Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(12): 1411-1424, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36240433

RESUMEN

STAT3 mutations, predominantly in the DNA-binding domain (DBD) and Src-homology 2 domain (SH2D), cause rare cases of immunodeficiency, malignancy, and autoimmunity. The exact mechanisms by which these mutations abrogate or enhance STAT3 function are not completely understood. Here, we examined how loss-of-function (LOF) and gain-of-function (GOF) STAT3 mutations within the DBD and SH2D affect monomer and homodimer protein stability as well as their effect on key STAT3 activation events, including recruitment to phosphotyrosine (pY) sites within peptide hormone receptors, tyrosine phosphorylation at Y705, dimerization, nuclear translocation, and DNA binding. The DBD LOF mutants showed reduced DNA binding when homodimerized, whereas the DBD GOF mutants showed increased DNA binding. DBD LOF and GOF mutants showed minimal changes in other STAT3 functions or in monomer or homodimer protein stability. However, SH2D LOF mutants demonstrated reduced conformational stability as either monomers or homodimers, leading to decreased pY-peptide recruitment, tyrosine phosphorylation, dimerization, nuclear localization, and DNA binding. In contrast, cancer-causing SH2D GOF mutants showed increased STAT3 homodimer stability, which increased their DNA binding. Of note, a small-molecule inhibitor of STAT3 that targets the tyrosine phosphopeptide-binding pocket within the STAT3 SH2D potently inhibited cell proliferation driven by STAT3 SH2D GOF mutants. These findings indicate that the stability of STAT3 protein monomer and homodimer is critical for the pathogenesis of diseases caused by SH2D LOF and GOF mutations and suggest that agents that modulate STAT3 monomer and/or homodimer protein stability may have therapeutic value in diseases caused by these mutations.


Asunto(s)
Factor de Transcripción STAT3 , Dominios Homologos src , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Mutación , Dominios Homologos src/genética , ADN/metabolismo , Tirosina/genética
2.
Pharmacol Rev ; 72(2): 486-526, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32198236

RESUMEN

Before it was molecularly cloned in 1994, acute-phase response factor or signal transducer and activator of transcription (STAT)3 was the focus of intense research into understanding the mammalian response to injury, particularly the acute-phase response. Although known to be essential for liver production of acute-phase reactant proteins, many of which augment innate immune responses, molecular cloning of acute-phase response factor or STAT3 and the research this enabled helped establish the central function of Janus kinase (JAK) family members in cytokine signaling and identified a multitude of cytokines and peptide hormones, beyond interleukin-6 and its family members, that activate JAKs and STAT3, as well as numerous new programs that their activation drives. Many, like the acute-phase response, are adaptive, whereas several are maladaptive and lead to chronic inflammation and adverse consequences, such as cachexia, fibrosis, organ dysfunction, and cancer. Molecular cloning of STAT3 also enabled the identification of other noncanonical roles for STAT3 in normal physiology, including its contribution to the function of the electron transport chain and oxidative phosphorylation, its basal and stress-related adaptive functions in mitochondria, its function as a scaffold in inflammation-enhanced platelet activation, and its contributions to endothelial permeability and calcium efflux from endoplasmic reticulum. In this review, we will summarize the molecular and cellular biology of JAK/STAT3 signaling and its functions under basal and stress conditions, which are adaptive, and then review maladaptive JAK/STAT3 signaling in animals and humans that lead to disease, as well as recent attempts to modulate them to treat these diseases. In addition, we will discuss how consideration of the noncanonical and stress-related functions of STAT3 cannot be ignored in efforts to target the canonical functions of STAT3, if the goal is to develop drugs that are not only effective but safe. SIGNIFICANCE STATEMENT: Key biological functions of Janus kinase (JAK)/signal transducer and activator of transcription (STAT)3 signaling can be delineated into two broad categories: those essential for normal cell and organ development and those activated in response to stress that are adaptive. Persistent or dysregulated JAK/STAT3 signaling, however, is maladaptive and contributes to many diseases, including diseases characterized by chronic inflammation and fibrosis, and cancer. A comprehensive understanding of JAK/STAT3 signaling in normal development, and in adaptive and maladaptive responses to stress, is essential for the continued development of safe and effective therapies that target this signaling pathway.


Asunto(s)
Fibrosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Neoplasias/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ensayos Clínicos Fase II como Asunto , Fibrosis/metabolismo , Humanos , Inflamación/metabolismo , Quinasas Janus/genética , Terapia Molecular Dirigida , Neoplasias/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor de Transcripción STAT3/genética
3.
Trans Am Clin Climatol Assoc ; 132: 61-76, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36196170

RESUMEN

Signal transducer and activator of transcription (STAT) 3 has been assigned to the group of "undruggable" disease-causing proteins, despite its containing a Src-homology (SH) 2 domain, a potential Achilles' heel that has eluded successful targeting by academic and pharmaceutical groups over the past 30 years. Based on mutational and modeling studies, our group developed a unique virtual ligand screening strategy targeting the STAT3 SH2 domain that was coupled to robust biochemical and cellular assays and structure-based medicinal chemistry and led to the identification of TTI-101. TTI-101 represents one of the most advanced, direct, small-molecule inhibitors of an SH2 domain-containing, disease-causing protein in clinical development. TTI-101 is currently being evaluated in a Phase 1 study to determine safety and tolerability in addition to pharmacodynamic effects and efficacy in patients with advanced solid tumors.


Asunto(s)
Naftoles , Sulfonamidas , Humanos , Ligandos , Preparaciones Farmacéuticas
4.
Pharmacol Res ; 169: 105637, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33932608

RESUMEN

Efforts to develop STAT3 inhibitors have focused on its SH2 domain starting with short phosphotyrosylated peptides based on STAT3 binding motifs, e.g. pY905LPQTV within gp130. Despite binding to STAT3 with high affinity, issues regarding stability, bioavailability, and membrane permeability of these peptides, as well as peptidomimetics such as CJ-887, have limited their further clinical development and led to interest in small-molecule inhibitors. Some small molecule STAT3 inhibitors, identified using structure-based virtual ligand screening (SB-VLS); while having favorable drug-like properties, suffer from weak binding affinities, possibly due to the high flexibility of the target domain. We conducted molecular dynamic (MD) simulations of the SH2 domain in complex with CJ-887, and used an averaged structure from this MD trajectory as an "induced-active site" receptor model for SB-VLS of 110,000 compounds within the SPEC database. Screening was followed by re-docking and re-scoring of the top 30% of hits, selection for hit compounds that directly interact with pY + 0 binding pocket residues R609 and S613, and testing for STAT3 targeting in vitro, which identified two lead hits with good activity and favorable drug-like properties. Unlike most small-molecule STAT3 inhibitors previously identified, which contain negatively-charged moieties that mediate binding to the pY + 0 binding pocket, these compounds are uncharged and likely will serve as better candidates for anti-STAT3 drug development. IMPLICATIONS: SB-VLS, using an averaged structure from molecular dynamics (MD) simulations of STAT3 SH2 domain in a complex with CJ-887, a known peptidomimetic binder, identify two highly potent, neutral, low-molecular weight STAT3-inhibitors with favorable drug-like properties.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Factor de Transcripción STAT3/antagonistas & inhibidores , Dominios Homologos src , Alquilación , Sitios de Unión/efectos de los fármacos , Western Blotting , Línea Celular Tumoral/efectos de los fármacos , Cromatografía de Gases y Espectrometría de Masas , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/genética , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Dominios Homologos src/efectos de los fármacos
5.
Am J Physiol Renal Physiol ; 319(1): F84-F92, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32475130

RESUMEN

Loss of muscle proteins increases the morbidity and mortality of patients with chronic kidney disease (CKD), and there are no reliable preventive treatments. We uncovered a STAT3/CCAAT-enhancer-binding protein-δ to myostatin signaling pathway that activates muscle protein degradation in mice with CKD or cancer; we also identified a small-molecule inhibitor of STAT3 (TTI-101) that blocks this pathway. To evaluate TTI-101 as a treatment for CKD-induced cachexia, we measured TTI-101 pharmacokinetics and pharmacodynamics in control and CKD rats that were orally administered TTI-101or its diluent. The following two groups of gavage-fed rats were studied: sham-operated control rats and CKD rats. Plasma was collected serially (0, 0.25, 0.5, 1, 2, 4, 8, and 24 h) following TTI-101 administration (at oral doses of 0, 10, 30, or 100 mg/kg). Plasma levels of TTI-101 were measured by LC-MS/MS, and pharmacokinetic results were analyzed with the PKSolver program. Plasma TTI-101 levels increased linearly with doses; the maximum plasma concentrations and time to maximal plasma levels (~1 h) were similar in sham-operated control rats and CKD rats. Notably, gavage treatment of TTI-101 for 3 days produced TTI-101 muscle levels in sham control rats and CKD rats that were not significantly different. CKD rats that received TTI-101 for 7 days had suppression of activated STAT3 and improved muscle grip strength; there also was a trend for increasing body and muscle weights. TTI-101 was tolerated at doses of 100 mg·kg-1·day-1 for 7 days. These results with TTI-101 in rats warrant its development as a treatment for cachexia in humans.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Músculo Esquelético/efectos de los fármacos , Naftoles/farmacología , Proteolisis/efectos de los fármacos , Insuficiencia Renal Crónica/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Peso Corporal/efectos de los fármacos , Cromatografía Liquida , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacocinética , Fuerza de la Mano , Músculo Esquelético/metabolismo , Naftoles/farmacocinética , Ratas , Sulfonamidas/farmacocinética , Espectrometría de Masas en Tándem
6.
Am J Physiol Endocrinol Metab ; 318(5): E625-E635, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32101031

RESUMEN

Cellular mechanisms causing insulin resistance (IR) in chronic kidney disease (CKD) are poorly understood. One potential mechanism is that CKD-induced inflammation activates the signal transducer and activator of transcription 3 (Stat3) in muscle. We uncovered increased p-Stat3 in muscles of mice with CKD or mice fed high-fat diet (HFD). Activated Stat3 stimulates the expression of Fbxo40, a muscle-specific E3 ubiquitin ligase that stimulates ubiquitin conjugation leading to degradation of insulin receptor substrate 1 (IRS1). Evidence that Stat3 activates Fbxo40 includes 1) potential Stat3 binding sites in Fbxo40 promoters; 2) Stat3 binding to the Fbxo40 promoter; and 3) constitutively active Stat3 stimulating both Fbxo40 expression and its promoter activity. We found that IL-6 activates Stat3 in myotubes, increasing Fbxo40 expression with reduced IRS1 and p-Akt. Knockdown Fbxo40 using siRNA from myotubes results in higher levels of IRS1 and p-Akt despite the presence of IL-6. We treated mice with a small-molecule inhibitor of Stat3 (TTI-101) and found improved glucose tolerance and insulin signaling in skeletal muscles of mice with CKD or fed an HFD. Finally, we uncovered improved glucose tolerance in mice with muscle-specific Stat3 KO versus results in Stat3f/f mice in response to the HFD. Thus Stat3 activation in muscle increases IR in mice. Inhibition of Stat3 by TTI-101 could be developed into clinical strategies to improve muscle insulin signaling in inflammation and other catabolic diseases.


Asunto(s)
Proteínas F-Box/metabolismo , Resistencia a la Insulina/fisiología , Músculo Esquelético/metabolismo , Insuficiencia Renal Crónica/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Dieta Alta en Grasa , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT3/genética , Transducción de Señal/fisiología
7.
Org Biomol Chem ; 18(17): 3288-3296, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32286579

RESUMEN

Recurrence and drug resistance are major challenges in the treatment of acute myeloid leukemia (AML) that spur efforts to identify new clinical targets and active agents. STAT3 has emerged as a potential target in resistant AML, but inhibiting STAT3 function has proven challenging. This paper describes synthetic studies and biological assays for a naphthalene sulfonamide inhibitor class of molecules that inhibit G-CSF-induced STAT3 phosphorylation in cellulo and induce apoptosis in AML cells. We describe two different approaches to inhibitor design: first, variation of substituents on the naphthalene sulfonamide core allows improvements in anti-STAT activity and creates a more thorough understanding of anti-STAT SAR. Second, a novel approach involving hybrid sulfonamide-rhodium(ii) conjugates tests our ability to use cooperative organic-inorganic binding for drug development, and to use SAR studies to inform metal conjugate design. Both approaches have produced compounds with improved binding potency. In vivo and in cellulo experiments further demonstrate that these approaches can also lead to improved activity in living cells, and that compound 3aa slows disease progression in a xenograft model of AML.


Asunto(s)
Antineoplásicos/química , Leucemia Mieloide Aguda/tratamiento farmacológico , Naftalenos/química , Inhibidores de Proteínas Quinasas/química , Factor de Transcripción STAT3/antagonistas & inhibidores , Sulfonamidas/química , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Modelos Moleculares , Terapia Molecular Dirigida , Neoplasias Experimentales , Oxidación-Reducción , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Factor de Transcripción STAT3/genética , Relación Estructura-Actividad
8.
Blood ; 128(26): 3061-3072, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-27799162

RESUMEN

Autosomal dominant hyper-IgE syndrome (AD-HIES) is caused by dominant-negative mutations in STAT3; however, the molecular basis for mutant STAT3 allele dysfunction is unclear and treatment remains supportive. We hypothesized that AD-HIES mutations decrease STAT3 protein stability and that mutant STAT3 activity can be improved by agents that increase chaperone protein activity. We used computer modeling to characterize the effect of STAT3 mutations on protein stability. We measured STAT3 protein half-life (t1/2) and determined levels of STAT3 phosphorylated on tyrosine (Y) 705 (pY-STAT3) and mRNA levels of STAT3 gene targets in Epstein-Barr virus-transformed B (EBV) cells, human peripheral blood mononuclear cells (PBMCs), and mouse splenocytes incubated without or with chaperone protein modulators-HSF1A, a small-molecule TRiC modulator, or geranylgeranylacetone (GGA), a drug that upregulates heat shock protein (HSP) 70 and HSP90. Computer modeling predicted that 81% of AD-HIES mutations are destabilizing. STAT3 protein t1/2 in EBV cells from AD-HIES patients with destabilizing STAT3 mutations was markedly reduced. Treatment of EBV cells containing destabilizing STAT3 mutations with either HSF1A or GGA normalized STAT3 t1/2, increased pY-STAT3 levels, and increased mRNA levels of STAT3 target genes up to 79% of control. In addition, treatment of human PBMCs or mouse splenocytes containing destabilizing STAT3 mutations with either HSF1A or GGA increased levels of cytokine-activated pY-STAT3 within human CD4+ and CD8+ T cells and numbers of IL-17-producing CD4+ mouse splenocytes, respectively. Thus, most AD-HIES STAT3 mutations are destabilizing; agents that modulate chaperone protein function improve STAT3 stability and activity in T cells and may provide a specific treatment.


Asunto(s)
Síndrome de Job/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Citocinas/farmacología , Proteínas de Unión al ADN/metabolismo , Diterpenos/farmacología , Semivida , Factores de Transcripción del Choque Térmico , Herpesvirus Humano 4/fisiología , Humanos , Interleucina-17/metabolismo , Síndrome de Job/patología , Ratones , Modelos Moleculares , Proteínas Mutantes/metabolismo , Mutación/genética , Fosfotirosina/metabolismo , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/genética , Bazo/patología , Factores de Transcripción/metabolismo
9.
Int J Mol Sci ; 19(8)2018 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-30081609

RESUMEN

Signal transducer and activator of transcription (STAT) 3 plays a central role in the host response to injury. It is activated rapidly within cells by many cytokines, most notably those in the IL-6 family, leading to pro-proliferative and pro-survival programs that assist the host in regaining homeostasis. With persistent activation, however, chronic inflammation and fibrosis ensue, leading to a number of debilitating diseases. This review summarizes advances in our understanding of the role of STAT3 and its targeting in diseases marked by chronic inflammation and/or fibrosis with a focus on those with the largest unmet medical need.


Asunto(s)
Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Fibrosis/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Asma/inmunología , Asma/metabolismo , Caquexia/inmunología , Caquexia/metabolismo , Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , Fibrosis/inmunología , Humanos
10.
J Biol Chem ; 291(9): 4732-41, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26706127

RESUMEN

AML1-ETO is the most common fusion oncoprotein causing acute myeloid leukemia (AML), a disease with a 5-year survival rate of only 24%. AML1-ETO functions as a rogue transcription factor, altering the expression of genes critical for myeloid cell development and differentiation. Currently, there are no specific therapies for AML1-ETO-positive AML. While known for decades to be the translational product of a chimeric gene created by the stable chromosome translocation t(8;21)(q22;q22), it is not known how AML1-ETO achieves its native and functional conformation or whether this process can be targeted for therapeutic benefit. Here, we show that the biosynthesis and folding of the AML1-ETO protein is facilitated by interaction with the essential eukaryotic chaperonin TRiC (or CCT). We demonstrate that a folding intermediate of AML1-ETO binds to TRiC directly, mainly through its ß-strand rich, DNA-binding domain (AML-(1-175)), with the assistance of HSP70. Our results suggest that TRiC contributes to AML1-ETO proteostasis through specific interactions between the oncoprotein's DNA-binding domain, which may be targeted for therapeutic benefit.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Animales , Bovinos , Supervivencia Celular , Chaperonina con TCP-1/antagonistas & inhibidores , Chaperonina con TCP-1/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células HEK293 , Humanos , Inmunoprecipitación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Subunidades de Proteína , Proteína 1 Compañera de Translocación de RUNX1 , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Reticulocitos/metabolismo
11.
FASEB J ; 30(1): 129-40, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26324850

RESUMEN

Lung fibrosis is the hallmark of the interstitial lung diseases. Alveolar epithelial cell (AEC) injury is a key step that contributes to a profibrotic microenvironment. Fibroblasts and myofibroblasts subsequently accumulate and deposit excessive extracellular matrix. In addition to TGF-ß, the IL-6 family of cytokines, which signal through STAT-3, may also contribute to lung fibrosis. In the current manuscript, the extent to which STAT-3 inhibition decreases lung fibrosis is investigated. Phosphorylated STAT-3 was elevated in lung biopsies from patients with idiopathic pulmonary fibrosis and bleomycin (BLM)-induced fibrotic murine lungs. C-188-9, a small molecule STAT-3 inhibitor, decreased pulmonary fibrosis in the intraperitoneal BLM model as assessed by arterial oxygen saturation (control, 84.4 ± 1.3%; C-188-9, 94.4 ± 0.8%), histology (Ashcroft score: untreated, 5.4 ± 0.25; C-188-9, 3.3 ± 0.14), and attenuated fibrotic markers such as diminished α-smooth muscle actin, reduced collagen deposition. In addition, C-188-9 decreased the expression of epithelial injury markers, including hypoxia-inducible factor-1α (HIF-1α) and plasminogen activator inhibitor-1 (PAI-1). In vitro studies show that inhibition of STAT-3 decreased IL-6- and TGF-ß-induced expression of multiple genes, including HIF-1α and PAI-1, in AECs. Furthermore, C-188-9 decreased fibroblast-to-myofibroblast differentiation. Finally, TGF-ß stimulation of lung fibroblasts resulted in SMAD2/SMAD3-dependent phosphorylation of STAT-3. These findings demonstrate that STAT-3 contributes to the development of lung fibrosis and suggest that STAT-3 may be a therapeutic target in pulmonary fibrosis.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Miofibroblastos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Bleomicina/farmacología , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrosis Pulmonar Idiopática/genética , Masculino , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/genética
12.
PLoS Biol ; 12(4): e1001844, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24756126

RESUMEN

Signal transducer and activator of transcription 3 (Stat3) transduces signals of many peptide hormones from the cell surface to the nucleus and functions as an oncoprotein in many types of cancers, yet little is known about how it achieves its native folded state within the cell. Here we show that Stat3 is a novel substrate of the ring-shaped hetero-oligomeric eukaryotic chaperonin, TRiC/CCT, which contributes to its biosynthesis and activity in vitro and in vivo. TRiC binding to Stat3 was mediated, at least in part, by TRiC subunit CCT3. Stat3 binding to TRiC mapped predominantly to the ß-strand rich, DNA-binding domain of Stat3. Notably, enhancing Stat3 binding to TRiC by engineering an additional TRiC-binding domain from the von Hippel-Lindau protein (vTBD), at the N-terminus of Stat3, further increased its affinity for TRiC as well as its function, as determined by Stat3's ability to bind to its phosphotyrosyl-peptide ligand, an interaction critical for Stat3 activation. Thus, Stat3 levels and function are regulated by TRiC and can be modulated by manipulating its interaction with TRiC.


Asunto(s)
Chaperonina con TCP-1/genética , Chaperonina con TCP-1/metabolismo , Pliegue de Proteína , Factor de Transcripción STAT3/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Activación Enzimática , Células Hep G2 , Humanos , Interleucina-6/metabolismo , Ratones , Neoplasias/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño
13.
J Allergy Clin Immunol ; 138(1): 187-199, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26948077

RESUMEN

BACKGROUND: During IgE-mediated immediate hypersensitivity reactions, vascular endothelial cells permeabilize in response to mast cell mediators. We have demonstrated previously that patients and mice with signal transducer and activator of transcription 3 (STAT3) mutations (autosomal dominant hyper-IgE syndrome [AD-HIES]) are partially protected from anaphylaxis. OBJECTIVES: We sought to study the mechanism by which STAT3 contributes to anaphylaxis and determine whether small-molecule inhibition of STAT3 can prevent anaphylaxis. METHODS: Using unaffected and STAT3-inhibited or genetic loss-of-function samples, we performed histamine skin prick tests, investigated the contribution of STAT3 to animal models of anaphylaxis, and measured endothelial cell permeability, gene and protein expression, and histamine receptor-mediated signaling. RESULTS: Although mouse mast cell degranulation was minimally affected by STAT3 blockade, mast cell mediator-induced anaphylaxis was blunted in Stat3 mutant mice with AD-HIES and in wild-type mice subjected to small-molecule STAT3 inhibition. Histamine skin prick test responses were diminished in patients with AD-HIES. Human umbilical vein endothelial cells derived from patients with AD-HIES or treated with a STAT3 inhibitor did not signal properly through Src or cause appropriate dissolution of the adherens junctions made up of the proteins vascular endothelial-cadherin and ß-catenin. Furthermore, we found that diminished STAT3 target microRNA17-92 expression in human umbilical vein endothelial cells from patients with AD-HIES is associated with increased phosphatase and tensin homolog (PTEN) expression, which inhibits Src, and increased E2F transcription factor 1 expression, which regulates ß-catenin cellular dynamics. CONCLUSIONS: These data demonstrate that STAT3-dependent transcriptional activity regulates critical components for the architecture and functional dynamics of endothelial junctions, thus permitting vascular permeability.


Asunto(s)
Anafilaxia/inmunología , Anafilaxia/metabolismo , Permeabilidad Capilar/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Uniones Adherentes/metabolismo , Anafilaxia/diagnóstico , Anafilaxia/genética , Animales , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/genética , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoglobulina E/inmunología , Mediadores de Inflamación/metabolismo , Mastocitos/metabolismo , Ratones , Ratones Noqueados , Mutación , Receptores Histamínicos/inmunología , Receptores Histamínicos/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos , Pruebas Cutáneas , beta Catenina/metabolismo , Familia-src Quinasas/metabolismo
14.
Biophys J ; 110(11): 2377-2385, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27276256

RESUMEN

AML1-ETO is the translational product of a chimeric gene created by the stable chromosome translocation t (8;21)(q22;q22). It causes acute myeloid leukemia (AML) by dysregulating the expression of genes critical for myeloid cell development and differentiation and recently has been reported to bind multiple subunits of the mammalian cytosolic chaperonin TRiC (or CCT), primarily through its DNA binding domain (AML1-175). Through these interactions, TRiC plays an important role in the synthesis, folding, and activity of AML1-ETO. Using single-particle cryo-electron microscopy, we demonstrate here that a folding intermediate of AML1-ETO's DNA-binding domain (AML1-175) forms a stable complex with apo-TRiC. Our structure reveals that AML1-175 associates directly with a specific subset of TRiC subunits in the open conformation.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Western Blotting , Cromatografía en Gel , Microscopía por Crioelectrón , ADN/metabolismo , Compuestos de Oro , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Humanos , Imagenología Tridimensional , Espectrometría de Masas , Nanopartículas del Metal , Dominios Proteicos , Pliegue de Proteína , Multimerización de Proteína , Estabilidad Proteica , Proteína 1 Compañera de Translocación de RUNX1
15.
J Biol Chem ; 290(17): 11177-87, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25787076

RESUMEN

Cachexia occurs in patients with advanced cancers. Despite the adverse clinical impact of cancer-induced muscle wasting, pathways causing cachexia are controversial, and clinically reliable therapies are not available. A trigger of muscle protein loss is the Jak/Stat pathway, and indeed, we found that conditioned medium from C26 colon carcinoma (C26) or Lewis lung carcinoma cells activates Stat3 (p-Stat3) in C2C12 myotubes. We identified two proteolytic pathways that are activated in muscle by p-Stat3; one is activation of caspase-3, and the other is p-Stat3 to myostatin, MAFbx/Atrogin-1, and MuRF-1 via CAAT/enhancer-binding protein δ (C/EBPδ). Using sequential deletions of the caspase-3 promoter and CHIP assays, we determined that Stat3 activation increases caspase-3 expression in C2C12 cells. Caspase-3 expression and proteolytic activity were stimulated by p-Stat3 in muscles of tumor-bearing mice. In mice with cachexia caused by Lewis lung carcinoma or C26 tumors, knock-out of p-Stat3 in muscle or with a small chemical inhibitor of p-Stat3 suppressed muscle mass losses, improved protein synthesis and degradation in muscle, and increased body weight and grip strength. Activation of p-Stat3 stimulates a pathway from C/EBPδ to myostatin and expression of MAFbx/Atrogin-1 and increases the ubiquitin-proteasome system. Indeed, C/EBPδ KO decreases the expression of MAFbx/Atrogin-1 and myostatin, while increasing muscle mass and grip strength. In conclusion, cancer stimulates p-Stat3 in muscle, activating protein loss by stimulating caspase-3, myostatin, and the ubiquitin-proteasome system. These results could lead to novel strategies for preventing cancer-induced muscle wasting.


Asunto(s)
Caquexia/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Caspasa 3/metabolismo , Neoplasias del Colon/metabolismo , Músculo Esquelético/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factor de Transcripción STAT3/metabolismo , Ubiquitina/metabolismo , Animales , Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Caquexia/genética , Caquexia/patología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Caspasa 3/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Miostatina/genética , Miostatina/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Proteolisis , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Factor de Transcripción STAT3/genética , Proteínas de Motivos Tripartitos , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
16.
J Cell Sci ; 126(Pt 11): 2446-58, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23572509

RESUMEN

In this report, we have shown that miR146b promotes the maintenance of pregnancy-derived mammary luminal alveolar progenitors. MiR146b expression was significantly higher in the mammary glands of pregnant and lactating mice than in virgin mice. Furthermore, miR146b levels were significantly higher in mouse mammary glands exposed to the sex hormones, estrogen and progesterone, compared with those of untreated control animals. Pregnancy-derived primary mouse mammary epithelial cells in which miR146b was knocked down showed a significant reduction in the number of hollow acinar organoid structures formed on three-dimensional Matrigel and in ß-casein expression. This demonstrates that miR146b promotes the maintenance of pregnancy-derived mammary luminal alveolar progenitors. It has been shown that mouse mammary luminal progenitors give rise to hollow organoid structures, whereas solid organoid structures are derived from stem cells. Among several miR146b targets, miR146b knockdown resulted in preferential STAT3ß overexpression. In the primary mouse mammary epithelial cells, overexpression of STAT3ß isoform caused mammary epithelial cell death and a significant reduction in ß-casein mRNA expression. Therefore, we conclude that during pregnancy miR146b is involved in luminal alveolar progenitor cell maintenance, at least partially, by regulating STAT3ß.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , MicroARNs/metabolismo , Embarazo/fisiología , Células Madre/metabolismo , Animales , Caseínas/biosíntesis , Estrógenos/genética , Estrógenos/metabolismo , Femenino , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Lactancia/fisiología , Glándulas Mamarias Animales/citología , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Progesterona/genética , Progesterona/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Madre/citología
17.
Blood ; 121(7): 1083-93, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23243289

RESUMEN

Signal transducer and activator of transcription 3 (Stat3) and Stat5 are critical signaling intermediates that promote survival in myeloid leukemias. We examined Stat3 and Stat5 activation patterns in resting and ligand-stimulated primary samples from pediatric patients with acute myeloid leukemia. Phosphorylated Stats were measured by FACS before and after stimulation with increasing doses of granulocyte-colony stimulating factor or IL-6. We also measured positive and negative regulators of Stat signaling, and we compared the variation in multiple parameters to identify biologic relationships. Levels of constitutively phosphorylated Stats were variable and did not correlate with survival. In terms of induced phospho-Stats, 15 of 139 specimens (11%) phosphorylated Stat3 in response to moderate doses of both granulocyte-colony stimulating factor and IL-6. Compared with groups that were resistant to 1 or both ligands, this pattern of dual sensitivity was associated with a superior outcome, with a 5-year event-free survival of 79% (P = .049) and 5-year overall survival of 100% (P = .006). This study provides important and novel insights into the biology of Stat3 and Stat5 signaling in acute myeloid leukemia. Patterns of ligand sensitivity may be valuable for improving risk identification, and for developing new agents for individualized therapy.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Adolescente , Separación Celular , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Filgrastim , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Lactante , Interleucina-6/farmacología , Masculino , Fosforilación , Pronóstico , Proteínas Recombinantes/farmacología , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Adulto Joven
18.
Stem Cells ; 32(10): 2571-82, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24891218

RESUMEN

In breast cancer, a subset of tumor-initiating cells (TIC) or "cancer stem cells" are thought to be responsible for tumor maintenance, treatment resistance, and disease recurrence. While current breast cancer stem cell markers (e.g., CD44(high) /CD24(low/neg) , ALDH positive) have allowed enrichment for such cells, they are not universally expressed and may actually identify distinct TIC subpopulations in the same tumor. Thus, additional markers of functional stem cells are needed. The STAT3 pathway is a critical regulator of the function of normal stem cells, and evidence is accumulating for its important role in breast cancer stem cells. However, due to the lack of a method for separating live cells based on their level of STAT3 activity, it remains unknown whether STAT3 functions in the cancer stem cells themselves, or in surrounding niche cells, or in both. To approach this question, we constructed a series of lentiviral fluorescent (enhanced green fluorescent protein, EGFP) reporters that enabled flow cytometric enrichment of cells differing in STAT3-mediated transcriptional activity, as well as in vivo/in situ localization of STAT3 responsive cells. Using in vivo claudin-low cell line xenograft models of human breast cancer, we found that STAT3 signaling reporter activity (EGFP(+) ) is associated with a subpopulation of cancer cells enriched for mammosphere-forming efficiency, as well as TIC function in limiting dilution transplantation assays compared to negative or unsorted populations. Our results support STAT3 signaling activity as another functional marker for human breast cancer stem cells thus making it an attractive therapeutic target for stem-cell-directed therapy in some breast cancer subtypes.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Claudinas/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Femenino , Fluorescencia , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lentivirus/metabolismo , Ratones SCID , Modelos Biológicos , Reproducibilidad de los Resultados , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Mol Sci ; 16(11): 26706-20, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26561808

RESUMEN

The folding of newly synthesized proteins and the maintenance of pre-existing proteins are essential in sustaining a living cell. A network of molecular chaperones tightly guides the folding, intracellular localization, and proteolytic turnover of proteins. Many of the key regulators of cell growth and differentiation have been identified as clients of molecular chaperones, which implies that chaperones are potential mediators of oncogenesis. In this review, we briefly provide an overview of the role of chaperones, including HSP70 and HSP90, in cancer. We further summarize and highlight the emerging the role of chaperonin TRiC (T-complex protein-1 ring complex, also known as CCT) in the development and progression of cancer mediated through its critical interactions with oncogenic clients that modulate growth deregulation, apoptosis, and genome instability in cancer cells. Elucidation of how TRiC modulates the folding and function of oncogenic clients will provide strategies for developing novel cancer therapies.


Asunto(s)
Carcinogénesis/genética , Chaperonina con TCP-1/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/química , Neoplasias/genética , Apoptosis , Carcinogénesis/metabolismo , Carcinogénesis/patología , Chaperonina con TCP-1/metabolismo , Progresión de la Enfermedad , Inestabilidad Genómica , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Conformación Proteica , Pliegue de Proteína , Transducción de Señal
20.
Angew Chem Int Ed Engl ; 54(44): 13085-9, 2015 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-26480340

RESUMEN

Nearly 40 % of children with acute myeloid leukemia (AML) suffer relapse arising from chemoresistance, often involving upregulation of the oncoprotein STAT3 (signal transducer and activator of transcription 3). Herein, rhodium(II)-catalyzed, proximity-driven modification identifies the STAT3 coiled-coil domain (CCD) as a novel ligand-binding site, and we describe a new naphthalene sulfonamide inhibitor that targets the CCD, blocks STAT3 function, and halts its disease-promoting effects in vitro, in tumor growth models, and in a leukemia mouse model, validating this new therapeutic target for resistant AML.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Naftalenos/farmacología , Rodio/química , Factor de Transcripción STAT3/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Sitios de Unión/efectos de los fármacos , Catálisis , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Naftalenos/química , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Factor de Transcripción STAT3/metabolismo , Relación Estructura-Actividad , Sulfonamidas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA