Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Cell Rep ; 37(6): 109974, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758313

RESUMEN

The mechanisms of Myc-driven liver tumorigenesis are inadequately understood. Herein we show that Myc-driven hepatocellular carcinoma (HCC) is dramatically aggravated in mice with hepatocyte-specific Ptpn11/Shp2 deletion. However, Myc-induced tumors develop selectively from the rare Shp2-positive hepatocytes in Shp2-deficent liver, and Myc-driven oncogenesis depends on an intact Ras-Erk signaling promoted by Shp2 to sustain Myc stability. Despite a stringent requirement of Shp2 cell autonomously, Shp2 deletion induces an immunosuppressive environment, resulting in defective clearance of tumor-initiating cells and aggressive tumor progression. The basal Wnt/ß-catenin signaling is upregulated in Shp2-deficient liver, which is further augmented by Myc transfection. Ablating Ctnnb1 suppresses Myc-induced HCC in Shp2-deficient livers, revealing an essential role of ß-catenin. Consistently, Myc overexpression and CTNNB1 mutations are frequently co-detected in HCC patients with poor prognosis. These data elucidate complex mechanisms of liver tumorigenesis driven by cell-intrinsic oncogenic signaling in cooperation with a tumor-promoting microenvironment generated by disrupting the specific oncogenic pathway.


Asunto(s)
Carcinoma Hepatocelular/patología , Hepatocitos/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Análisis de la Célula Individual/métodos , Microambiente Tumoral , Animales , Biomarcadores de Tumor , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Hepatocitos/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-myc/genética , Transcriptoma , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
3.
Oncogene ; 40(4): 746-762, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33247204

RESUMEN

Leukemias are routinely sub-typed for risk/outcome prediction and therapy choice using acquired mutations and chromosomal rearrangements. Down syndrome acute lymphoblastic leukemia (DS-ALL) is characterized by high frequency of CRLF2-rearrangements, JAK2-mutations, or RAS-pathway mutations. Intriguingly, JAK2 and RAS-mutations are mutually exclusive in leukemic sub-clones, causing dichotomy in therapeutic target choices. We prove in a cell model that elevated CRLF2 in combination with constitutionally active JAK2 is sufficient to activate wtRAS. On primary clinical DS-ALL samples, we show that wtRAS-activation is an obligatory consequence of mutated/hyperphosphorylated JAK2. We further prove that CRLF2-ligand TSLP boosts the direct binding of active PTPN11 to wtRAS, providing the molecular mechanism for the wtRAS activation. Pre-inhibition of RAS or PTPN11, but not of PI3K or JAK-signaling, prevented TSLP-induced RAS-GTP boost. Cytotoxicity assays on primary clinical DS-ALL samples demonstrated that, regardless of mutation status, high-risk leukemic cells could only be killed using RAS-inhibitor or PTPN11-inhibitor, but not PI3K/JAK-inhibitors, suggesting a unified treatment target for up to 80% of DS-ALL. Importantly, protein activities-based principal-component-analysis multivariate clusters analyzed for independent outcome prediction using Cox proportional-hazards model showed that protein-activity (but not mutation-status) was independently predictive of outcome, demanding a paradigm-shift in patient-stratification strategy for precision therapy in high-risk ALL.


Asunto(s)
Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas ras/fisiología , Animales , Citocinas/fisiología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/fisiología , Ratones , Fosfatidilinositol 3-Quinasas/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Receptores de Citocinas/genética , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología , Proteínas ras/antagonistas & inhibidores , Proteínas ras/genética
4.
Sci Rep ; 10(1): 18530, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33116231

RESUMEN

SHP2 is a protein tyrosine phosphatase (PTP) involved in multiple signaling pathways. Mutations of SHP2 can result in Noonan syndrome or pediatric malignancies. Inhibition of wild-type SHP2 represents a novel strategy against several cancers. SHP2 is activated by binding of a phosphopeptide to the N-SH2 domain of SHP2, thereby favoring dissociation of the N-SH2 domain and exposing the active site on the PTP domain. The conformational transitions controlling ligand affinity and PTP dissociation remain poorly understood. Using molecular simulations, we revealed an allosteric interaction restraining the N-SH2 domain into a SHP2-activating and a stabilizing state. Only ligands selecting for the activating N-SH2 conformation, depending on ligand sequence and binding mode, are effective activators. We validate the model of SHP2 activation by rationalizing modified basal activity and responsiveness to ligand stimulation of several N-SH2 variants. This study provides mechanistic insight into SHP2 activation and may open routes for SHP2 regulation.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Simulación por Computador , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación/genética , Unión Proteica/genética , Conformación Proteica , Dominios Proteicos/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal/genética , Tirosina/metabolismo , Dominios Homologos src/genética
5.
J Pharmacol Sci ; 144(3): 139-146, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32921395

RESUMEN

SHP2 is a non-receptor protein tyrosine phosphatase encoded by the PTPN11 gene in human. Clinically, SHP2 has been identified as a causal factor of several diseases, such as Noonan syndrome, LEOPARD syndrome as well as myeloid malignancies. Interestingly, both loss-of-function and gain-of-function mutations occur in the PTPN11 gene. Analyses by biochemical and cell biological means as well as probing with small molecule compounds have demonstrated that SHP2 has both phosphatase-dependent and independent functions. In comparison with its phosphatase activity, the non-phosphatase-like function of SHP2 has not been well introduced or summarized. This review mainly focuses on the phosphatase-independent functions and its regulation by small molecule compounds as well as their use for disease therapy.


Asunto(s)
Monoéster Fosfórico Hidrolasas , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Cerebrósidos , Depsipéptidos , Mutación con Ganancia de Función , Humanos , Síndrome LEOPARD/genética , Mutación con Pérdida de Función , Terapia Molecular Dirigida , Síndrome de Noonan/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Piperidinas , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Pirimidinas , Transducción de Señal/genética
6.
Dev Genes Evol ; 230(3): 227-238, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32198667

RESUMEN

Across the Bilateria, FGF/FGFR signaling is critical for normal development, and in both Drosophila and vertebrates, docking proteins are required to connect activated FGFRs with downstream pathways. While vertebrates use Frs2 to dock FGFR to the RAS/MAPK or PI3K pathways, the unrelated protein, downstream of FGFR (Dof/stumps/heartbroken), fulfills the corresponding function in Drosophila. To better understand the evolution of the signaling pathway downstream of FGFR, the available sequence databases were screened to identify Frs2, Dof, and other key pathway components in phyla that diverged early in animal evolution. While Frs2 homologues were detected only in members of the Bilateria, canonical Dof sequences (containing Dof, ankyrin, and SH2/SH3 domains) were present in cnidarians as well as bilaterians (but not in other animals or holozoans), correlating with the appearance of FGFR. Although these data suggested that Dof coupling might be ancestral, gene expression analysis in the cnidarian Hydra revealed that Dof is not upregulated in the zone of strong FGFRa and FGFRb expression at the bud base, where FGFR signaling controls detachment. In contrast, transcripts encoding other, known elements of FGFR signaling in Bilateria, namely the FGFR adaptors Grb2 and Crkl, which are acting downstream of Dof (and Frs2), as well as the guanyl nucleotide exchange factor Sos, and the tyrosine phosphatase Csw/Shp2, were strongly upregulated at the bud base. Our expression analysis, thus, identified transcriptional upregulation of known elements of FGFR signaling at the Hydra bud base indicating a highly conserved toolkit. Lack of transcriptional Dof upregulation raises the interesting question, whether Hydra FGFR signaling requires either of the docking proteins known from Bilateria.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Evolución Biológica , Hydra/genética , Hydra/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Animales , Cnidarios/genética , Cnidarios/metabolismo , Proteína Adaptadora GRB2/fisiología , Regulación del Desarrollo de la Expresión Génica , Filogenia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Transducción de Señal , Proteínas Son Of Sevenless/fisiología
7.
Biochemistry (Mosc) ; 85(1): 108-118, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32079522

RESUMEN

The MAPK (RAS/BRAF/MEK/ERK) signaling pathway is a kinase cascade involved in the regulation of cell proliferation, differentiation, and survival in response to external stimuli. The V600E mutation in the BRAF gene has been detected in various tumors, resulting in a 500-fold increase in BRAF kinase activity. However, monotherapy with selective BRAF V600E inhibitors often leads to reactivation of MAPK signaling cascade and emergence of drug resistance. Therefore, new targets are being developed for the inhibition of components of the aberrantly activated cascade. It was recently discovered that resistance to BRAF V600E inhibitors may be associated with the activity of the tyrosine phosphatase SHP-2 encoded by the PTPN11 gene. In this paper, we analyzed transcriptional effects of PTPN11 gene knockdown and selective suppression of BRAF V600E in a model of thyroid follicular epithelium. We found that the siRNA-mediated knockdown of PTPN11 after vemurafenib treatment prevented an increase in the expression CCNA1 and NOTCH4 genes involved in the formation of drug resistance of tumors. On the other hand, downregulation of PTPN11 expression blocked the transcriptional activation of genes (p21, p15, p16, RB1, and IGFBP7) involved in cell cycle regulation and oncogene-induced senescence in response to BRAF V600E expression. Therefore, it can be assumed that SHP-2 participates not only in emergence of drug resistance in cancer cells, but also in oncogene-induced cell senescence.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Células Epiteliales Tiroideas , Neoplasias de la Tiroides/metabolismo , Ciclo Celular , Línea Celular , Senescencia Celular , Resistencia a Antineoplásicos/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Células Epiteliales Tiroideas/citología , Células Epiteliales Tiroideas/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Vemurafenib/uso terapéutico
8.
Front Immunol ; 10: 2468, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708921

RESUMEN

Somewhat counterintuitively, the tyrosine phosphatase SHP-2 (SH2 domain-containing protein tyrosine phosphatase-2) is crucial for the activation of extracellular signal-regulated kinase (ERK) downstream of various growth factor receptors, thereby exerting essential developmental functions. This phosphatase also deploys proto-oncogenic functions and specific inhibitors have recently been developed. With respect to the immune system, the role of SHP-2 in the signaling of cytokines relevant for myelopoiesis and myeloid malignancies has been intensively studied. The function of this phosphatase downstream of cytokines important for lymphocytes is less understood, though multiple lines of evidence suggest its importance. In addition, SHP-2 has been proposed to mediate the suppressive effects of inhibitory receptors (IRs) that sustain a dysfunctional state in anticancer T cells. Molecules involved in IR signaling are of potential pharmaceutical interest as blockade of these inhibitory circuits leads to remarkable clinical benefit. Here, we discuss the dichotomy in the functions ascribed to SHP-2 downstream of cytokine receptors and IRs, with a focus on T and NK lymphocytes. Further, we highlight the importance of broadening our understanding of SHP-2's relevance in lymphocytes, an essential step to inform on side effects and unanticipated benefits of its therapeutic blockade.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Receptores de Citocinas/fisiología , Receptores KIR/fisiología , Linfocitos T/inmunología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Células Asesinas Naturales/inmunología , Neoplasias/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Transducción de Señal/fisiología
9.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 144-152, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30026077

RESUMEN

Cancer cells depend on a supportive niche (the tumor microenvironment) that promotes tumor cell survival while protecting the malignant cells from therapeutic challenges and the host's defense systems. Cancer cells and the support cells in the tumor microenvironment communicate via cytokines/chemokines, cell:cell contact, or alterations in the metabolic state of the niche (e.g. hypoxia) that promote growth and survival of the tumor cell, influence metastasis, and defeat immune surveillance. These signaling pathways involve dysregulation of not only protein kinases but also protein phosphatases as normal signal transduction processes require both activation and deactivation. For instance, aberrant receptor signaling can result from constitutive activation of a tyrosine kinase such as FLT3 or inactivation of a tyrosine protein phosphatase such as SHP-2 (PTPN11). Activation of serine/threonine kinases such as AKT and ERK are often observed during the development of drug resistance while genomic and non-genomic suppression of serine/threonine protein phosphatases such as PP2A achieve similar results. It is fairly clear that the various protein phosphatases will impact processes that support drug resistance. Of growing interest is the emerging model whereby the support cells in the tumor microenvironment actually serve as drivers of tumorigenesis. This phenomenon has been most prominently observed in osteoblast cells in leukemic niches. At least one protein phosphatase, PTPN11, has emerged as a critical driver of this process in juvenile myelomonocytic leukemia. This review will cover the role of various serine/threonine and tyrosine protein phosphatases in processes that are central to tumor microenvironment function.


Asunto(s)
Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas Fosfatasas/fisiología , Microambiente Tumoral/fisiología , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia Mieloide/genética , Neoplasias/metabolismo , Neoplasias/fisiopatología , Fosforilación , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
10.
Integr Biol (Camb) ; 9(7): 584-594, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28548171

RESUMEN

High shear stress (SS) causes local changes around arterial bifurcations, which are common sites for cerebral aneurysms. High SS and SS spatial gradient (SSG) are thought to play important roles in the pathology of cerebral aneurysms. However, whether SS and SSG independently affect the function and morphology of vascular endothelial cells (ECs) exposed to fluid flow remains unclear. This study evaluated the morphology of ECs exposed to various SS and SSG combinations. Confluent ECs were exposed to a SS of 2-60 Pa and a uniform SSG of 0, 5, 10, or 15 Pa mm-1 for 24 h. Although ECs exposed to lower levels of SS/SSG were not oriented or elongated in the direction of flow, they began to exhibit orientation, elongation, and development of actin stress fibers under the conditions of SS with a SSG when the SS exceeded a threshold value depending on the magnitude of SSG. Using a simplified computational model, we found that the presence of a SSG affects the strain field in ECs, resulting in a morphological response. SS combined with a SSG can alter the localization of SS mechano-sensing proteins along the strain field as a result of shear flow. Our results suggest that the magnitude of the relationship between SS and SSG plays an important role in regulating morphological changes in ECs in response to fluid flow by regulating EC polarity.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/fisiología , Citoesqueleto de Actina/fisiología , Fenómenos Biomecánicos , Forma de la Célula , Células Cultivadas , Humanos , Hidrodinámica , Aneurisma Intracraneal/etiología , Aneurisma Intracraneal/patología , Aneurisma Intracraneal/fisiopatología , Mecanotransducción Celular , Modelos Biológicos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Fibras de Estrés , Estrés Mecánico
11.
PLoS One ; 12(5): e0177334, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28481957

RESUMEN

Shp2 played an important role in cigarette-smoke-mediated inflammation, surfactant homeostasis and asthmatic airway remodeling. However, whether shp2 plays a key role in epithelium-associated allergic reaction is still unknown. In this study, LPS and OVA were observed to induce the production of IL-25 in bronchial epithelial cells in vitro via the activation of MAPK p38 and JNK. Furthermore, blockage of Shp2 by its specific inhibitor PHPS1 or by siRNA-mediated depletion was found to reduce the production of IL-25 in epithelial cells as well as the up-regulated LPS-triggered activation of JNK but not p38. To confirm the role of intra-bronchial epithelial Shp2 in OVA-induced allergic reaction, we generated CC10-rtTA/(tetO)7-Cre/Shp2f/f mice, where Shp2 was conditionally knocked out in bronchial epithelial cells. Surprisingly, specific deletion of Shp2 in bronchial epithelial cells showed a mild but insignificant effect on the expressions of epithelium-derived cytokines as well as TH2 and TH17 polarization following allergen-induced murine airway inflammation. Collectively, our data suggested that deletion of Shp2 impaired IL-25 production in bronchial epithelial cells in vitro, but might yet have minor influence on OVA-induced allergic reaction in vivo.


Asunto(s)
Asma/metabolismo , Bronquios/metabolismo , Interleucinas/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Animales , Bronquios/citología , Línea Celular , Activación Enzimática , Células Epiteliales/metabolismo , Femenino , Humanos , Técnicas In Vitro , Inflamación/metabolismo , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , ARN Interferente Pequeño/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Int J Cancer ; 140(5): 1159-1172, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27859216

RESUMEN

PTPN11 mutation, a RAS signaling pathway mutation, is associated with MLL translocations in acute leukemia. A girl with MLL/AF10 AML was found to carry PTPN11G503A . To study the impact of PTPN11G503A cooperating with MLL/AF10 on leukemogenesis, we established a retroviral transduction/transplantation mouse model. Compared to the MLL/AF10(OM-LZ) leukemia cells harboring PTPN11wt , the cells harboring PTPN11G503A were hypersensitive to GM-CSF and IL3, and more resistant to death upon treatment with daunorubicin but sensitive to cytarabine. The cells harboring PTPN11G503A autonomously differentiated into macrophages (1.8%) in the medium containing IL3. Further studies showed that the cells had an elevated (∼2.9-fold) Csf1 transcription level and secreted more (∼4.5-fold) M-CSF to the medium which can stimulate monocyte/macrophage differentiation of BM cells. Mice transplanted with the cells harboring PTPN11G503A had a higher concentration of M-CSF in plasma. When mixed with the MLL/AF10(OM-LZ) leukemia cells harboring PTPN11wt , the cells harboring PTPN11G503A had an increased competitive engraftment and clonal expansion in the BM and spleen of recipient mice, although no competitive growth advantage was observed in the in vitro co-culturing assays. The mice transplanted with the MLL/AF10(OM-LZ) cells harboring PTPN11wt developed myelomonocytic leukemia, while those transplanted with the cells harboring PTPN11G503A -induced monocytic leukemia in a shorter latency. Our results demonstrated that addition of PTPN11G503A to MLL/AF10 affected cell proliferation, chemo-resistance, differentiation, in vivo BM recruitment/clonal expansion and accelerated disease progression.


Asunto(s)
Transformación Celular Neoplásica/genética , Leucemia Monocítica Aguda/etiología , Leucemia Mielomonocítica Aguda/etiología , Mutación Missense , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Fusión Oncogénica/fisiología , Mutación Puntual , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Animales , Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , Citarabina/uso terapéutico , Daunorrubicina/uso terapéutico , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Activación Enzimática/genética , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Lactante , Interleucina-3/farmacología , Leucemia Monocítica Aguda/genética , Leucemia Mielomonocítica Aguda/genética , Factor Estimulante de Colonias de Macrófagos/sangre , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Quimera por Radiación , Transducción Genética , Células Tumorales Cultivadas/trasplante
13.
Leukemia ; 31(6): 1415-1422, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27840422

RESUMEN

Activating mutations, such as E76K and D61Y, in PTPN11 (SHP2), a protein tyrosine phosphatase implicated in multiple cell signaling processes, are associated with 35% of patients with juvenile myelomonocytic leukemia (JMML), an aggressive childhood myeloproliferative neoplasm (MPN). Here we show that the interaction between leukemia-associated mutant Shp2 and Gab2, a scaffolding protein important for cytokine-induced PI3K/Akt signaling, was enhanced, and that the mTOR pathway was elevated in Ptpn11E76K/+ leukemic cells. Importantly, MPN induced by the Ptpn11E76K/+ mutation was markedly attenuated in Ptpn11E76K/+/Gab2-/- double mutant mice-overproduction of myeloid cells was alleviated, splenomegaly was diminished and myeloid cell infiltration in nonhematopoietic organs was decreased in these double mutants. Excessive myeloid differentiation of stem cells was also normalized by depletion of Gab2. Acute leukemia progression of MPN was reduced in the double mutant mice and, as such, their survival was much prolonged. Furthermore, treatment of Ptpn11E76K/+ mice with Rapamycin, a specific and potent mTOR inhibitor, mitigated MPN phenotypes. Collectively, this study reveals an important role of the Gab2/PI3K/mTOR pathway in mediating the pathogenic signaling of the PTPN11 gain-of-function mutations and a therapeutic potential of Rapamycin for PTPN11 mutation-associated JMML.


Asunto(s)
Mutación , Trastornos Mieloproliferativos/etiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfoproteínas/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Antibióticos Antineoplásicos/farmacología , Ratones , Ratones Noqueados , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfoproteínas/genética , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética
14.
Cancer Res ; 76(21): 6396-6409, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27803106

RESUMEN

Little is known about the factors that regulate the asymmetric division of cancer stem-like cells (CSC). Here, we demonstrate that EGFL6, a stem cell regulatory factor expressed in ovarian tumor cells and vasculature, regulates ALDH+ ovarian CSC. EGFL6 signaled at least in part via the oncoprotein SHP2 with concomitant activation of ERK. EGFL6 signaling promoted the migration and asymmetric division of ALDH+ ovarian CSC. As such, EGFL6 increased not only tumor growth but also metastasis. Silencing of EGFL6 or SHP2 limited numbers of ALDH+ cells and reduced tumor growth, supporting a critical role for EGFL6/SHP2 in ALDH+ cell maintenance. Notably, systemic administration of an EGFL6-neutralizing antibody we generated restricted tumor growth and metastasis, specifically blocking ovarian cancer cell recruitment to the ovary. Together, our results offer a preclinical proof of concept for EGFL6 as a novel therapeutic target for the treatment of ovarian cancer. Cancer Res; 76(21); 6396-409. ©2016 AACR.


Asunto(s)
Aldehído Deshidrogenasa/análisis , División Celular , Glicoproteínas de Membrana/fisiología , Neoplasias Ováricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Humanos , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Neoplasias Ováricas/enzimología , Pronóstico , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología
15.
Oncotarget ; 7(40): 65676-65695, 2016 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-27582544

RESUMEN

A major risk factor of developing colorectal cancer (CRC) is the presence of chronic inflammation in the colon. In order to understand how inflammation contributes to CRC development, the present study focused on SHP-2, a tyrosine phosphatase encoded by PTPN11 gene in which polymorphisms have been shown to be markers of colitis susceptibility. Conversely, gain-of-function mutations in PTPN11 gene (E76 residue) have been found in certain sporadic CRC. Results shown herein demonstrate that SHP-2 expression was markedly increased in sporadic human adenomas but not in advanced colorectal tumors. SHP-2 silencing inhibited proliferative, invasive and tumoral properties of both intestinal epithelial cells (IECs) transformed by oncogenic KRAS and of human CRC cells. IEC-specific expression of a SHP-2E76K activated mutant in mice was not sufficient to induce tumorigenesis but markedly promoted tumor growth under the ApcMin/+ background. Conversely, mice with a conditional deletion of SHP-2 in IECs developed colitis-associated adenocarcinomas with age, associated with sustained activation of Wnt/ß-catenin, NFκB and STAT3 signalings in the colonic mucosae. Moreover, SHP-2 epithelial deficiency considerably increased tumor load in ApcMin/+ mice, shifting tumor incidence toward the colon. Overall, these results reveal that SHP-2 can exert opposing functions in the large intestine: it can promote or inhibit tumorigenesis depending of the inflammatory context.


Asunto(s)
Adenocarcinoma/prevención & control , Biomarcadores de Tumor/metabolismo , Colitis/complicaciones , Neoplasias Colorrectales/prevención & control , Neoplasias Intestinales/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma/etiología , Adenocarcinoma/patología , Animales , Apoptosis , Carcinogénesis , Proliferación Celular , Colitis/fisiopatología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , Femenino , Humanos , Neoplasias Intestinales/etiología , Neoplasias Intestinales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pronóstico , Transducción de Señal , Células Tumorales Cultivadas
16.
Proc Natl Acad Sci U S A ; 113(4): 984-9, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26755576

RESUMEN

Gain-of-function (GOF) mutations of protein tyrosine phosphatase nonreceptor type 11 Ptpn11 (Shp2), a protein tyrosine phosphatase implicated in multiple cell signaling pathways, are associated with childhood leukemias and solid tumors. The underlying mechanisms are not fully understood. Here, we report that Ptpn11 GOF mutations disturb mitosis and cytokinesis, causing chromosomal instability and greatly increased susceptibility to DNA damage-induced malignancies. We find that Shp2 is distributed to the kinetochore, centrosome, spindle midzone, and midbody, all of which are known to play critical roles in chromosome segregation and cytokinesis. Mouse embryonic fibroblasts with Ptpn11 GOF mutations show a compromised mitotic checkpoint. Centrosome amplification and aberrant mitosis with misaligned or lagging chromosomes are significantly increased in Ptpn11-mutated mouse and patient cells. Abnormal cytokinesis is also markedly increased in these cells. Further mechanistic analyses reveal that GOF mutant Shp2 hyperactivates the Polo-like kinase 1 (Plk1) kinase by enhancing c-Src kinase-mediated tyrosine phosphorylation of Plk1. This study provides novel insights into the tumorigenesis associated with Ptpn11 GOF mutations and cautions that DNA-damaging treatments in Noonan syndrome patients with germ-line Ptpn11 GOF mutations could increase the risk of therapy-induced malignancies.


Asunto(s)
Daño del ADN , Mitosis , Neoplasias/etiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Animales , Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas/metabolismo , Quinasa Tipo Polo 1
17.
J Hepatol ; 63(3): 651-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25865556

RESUMEN

BACKGROUND & AIMS: We have previously reported that Shp2, a tyrosine phosphatase previously known as a pro-leukemogenic molecule, suppresses the initiation of hepatocellular carcinoma (HCC). However, the role of Shp2 in HCC progression remains obscure. METHODS: Shp2 expression was determined in human HCC using real-time PCR, immunoblotting and immunohistochemistry. Clinical significance of Shp2 expression was analyzed in 301 HCC tissues with clinico-pathological characteristics and follow-up information. Short hairpin RNA was utilized to investigate the function of Shp2 in hepatoma cell behavior. Role of Shp2 in HCC progression was monitored through nude mice xenograft assay. Kinase activity assay and co-immunoprecipitation were used for mechanism analysis. RESULTS: Elevated expression of Shp2 was detected in 65.9% (394/598) of human HCCs, and its levels were even higher in metastasized foci. Overexpression of Shp2 correlated well with the malignant clinico-pathological characteristics of HCC and predicted the poor prognosis of patients. Interference of Shp2 expression suppressed the proliferation of hepatoma cells in vitro and inhibited the growth of HCC xenografts in vivo. Down-regulation of Shp2 attenuated the adhesion and migration of hepatoma cells and diminished metastasized HCC formation in mice. Our data demonstrated that Shp2 promotes HCC growth and metastasis by coordinately activating Ras/Raf/Erk pathway and PI3-K/Akt/mTOR cascade. Moreover, down-regulation of Shp2 enhanced the sensitivity of hepatoma cells upon sorafenib treatment, and patients with low Shp2 expression exhibited superior prognosis to sorafenib. CONCLUSIONS: Shp2 promotes the progression of HCC and may serve as a prognostic biomarker for patients.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Animales , Carcinoma Hepatocelular/mortalidad , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Humanos , Neoplasias Hepáticas/mortalidad , Sistema de Señalización de MAP Quinasas , Ratones , Niacinamida/análogos & derivados , Niacinamida/farmacología , Compuestos de Fenilurea/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Pronóstico , Sorafenib , Quinasas raf/fisiología
18.
FASEB J ; 29(5): 1635-45, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25593124

RESUMEN

Genes that regulate osteoclast (OC) development and function in both physiologic and disease conditions remain incompletely understood. Shp2 (the Src homology-2 domain containing protein tyrosine phosphatase 2), a ubiquitously expressed cytoplasmic protein tyrosine phosphatase, is implicated in regulating M-CSF and receptor activator of nuclear factor-κB ligand (RANKL)-evoked signaling; its role in osteoclastogenesis and bone homeostasis, however, remains unknown. Using a tissue-specific gene knockout approach, we inactivated Shp2 expression in murine OCs. Shp2 mutant mice are phenotypically osteopetrotic, featuring a marked increase of bone volume (BV)/total volume (TV) (+42.8%), trabeculae number (Tb.N) (+84.1%), structure model index (+119%), and a decrease of trabecular thickness (Tb.Th) (-34.1%) and trabecular spacing (Tb.Sp) (-41.0%). Biochemical analyses demonstrate that Shp2 is required for RANKL-induced formation of giant multinucleated OCs by up-regulating the expression of nuclear factor of activated T cells, cytoplasmic 1 (Nfatc1), a master transcription factor that is indispensable for terminal OC differentiation. Shp2 deletion, however, has minimal effect on M-CSF-dependent survival and proliferation of OC precursors. Instead, its deficiency aborts the fusion of OC precursors and formation of multinucleated OCs and decreases bone matrix resorption. Moreover, pharmacological intervention of Shp2 is sufficient to prevent preosteoclast fusion in vitro. These findings uncover a novel mechanism through which Shp2 regulates osteoclastogenesis by promoting preosteoclast fusion. Shp2 or its signaling partners could potentially serve as pharmacological targets to regulate the population of OCs locally and/or systematically, and thus treat OC-related diseases, such as periprosthetic osteolysis and osteoporosis.


Asunto(s)
Médula Ósea/crecimiento & desarrollo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Osteopetrosis/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Ligando RANK/metabolismo , Animales , Apoptosis , Western Blotting , Médula Ósea/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción NFATC/genética , Osteoclastos/metabolismo , Osteopetrosis/metabolismo , Ligando RANK/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
19.
J Biol Chem ; 290(7): 3894-909, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25538234

RESUMEN

Macrophages are vital to innate immunity and express pattern recognition receptors and integrins for the rapid detection of invading pathogens. Stimulation of Dectin-1 and complement receptor 3 (CR3) activates Erk- and Akt-dependent production of reactive oxygen species (ROS). Shp2, a protein-tyrosine phosphatase encoded by Ptpn11, promotes activation of Ras-Erk and PI3K-Akt and is crucial for hematopoietic cell function; however, no studies have examined Shp2 function in particulate-stimulated ROS production. Maximal Dectin-1-stimulated ROS production corresponded kinetically to maximal Shp2 and Erk phosphorylation. Bone marrow-derived macrophages (BMMs) from mice with a conditionally deleted allele of Ptpn11 (Shp2(flox/flox);Mx1Cre+) produced significantly lower ROS levels compared with control BMMs. Although YFP-tagged phosphatase dead Shp2-C463A was strongly recruited to the early phagosome, its expression inhibited Dectin-1- and CR3-stimulated phospho-Erk and ROS levels, placing Shp2 phosphatase function and Erk activation upstream of ROS production. Further, BMMs expressing gain of function Shp2-D61Y or Shp2-E76K and peritoneal exudate macrophages from Shp2D61Y/+;Mx1Cre+ mice produced significantly elevated levels of Dectin-1- and CR3-stimulated ROS, which was reduced by pharmacologic inhibition of Erk. SIRPα (signal regulatory protein α) is a myeloid inhibitory immunoreceptor that requires tyrosine phosphorylation to exert its inhibitory effect. YFP-Shp2C463A-expressing cells have elevated phospho-SIRPα levels and an increased Shp2-SIRPα interaction compared with YFP-WT Shp2-expressing cells. Collectively, these findings indicate that Shp2 phosphatase function positively regulates Dectin-1- and CR3-stimulated ROS production in macrophages by dephosphorylating and thus mitigating the inhibitory function of SIRPα and by promoting Erk activation.


Asunto(s)
Macrófagos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio/fisiología , Animales , Western Blotting , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Integrasas/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Noqueados , Fagocitosis , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Tirosina/metabolismo
20.
Adv Exp Med Biol ; 846: 163-88, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25472538

RESUMEN

Prolactin (PRL) is a polypeptide hormone/cytokine mainly synthesized by the lactotrophic cells of the adenohypophysis. In addition to the best-known role in mammary gland development and the functional differentiation of its epithelium, PRL is involved in regulation of multiple physiological processes in higher organisms contributing to their homeostasis. PRL has been also associated with pathology, including breast cancer. Therefore, it is relevant to determine the molecular mechanisms by which PRL controls cellular functions. Here, we analyze the role of Src family kinases (SFKs) in the intracellular signaling pathways controlled by PRL in several model systems. The data show that SFKs are essential components in transmitting signals upon PRL receptor stimulation, as they control activation of Jak2/Stat5 and other routes that regulate PRL cellular responses.


Asunto(s)
Prolactina/metabolismo , Familia-src Quinasas/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Activación Enzimática , Humanos , Janus Quinasa 2/metabolismo , Quinasas Janus/fisiología , Prolactina/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Receptores de Prolactina/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...