responsiveness. STAT transcription factors play critical roles in mammary gland tumorigenesis, but the precise role of STAT1 remains unclear. Herein, we show that a subset of human breast cancers display reduced STAT1 expression and that mice lacking STAT1 surprisingly develop ERα+/PR+ mammary tumors. METHODS: We used a combination of approaches, including histological examination, gene targeted mice, gene expression analysis, tumor transplantaion, and immunophenotyping, to pursue this study. RESULTS: Forty-five percent (37/83) of human ERα+ and 22% (17/78) of ERα- breast cancers display undetectable or low levels of STAT1 expression in neoplastic cells. In contrast, STAT1 expression is elevated in epithelial cells of normal breast tissues adjacent to the malignant lesions, suggesting that STAT1 is selectively downregulated in the tumor cells during tumor progression. Interestingly, the expression levels of STAT1 in the tumor-infiltrating stromal cells remain elevated, indicating that single-cell resolution analysis of STAT1 level in primary breast cancer biopsies is necessary for accurate assessment. Female mice lacking functional STAT1 spontaneously develop mammary adenocarcinomas that comprise > 90% ERα+/PR+ tumor cells, and depend on estrogen for tumor engraftment and progression. Phenotypic marker analyses demonstrate that STAT1-/- mammary tumors arise from luminal epithelial cells, but not myoepithelial cells. In addition, the molecular signature of the STAT1-/- mammary tumors overlaps closely to that of human luminal breast cancers. Finally, introduction of wildtype STAT1, but not a STAT1 mutant lacking the critical Tyr701 residue, into STAT1-/- mammary tumor cells results in apoptosis, demonstrating that the tumor suppressor function of STAT1 is cell-autonomous and requires its transcriptional activity. CONCLUSIONS: Our findings demonstrate that STAT1 suppresses mammary tumor formation and its expression is frequently lost during breast cancer progression. Spontaneous mammary tumors that develop in STAT1-/- mice closely recapitulate the progression, ovarian hormone responsiveness, and molecular characteristics of human luminal breast cancer, the most common subtype of human breast neoplasms, and thus represent a valuable platform for testing novel treatments and detection modalities.


Subject(s)
Adenocarcinoma/metabolism , Estrogen Receptor alpha/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasms, Hormone-Dependent/metabolism , STAT1 Transcription Factor/deficiency , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Down-Regulation , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Kaplan-Meier Estimate , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Transcriptome
18.
Nat Med ; 11(6): 623-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15895073

ABSTRACT

Anaplastic large cell lymphomas (ALCLs) are caused by chromosomal translocations that juxtapose the anaplastic lymphoma kinase (ALK) proto-oncogene to a dimerization partner, resulting in constitutive expression of ALK and ALK tyrosine kinase activity. One substrate of activated ALK in human ALCLs is the transcription factor Stat3, and its phosphorylation is accurately recapitulated in a new nucleophosmin (NPM)-ALK transgenic mouse model of lymphomagenesis. Here we show by gene targeting that Stat3 is required for the transformation of mouse embryonic fibroblasts in vitro, for the development of B-cell lymphoma in transgenic mice and for the growth and survival of both human and mouse NPM-ALK-transformed B and T cells. Ablation of Stat3 expression by antisense oligonucleotides significantly (P < 0.0001) impaired the growth of human and mouse NPM-ALK tumors in vivo. Pharmacological ablation of Stat3 represents a new candidate approach for the treatment of human lymphoma


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins/physiology , Lymphoma, Large B-Cell, Diffuse/physiopathology , Protein-Tyrosine Kinases/physiology , Trans-Activators/physiology , Anaplastic Lymphoma Kinase , Animals , Cell Line , Fibroblasts/physiology , Humans , Lymphoma, T-Cell/physiopathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Molecular Sequence Data , Multiple Myeloma/physiopathology , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Mas , Receptor Protein-Tyrosine Kinases , STAT3 Transcription Factor
19.
Bio Protoc ; 12(11): e4438, 2022 Jun 05.
Article in English | MEDLINE | ID: mdl-35799901

ABSTRACT

Transplantation of hematopoietic material into recipient mice is an assay routinely used to determine the presence and function of hematopoietic stem and progenitor cells (HSPCs) in vivo . The principle of the method is to transplant donor cells being tested for HSPCs into a recipient mouse following bone marrow ablation and testing for reconstitution of hematopoiesis. Congenic mouse strains where donor and recipient differ by a distinct cell surface antigen (commonly CD45.1 versus CD45.2) are used to distinguish between cells derived from the donor and any residual recipient cells. Typically, the transplantation is performed using bone marrow cells, which are enriched for HSPCs. Here, we describe an analogous procedure using hematopoietic material from spleen, allowing detection of functional progenitors and/or stem cells in the spleen that can occur under certain pathologies. Key to the success of this procedure is the prior removal of mature T cells from the donor sample, to minimize graft versus host reactions. As such, this protocol is highly analogous to standard bone marrow transplant procedures, differing mainly only in the source of stem cells (spleen rather than bone marrow) and the recommendation for T cell depletion to avoid potential immune incompatibilities. Graphical abstract: Schematic overview for assessment of stem cells in spleen by transplantation. Single cell suspensions from spleens are depleted of potentially pathogenic mature T lymphocytes by magnetic bead immunoselection using biotinylated antibodies against CD4 and CD8, followed by streptavidin magnetic beads, which are subsequently removed by using a magnet (MojoSort, Biolegend). Successful T cell depletion is then evaluated by Fluorescence Activated Cell Sorting (FACS). T-cell depleted cell suspension is injected intravenously through the retro-orbital sinus into lethally irradiated recipients. Recipients are analyzed for successful engraftment by FACS analysis for the presence of donor-derived mature hematopoietic lineages in the peripheral blood. A second serial transplantation can be used to document the presence of long-term reconstituting stem cells in the periphery of the original donor mice.

20.
Stroke ; 42(11): 3277-80, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21868728

ABSTRACT

BACKGROUND AND PURPOSE: Ancrod, derived from Malayan pit viper venom, has been tested as ischemic stroke treatment in clinical trials with inconsistent results. We studied the actions of ancrod on fibrinolysis pathways in patient plasma samples and endothelial cell culture systems. METHODS: We analyzed fibrinogen levels during the first 6 hours of ancrod infusion in patients entered in the Stroke Treatment with Ancrod Trial. For the in vitro study, human brain microvascular endothelial cells incubated with plasminogen or with human brain microvascular endothelial cell-conditioned medium were co-incubated with ancrod and fibrinogen under normal or oxygen-glucose deprivation conditions over 6 hours. RESULTS: Fibrinogen levels decreased both in vivo and in vitro. Ancrod generated fibrinopeptide A, caused visible clot formation, and reduced levels of tissue-type plasminogen activator antigen in the human brain microvascular endothelial cell system and in a cell-free system with conditioned media. CONCLUSIONS: The in vitro results indicate that ancrod causes local fibrin formation and secondary depletion of tissue-type plasminogen activator by binding to fibrin clot. Ancrod-induced fibrin formation could result in cerebral microvascular occlusion and may explain the suboptimal clinical effects of ancrod in human stroke trials.


Subject(s)
Ancrod/therapeutic use , Fibrin/metabolism , Stroke/blood , Stroke/drug therapy , Cells, Cultured , Culture Media, Conditioned , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL