Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 69(1): 87-99.e7, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29249655

RESUMEN

Loss of LKB1 is associated with increased metastasis and poor prognosis in lung cancer, but the development of targeted agents is in its infancy. Here we report that a glutaminolytic enzyme, glutamate dehydrogenase 1 (GDH1), upregulated upon detachment via pleomorphic adenoma gene 1 (PLAG1), provides anti-anoikis and pro-metastatic signals in LKB1-deficient lung cancer. Mechanistically, the GDH1 product α-KG activates CamKK2 by enhancing its substrate AMPK binding, which contributes to energy production that confers anoikis resistance. The effect of GDH1 on AMPK is evident in LKB1-deficient lung cancer, where AMPK activation predominantly depends on CamKK2. Targeting GDH1 with R162 attenuated tumor metastasis in patient-derived xenograft model and correlation studies in lung cancer patients further validated the clinical relevance of our finding. Our study provides insight into the molecular mechanism by which GDH1-mediated metabolic reprogramming of glutaminolysis mediates lung cancer metastasis and offers a therapeutic strategy for patients with LKB1-deficient lung cancer.


Asunto(s)
Anoicis/fisiología , Proteínas de Unión al ADN/metabolismo , Glutamato Deshidrogenasa/metabolismo , Neoplasias Pulmonares/patología , Proteínas Serina-Treonina Quinasas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Células A549 , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Línea Celular Tumoral , Activación Enzimática/fisiología , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias , Trasplante Heterólogo
2.
Nature ; 539(7628): 304-308, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27783593

RESUMEN

Germline activating mutations of the protein tyrosine phosphatase SHP2 (encoded by PTPN11), a positive regulator of the RAS signalling pathway, are found in 50% of patients with Noonan syndrome. These patients have an increased risk of developing leukaemia, especially juvenile myelomonocytic leukaemia (JMML), a childhood myeloproliferative neoplasm (MPN). Previous studies have demonstrated that mutations in Ptpn11 induce a JMML-like MPN through cell-autonomous mechanisms that are dependent on Shp2 catalytic activity. However, the effect of these mutations in the bone marrow microenvironment remains unclear. Here we report that Ptpn11 activating mutations in the mouse bone marrow microenvironment promote the development and progression of MPN through profound detrimental effects on haematopoietic stem cells (HSCs). Ptpn11 mutations in mesenchymal stem/progenitor cells and osteoprogenitors, but not in differentiated osteoblasts or endothelial cells, cause excessive production of the CC chemokine CCL3 (also known as MIP-1α), which recruits monocytes to the area in which HSCs also reside. Consequently, HSCs are hyperactivated by interleukin-1ß and possibly other proinflammatory cytokines produced by monocytes, leading to exacerbated MPN and to donor-cell-derived MPN following stem cell transplantation. Remarkably, administration of CCL3 receptor antagonists effectively reverses MPN development induced by the Ptpn11-mutated bone marrow microenvironment. This study reveals the critical contribution of Ptpn11 mutations in the bone marrow microenvironment to leukaemogenesis and identifies CCL3 as a potential therapeutic target for controlling leukaemic progression in Noonan syndrome and for improving stem cell transplantation therapy in Noonan-syndrome-associated leukaemias.


Asunto(s)
Transformación Celular Neoplásica/genética , Microambiente Celular/genética , Células Madre Hematopoyéticas/patología , Leucemia/genética , Leucemia/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Nicho de Células Madre/genética , Animales , Quimiocina CCL3/antagonistas & inhibidores , Quimiocina CCL3/metabolismo , Progresión de la Enfermedad , Células Endoteliales/citología , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-1beta/metabolismo , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/metabolismo , Leucemia Mielomonocítica Juvenil/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Monocitos/metabolismo , Mutación , Síndrome de Noonan/genética , Síndrome de Noonan/metabolismo , Síndrome de Noonan/patología , Osteoblastos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Trasplante de Células Madre
3.
J Mol Cell Cardiol ; 132: 120-135, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31082397

RESUMEN

Immature phenotypes of cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) limit the utility of these cells in clinical application and basic research. During cardiac development, postnatal cardiomyocytes experience high oxygen tension along with a concomitant downregulation of hypoxia-inducible factor 1α (HIF-1α), leading to increased fatty acid oxidation (FAO). We hypothesized that targeting HIF-1α alone or in combination with other metabolic regulators could promote the metabolic maturation of hiPSC-CMs. We examined the effect of HIF-1α inhibition on the maturation of hiPSC-CMs and investigated a multipronged approach to promote hiPSC-CM maturation by combining HIF-1α inhibition with molecules that target key pathways involved in the energy metabolism. Cardiac spheres of highly-enriched hiPSC-CMs were treated with a HIF-1α inhibitor alone or in combination with an agonist of peroxisome proliferator activated receptor α (PPARα) and three postnatal factors (triiodothyronine hormone T3, insulin-like growth factor-1 and dexamethasone). HIF-1α inhibition significantly increased FAO and basal and maximal respiration of hiPSC-CMs. Combining HIF-1α inhibition with PPARα activation and the postnatal factors further increased FAO and improved mitochondrial maturation in hiPSC-CMs. Compared with mock-treated cultures, the cultures treated with the five factors had increased mitochondrial content and contained more cells with mitochondrial distribution throughout the cells, which are features of more mature cardiomyocytes. Consistent with these observations, a number of transcriptional regulators of mitochondrial metabolic processes were upregulated in hiPSC-CMs treated with the five factors. Furthermore, these cells had significantly increased Ca2+ transient kinetics and contraction and relaxation velocities, which are functional features for more mature cardiomyocytes. Therefore, targeting HIF-1α in combination with other metabolic regulators significantly improves the metabolic maturation of hiPSC-CMs.


Asunto(s)
Benzamidas/farmacología , Sinergismo Farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Células Madre Pluripotentes Inducidas/fisiología , Mitocondrias/metabolismo , Miocitos Cardíacos/fisiología , PPAR alfa/agonistas , Antiinflamatorios/farmacología , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Dexametasona/farmacología , Metabolismo Energético , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Metabolismo de los Lípidos , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Oxidación-Reducción , Transcriptoma , Triyodotironina/farmacología
4.
Blood ; 125(10): 1562-5, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25593337

RESUMEN

The difficulty in maintaining the reconstituting capabilities of hematopoietic stem cells (HSCs) in culture outside of the bone marrow microenvironment has severely limited their utilization for clinical therapy. This hurdle is largely due to the differentiation of long-term stem cells. Emerging evidence suggests that energy metabolism plays an important role in coordinating HSC self-renewal and differentiation. Here, we show that treatment with alexidine dihydrochloride, an antibiotic and a selective inhibitor of the mitochondrial phosphatase Ptpmt1, which is crucial for the differentiation of HSCs, reprogrammed cellular metabolism from mitochondrial aerobic metabolism to glycolysis, resulting in a remarkable preservation of long-term HSCs ex vivo in part through hyperactivation of adenosine 5'-monophosphate-activated protein kinase (AMPK). In addition, inhibition of mitochondrial metabolism and activation of AMPK by metformin, a diabetes drug, also decreased differentiation and helped maintain stem cells in culture. Thus, manipulating metabolic pathways represents an effective new strategy for ex vivo maintenance of HSCs.


Asunto(s)
Biguanidas/farmacología , Reprogramación Celular/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Fosfohidrolasa PTEN/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Aerobiosis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucólisis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Hipoglucemiantes/farmacología , Metformina/farmacología , Ratones , Consumo de Oxígeno/efectos de los fármacos
5.
J Biol Chem ; 289(1): 251-63, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24265312

RESUMEN

CREB-binding protein (CBP)/p300 interacting transactivator with glutamic acid (Glu) and aspartic acid (Asp)-tail 2 (Cited2) was recently shown to be essential for gluconeogenesis in the adult mouse. The metabolic function of Cited2 in mouse embryonic stem cells (mESCs) remains elusive. In the current study, the metabolism of glucose was investigated in mESCs, which contained a deletion in the gene for Cited2 (Cited2(Δ/-)). Compared with its parental wild type counterpart, Cited2(Δ/-) ESCs have enhanced glycolysis, alternations in mitochondria morphology, reduced glucose oxidation, and decreased ATP content. Cited2 is recruited to the hexokinase 1 (HK1) gene promoter to regulate transcription of HK1, which coordinates glucose metabolism in wild type ESCs. Reduced glucose oxidation and enhanced glycolytic activity in Cited2(Δ/-) ESCs correlates with defective differentiation during hypoxia, which is reflected in an increased expression of pluripotency marker (Oct4) and epiblast marker (Fgf5) and decreased expression of lineage specification markers (T, Gata-6, and Cdx2). Knockdown of hypoxia inducible factor-1α in Cited2(Δ/-) ESCs re-initiates the expression of differentiation markers T and Gata-6. Taken together, a deletion of Cited2 in mESCs results in abnormal mitochondrial morphology and impaired glucose metabolism, which correlates with a defective cell fate decision.


Asunto(s)
Células Madre Embrionarias/metabolismo , Glucólisis/fisiología , Mitocondrias/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Transcripción Genética/fisiología , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/genética , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Hipoxia de la Célula/fisiología , Células Madre Embrionarias/citología , Glucosa/genética , Glucosa/metabolismo , Hexoquinasa/biosíntesis , Hexoquinasa/genética , Ratones , Ratones Noqueados , Mitocondrias/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oxidación-Reducción , Proteínas Represoras/genética , Transactivadores/genética
6.
Res Sq ; 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39149498

RESUMEN

Juvenile myelomonocytic leukemia (JMML), a clonal hematologic malignancy, originates from mutated hematopoietic stem cells (HSCs). The mechanism sustaining the persistence of mutant stem cells, leading to leukemia development, remains elusive. In this study, we conducted comprehensive examination of gene expression profiles, transcriptional factor regulons, and cell compositions/interactions throughout various stages of tumor cell development in Ptpn11 mutation-associated JMML. Our analyses revealed that leukemia-initiating Ptpn11 E76K/+ mutant stem cells exhibited de novo activation of the myeloid transcriptional program and aberrant developmental trajectories. These mutant stem cells displayed significantly elevated expression of innate immunity-associated anti-microbial peptides and pro-inflammatory proteins, particularly S100a9 and S100a8. Biological experiments confirmed that S100a9/S100a8 conferred a selective advantage to the leukemia-initiating cells through autocrine effects and facilitated immune evasion by recruiting and promoting immune suppressive myeloid-derived suppressor cells (MDSCs) in the microenvironment. Importantly, pharmacological inhibition of S100a9/S100a8 signaling effectively impeded leukemia development from Ptpn11 E76K/+ mutant stem cells. These findings collectively suggest that JMML tumor-initiating cells exploit evolutionarily conserved innate immune and inflammatory mechanisms to establish clonal dominance.

7.
Stem Cell Res Ther ; 14(1): 322, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37941041

RESUMEN

BACKGROUND: Cardiac pathological outcome of metabolic remodeling is difficult to model using cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) due to low metabolic maturation. METHODS: hiPSC-CM spheres were treated with AMP-activated protein kinase (AMPK) activators and examined for hiPSC-CM maturation features, molecular changes and the response to pathological stimuli. RESULTS: Treatment of hiPSC-CMs with AMPK activators increased ATP content, mitochondrial membrane potential and content, mitochondrial DNA, mitochondrial function and fatty acid uptake, indicating increased metabolic maturation. Conversely, the knockdown of AMPK inhibited mitochondrial maturation of hiPSC-CMs. In addition, AMPK activator-treated hiPSC-CMs had improved structural development and functional features-including enhanced Ca2+ transient kinetics and increased contraction. Transcriptomic, proteomic and metabolomic profiling identified differential levels of expression of genes, proteins and metabolites associated with a molecular signature of mature cardiomyocytes in AMPK activator-treated hiPSC-CMs. In response to pathological stimuli, AMPK activator-treated hiPSC-CMs had increased glycolysis, and other pathological outcomes compared to untreated cells. CONCLUSION: AMPK activator-treated cardiac spheres could serve as a valuable model to gain novel insights into cardiac diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Células Madre Pluripotentes Inducidas , Humanos , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Células Cultivadas , Proteómica , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Diferenciación Celular/fisiología
8.
Blood ; 116(18): 3611-21, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-20651068

RESUMEN

Germline and somatic gain-of-function mutations in tyrosine phosphatase PTPN11 (SHP-2) are associated with juvenile myelomonocytic leukemia (JMML), a myeloproliferative disease (MPD) of early childhood. The mechanism by which PTPN11 mutations induce this disease is not fully understood. Signaling partners that mediate the pathogenic effects of PTPN11 mutations have not been explored. Here we report that germ line mutation Ptpn11(D61G) in mice aberrantly accelerates hematopoietic stem cell (HSC) cycling, increases the stem cell pool, and elevates short-term and long-term repopulating capabilities, leading to the development of MPD. MPD is reproduced in primary and secondary recipient mice transplanted with Ptpn11(D61G/+) whole bone marrow cells or purified Lineage(-)Sca-1(+)c-Kit(+) cells, but not lineage committed progenitors. The deleterious effects of Ptpn11(D61G) mutation on HSCs are attributable to enhancing cytokine/growth factor signaling. The aberrant HSC activities caused by Ptpn11(D61G) mutation are largely corrected by deletion of Gab2, a prominent interacting protein and target of Shp-2 in cell signaling. As a result, MPD phenotypes are markedly ameliorated in Ptpn11(D61G/+)/Gab2(-/-) double mutant mice. Collectively, our data suggest that oncogenic Ptpn11 induces MPD by aberrant activation of HSCs. This study also identifies Gab2 as an important mediator for the pathogenic effects of Ptpn11 mutations.


Asunto(s)
Células Madre Hematopoyéticas/patología , Mutación , Trastornos Mieloproliferativos/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis , Células de la Médula Ósea/citología , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Regulación Neoplásica de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/metabolismo , Interleucina-3/inmunología , Ratones , Ratones Endogámicos C57BL , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Fosfoproteínas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo
9.
Cell Stem Cell ; 24(4): 608-620.e6, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30880025

RESUMEN

Hematopoietic stem cell (HSC) quiescence is a tightly regulated process crucial for hematopoietic regeneration, which requires a healthy and supportive microenvironmental niche within the bone marrow (BM). Here, we show that deletion of Ptpn21, a protein tyrosine phosphatase highly expressed in HSCs, induces stem cell egress from the niche due to impaired retention within the BM. Ptpn21-/- HSCs exhibit enhanced mobility, decreased quiescence, increased apoptosis, and defective reconstitution capacity. Ptpn21 deletion also decreased HSC stiffness and increased physical deformability, in part by dephosphorylating Spetin1 (Tyr246), a poorly described component of the cytoskeleton. Elevated phosphorylation of Spetin1 in Ptpn21-/- cells impaired cytoskeletal remodeling, contributed to cortical instability, and decreased cell rigidity. Collectively, these findings show that Ptpn21 maintains cellular mechanics, which is correlated with its important functions in HSC niche retention and preservation of hematopoietic regeneration capacity.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Septinas/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Nicho de Células Madre
10.
Nat Metab ; 1(3): 390-403, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-31535081

RESUMEN

Amino acid (AA) metabolism is involved in diverse cellular functions, including cell survival and growth, however it remains unclear how it regulates normal hematopoiesis versus leukemogenesis. Here, we report that knockout of Slc1a5 (ASCT2), a transporter of neutral AAs, especially glutamine, results in mild to moderate defects in bone marrow and mature blood cell development under steady state conditions. In contrast, constitutive or induced deletion of Slc1a5 decreases leukemia initiation and maintenance driven by the oncogene MLL-AF9 or Pten deficiency. Survival of leukemic mice is prolonged following Slc1a5 deletion, and pharmacological inhibition of ASCT2 also decreases leukemia development and progression in xenograft models of human acute myeloid leukemia. Mechanistically, loss of ASCT2 generates a global effect on cellular metabolism, disrupts leucine influx and mTOR signaling, and induces apoptosis in leukemic cells. Given the substantial difference in reliance on ASCT2-mediated AA metabolism between normal and malignant blood cells, this in vivo study suggests ASCT2 as a promising therapeutic target for the treatment of leukemia.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/fisiología , Aminoácidos/metabolismo , Leucemia Mieloide Aguda/metabolismo , Antígenos de Histocompatibilidad Menor/fisiología , Sistema de Transporte de Aminoácidos ASC/genética , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Hematopoyesis/genética , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Antígenos de Histocompatibilidad Menor/genética
11.
Sci Signal ; 11(522)2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29559584

RESUMEN

Catalytically activating mutations in Ptpn11, which encodes the protein tyrosine phosphatase SHP2, cause 50% of Noonan syndrome (NS) cases, whereas inactivating mutations in Ptpn11 are responsible for nearly all cases of the similar, but distinct, developmental disorder Noonan syndrome with multiple lentigines (NSML; formerly called LEOPARD syndrome). However, both types of disease mutations are gain-of-function mutations because they cause SHP2 to constitutively adopt an open conformation. We found that the catalytic activity of SHP2 was required for the pathogenic effects of gain-of-function, disease-associated mutations on the development of hydrocephalus in the mouse. Targeted pan-neuronal knockin of a Ptpn11 allele encoding the active SHP2 E76K mutant resulted in hydrocephalus due to aberrant development of ependymal cells and their cilia. These pathogenic effects of the E76K mutation were suppressed by the additional mutation C459S, which abolished the catalytic activity of SHP2. Moreover, ependymal cells in NSML mice bearing the inactive SHP2 mutant Y279C were also unaffected. Mechanistically, the SHP2 E76K mutant induced developmental defects in ependymal cells by enhancing dephosphorylation and inhibition of the transcription activator STAT3. Whereas STAT3 activity was reduced in Ptpn11E76K/+ cells, the activities of the kinases ERK and AKT were enhanced, and neural cell-specific Stat3 knockout mice also manifested developmental defects in ependymal cells and cilia. These genetic and biochemical data demonstrate a catalytic-dependent role of SHP2 gain-of-function disease mutants in the pathogenesis of hydrocephalus.


Asunto(s)
Mutación con Ganancia de Función , Hidrocefalia/genética , Síndrome LEOPARD/genética , Síndrome de Noonan/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Animales , Biocatálisis , Epéndimo/citología , Epéndimo/metabolismo , Predisposición Genética a la Enfermedad/genética , Humanos , Hidrocefalia/metabolismo , Síndrome LEOPARD/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Síndrome de Noonan/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo
12.
Oncotarget ; 9(31): 21831-21843, 2018 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-29774106

RESUMEN

Juvenile Myelomonocytic Leukemia (JMML) is a pediatric myeloproliferative neoplasm (MPN) that has a poor prognosis. Somatic mutations in Ptpn11 are the most frequent cause of JMML and they commonly occur in utero. Animal models of mutant Ptpn11 have probed the signaling pathways that contribute to JMML. However, existing models may inappropriately exacerbate MPN features by relying on non-hematopoietic-restricted Cre-loxP strains or transplantations into irradiated recipients. In this study we generate hematopoietic-restricted models of Ptpn11E76K-mediated disease using Csf1r-MCM and Flt3Cre. We show that these animals have indolent MPN progression despite robust GM-CSF hypersensitivity and Ras-Erk hyperactivation. Rather, the dominant pathology is pronounced thrombocytopenia with expanded extramedullary hematopoiesis. Furthermore, we demonstrate that the timing of tamoxifen administration in Csf1r-MCM mice can specifically induce recombinase activity in either fetal or adult hematopoietic progenitors. We take advantage of this technique to show more rapid monocytosis following Ptpn11E76K expression in fetal progenitors compared with adult progenitors. Finally, we demonstrate that Ptpn11E76K results in the progressive reduction of T cells, most notably of CD4+ and naïve T cells. This corresponds to an increased frequency of T cell progenitors in the thymus and may help explain the occasional emergence of T-cell leukemias in JMML patients. Overall, our study is the first to describe the consequences of hematopoietic-restricted Ptpn11E76K expression in the absence of irradiation. Our techniques can be readily adapted by other researchers studying somatically-acquired blood disorders.

13.
Sci Adv ; 4(10): eaat2681, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30338292

RESUMEN

While deregulation of mitochondrial metabolism and cytosolic glycolysis has been well recognized in tumor cells, the role of coordinated mitochondrial oxidation and cytosolic fermentation of pyruvate, a key metabolite derived from glucose, in physiological processes is not well understood. Here, we report that knockout of PTPMT1, a mitochondrial phosphoinositide phosphatase, completely blocked postnatal cerebellar development. Proliferation of granule cell progenitors, the most actively replicating cells in the developing cerebellum, was only moderately decreased, and proliferation of Purkinje cell progenitors did not seem to be affected in knockout mice. In contrast, generation of functional Bergmann glia from multipotent precursor cells (radial glia), which is essential for cerebellar corticogenesis, was totally disrupted. Moreover, despite a low turnover rate, neural stem cells were impaired in self-renewal in knockout mice. Mechanistically, loss of PTPMT1 decreased mitochondrial aerobic metabolism by limiting utilization of pyruvate, which resulted in bioenergetic stress in neural precursor/stem cells but not in progenitor or mature cells, leading to cell cycle arrest through activation of the AMPK-p19/p21 pathway. This study suggests that mitochondrial oxidation of the carbohydrate fuel is required for postnatal cerebellar development, and identifies a bioenergetic stress-induced cell cycle checkpoint in neural precursor/stem cells.


Asunto(s)
Metabolismo de los Hidratos de Carbono/fisiología , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Mitocondrias/metabolismo , Células-Madre Neurales/fisiología , Animales , Animales Recién Nacidos , Puntos de Control del Ciclo Celular/genética , Cerebelo/citología , Femenino , Glucólisis , Masculino , Ratones Noqueados , Células-Madre Neurales/citología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Ácido Pirúvico/metabolismo
14.
J Clin Invest ; 128(3): 944-959, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29376892

RESUMEN

Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12-/-) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor-mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of αMß2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12-/- mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12-/- hosts was sufficient to correct the neutrophil migration defect in F12-/- mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated.


Asunto(s)
Factor XII/metabolismo , Neutrófilos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Cicatrización de Heridas , Animales , Calcio/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Trampas Extracelulares , Femenino , Humanos , Inflamación , Leucocitos/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
15.
Cell Signal ; 18(11): 2049-55, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16765027

RESUMEN

Shp-2, a ubiquitously expressed protein tyrosine phosphatase containing two Src homology 2 domains, plays an important role in integrating signaling from the cell surface receptors to intracellular signaling mechanisms. Previous studies have demonstrated that the Shp-2 is involved in hepatocyte growth factor (HGF)-induced cell scattering. Here we report that Shp-2 is required for the HGF-induced activation of sphingosine kinase-1 (SPK1), a highly conserved lipid kinase that plays an important role in cell migration. Loss-of-function mutation of Shp-2 did not affect the expression of SPK1, but resulted in its inactivation and the blockage of HGF-induced migration in embryonic fibroblasts. Reintroduction of functional wild type (WT) Shp-2 into the mutant cells partially restored SPK1 activation, and overexpression of SPK1 in these mutant cells enhanced HGF-induced cell migration. Inhibition of expression or activity of SPK1 in WT cells markedly decreased intracellular S1P levels and HGF-induced cell migration. Furthermore, we found that Shp-2 co-immunoprecipitated with SPK1 and c-Met in embryonic fibroblasts. These studies suggest that Shp-2 is an SPK1-interacting protein and that it plays an indispensable role in HGF-induced SPK1 activation.


Asunto(s)
Movimiento Celular/fisiología , Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/farmacología , Línea Celular , Quinasa de Punto de Control 2 , Embrión de Mamíferos/citología , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Fosfotransferasas (Aceptor de Grupo Alcohol)/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/farmacología
16.
Leuk Res ; 50: 132-140, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27760406

RESUMEN

Current therapy for acute myeloid leukemia (AML) primarily includes high-dose cytotoxic chemotherapy with or without allogeneic stem cell transplantation. Targeting unique cellular metabolism of cancer cells is a potentially less toxic approach. Monotherapy with mitochondrial inhibitors like metformin have met with limited success since escape mechanisms such as increased glycolytic ATP production, especially in hyperglycemia, can overcome the metabolic blockade. As an alternative strategy for metformin therapy, we hypothesized that the combination of 6-benzylthioinosine (6-BT), a broad-spectrum metabolic inhibitor, and metformin could block this drug resistance mechanism. Metformin treatment alone resulted in significant suppression of ROS and mitochondrial respiration with increased glycolysis accompanied by modest cytotoxicity (10-25%). In contrast, 6-BT monotherapy resulted in inhibition of glucose uptake, decreased glycolysis, and decreased ATP with minimal changes in ROS and mitochondrial respiration. The combination of 6-BT with metformin resulted in significant cytotoxicity (60-70%) in monocytic AML cell lines and was associated with inhibition of FLT3-ITD activated STAT5 and reduced c-Myc and GLUT-1 expression. Therefore, although the anti-tumor and metabolic effects of metformin have been limited by the metabolic reprogramming within cells, the novel combination of 6-BT and metformin targets this bypass mechanism resulting in reduced glycolysis, STAT5 inhibition, and increased cell death.


Asunto(s)
Muerte Celular/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Metformina/uso terapéutico , Tioinosina/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Sinergismo Farmacológico , Sangre Fetal/citología , Glucólisis/efectos de los fármacos , Humanos , Secuencias Invertidas Repetidas , Leucemia Mieloide Aguda/genética , Factor de Transcripción STAT5/antagonistas & inhibidores , Tioinosina/uso terapéutico , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/fisiología
17.
Oncogene ; 23(20): 3659-69, 2004 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-15116097

RESUMEN

SHP-2 tyrosine phosphatase is highly expressed in hematopoietic cells; however, the function of SHP-2 in hematopoietic cell processes is not fully understood. Recent identification of SHP-2 mutations in childhood leukemia further emphasizes the importance of SHP-2 regulation in hematopoietic cells. We previously reported that SHP-2 played a positive role in IL-3-induced activation of Jak2 kinase in a catalytic-dependent manner. Interestingly, enforced expression of wild-type (WT) SHP-2 in Ba/F3 cells enhanced growth factor deprivation-induced apoptosis. Biochemical analyses revealed that although IL-3 activation of Jak2 kinase was increased, tyrosyl phosphorylation of its downstream substrate STAT5 was disproportionately decreased by the overexpression of SHP-2. Following IL-3 deprivation, the tyrosyl phosphorylation of STAT5 that is required for its antiapoptotic activity was rapidly diminished in SHP-2 overexpressing cells. As a result, reduction of the putative downstream targets of STAT5-Bcl-X(L) and pim-1 was accelerated by overexpression of SHP-2. Further investigation showed that SHP-2 associated with STAT5, and that it was indeed able to dephosphorylate STAT5. Finally, overexpression of SHP-2 in primary bone marrow hematopoietic progenitor cells compromised their differentiative and proliferative potential, and enhanced growth factor withdrawal-induced cell death. And, the effect of SHP-2 overexpression on growth factor-dependent survival was diminished in STAT5-deficient hematopoietic cells. Taken together, these results suggest that SHP-2 tyrosine phosphatase negatively regulates hematopoietic cell survival by dephosphorylation of STAT5.


Asunto(s)
Sustancias de Crecimiento/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas de la Leche , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas , Animales , Células de la Médula Ósea/metabolismo , Supervivencia Celular/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/fisiología , Péptidos y Proteínas de Señalización Intracelular , Janus Quinasa 2 , Ratones , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Quinasas/metabolismo , Factor de Transcripción STAT5 , Transactivadores/genética , Transactivadores/metabolismo
18.
Oncogene ; 22(38): 5995-6004, 2003 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-12955078

RESUMEN

SHP-2 tyrosine phosphatase is highly expressed in hematopoietic cells, however, the function of SHP-2 in hematopoietic cell signaling is not well understood. Here we focus on the role of SHP-2 phosphatase in the signal transduction of interleukin (IL)-3, a cytokine involved in hematopoietic cell survival, proliferation, and differentiation. We established immortalized SHP-2(-/-) hematopoietic cell pools and showed that IL-3-induced proliferative response was diminished in SHP-2(-/-) cells. Moreover, inhibition of the catalytic activity of SHP-2 in wild-type (WT) bone marrow hematopoietic progenitor cells and Ba/F3 cells by overexpression of catalytically inactive SHP-2 mutant suppressed their differentiative and proliferative responses to IL-3, demonstrating an important positive role for SHP-2 in IL-3 signal transduction. Further biochemical analyses revealed that IL-3-induced Jak/Stat, Erk, and PI3 kinase pathways in SHP-2(-/-) cells were impaired and reintroduction of WT SHP-2 into mutant cells partially restored IL-3 signaling. Interestingly, in catalytically inactive SHP-2-overexpressing Ba/F3 cells, although IL-3-induced activation of Jak2 and Erk kinases was reduced and shortened, PI3 kinase activation remained unaltered. Taken together, these results suggest that SHP-2 tyrosine phosphatase plays multiple roles in IL-3 signal transduction, functioning in both catalytic-dependent and -independent manners in the Jak/Stat, Erk, and PI3 kinase pathways.


Asunto(s)
Interleucina-3/metabolismo , Proteínas de la Leche , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Catálisis , División Celular/efectos de los fármacos , Línea Celular Transformada , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Interleucina-3/farmacología , Péptidos y Proteínas de Señalización Intracelular , Janus Quinasa 2 , Ratones , Ratones Mutantes , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Proteínas Oncogénicas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Quinasas/metabolismo , Receptores de Interleucina-3/metabolismo , Factor de Transcripción STAT5 , Transactivadores/metabolismo
19.
Acta Trop ; 96(2-3): 282-7, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16198300

RESUMEN

A surveillance system has been put in place in the Zhejiang province since achieving there the criteria for transmission interruption of schistosomiasis japonica. Suspected patients and special high-risk groups (e.g. the so called 'floating population' and children under 14 years of age) are screened for Schistosoma japonicum using serological tests. Those with positive serological result are subjected to faecal examination and if S. japonicum eggs are found they are treated with praziquantel and followed by regular re-examination until complete cure, i.e. absence of S. japonicum eggs in faecal samples. Patients with advanced schistosomiasis japonica are continuously followed-up. Implementation of the surveillance system from 1995 to 2002 detected two S. japonicum-infected persons; one came from Anhui province and the other from Jiangsu province, but no new infection occurred among local residents. The number of patients with advanced schistosomiasis japonica decreased from 1524 in 1995 to 906 by the end of 2002; a reduction of 40.6%. However, post-transmission schistosomiasis still continues to weigh on the medial resources. We conclude that the surveillance system in Zhejiang province is effective and a useful means for monitoring the endemic situation of schistosomiasis. Hence, it is recommended to be pursued in the years to come.


Asunto(s)
Esquistosomiasis Japónica/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , China , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sistema de Registros , Esquistosomiasis Japónica/parasitología , Esquistosomiasis Japónica/transmisión , Esplenectomía , Esplenomegalia/parasitología
20.
J Leukoc Biol ; 76(2): 484-90, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15123777

RESUMEN

Signal transducer and activator of transcription-6 (STAT6) plays important roles in cytokine signaling via interleukin-4 and -13 receptors (IL-4R and IL-13R). Mice in which STAT6 has been disrupted by homologous recombination show defects in T helper cell type 2 (Th2) lymphocyte production, resulting in an accumulation of Th1 cells. In addition to defects in differentiation and proliferation of T lymphocytes, STAT6-deficient mice show increased cell-cycle activation and frequency of myeloid progenitors. Although this has been shown to be mediated through Oncostatin M production by T cells, IL-4Ralpha and STAT6 have also recently been found to be enriched for expression in primitive hematopoietic stem cells (HSCs) in gene expression-profiling studies. Therefore, we have investigated whether defects in hematopoietic function in mice lacking STAT6 expression extended into the primitive hematopoietic compartments of the bone marrow. Here, we report that STAT6 deficiency increased bone marrow-committed myeloid progenitors but did not alter the number of cells enriched for HSC/multipotent progenitors, primitive cobblestone area-forming cells assayed in vitro, or bone marrow short-term or long-term repopulating cells assayed in vivo. Therefore, the requirement for STAT6 activation during hematopoiesis is limited, and primitive hematopoietic cell types are insulated against possible effects of cytokine stimulation by Th1 cells.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Células Progenitoras Mieloides/metabolismo , Transactivadores/genética , Animales , Recuento de Células , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción STAT6 , Transactivadores/biosíntesis , Transactivadores/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA