Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Int J Mol Sci ; 24(10)2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37240103

RESUMEN

The three subsets of human monocytes, classical, intermediate, and nonclassical, show phenotypic heterogeneity, particularly in their expression of CD14 and CD16. This has enabled researchers to delve into the functions of each subset in the steady state as well as in disease. Studies have revealed that monocyte heterogeneity is multi-dimensional. In addition, that their phenotype and function differ between subsets is well established. However, it is becoming evident that heterogeneity also exists within each subset, between health and disease (current or past) states, and even between individuals. This realisation casts long shadows, impacting how we identify and classify the subsets, the functions we assign to them, and how they are examined for alterations in disease. Perhaps the most fascinating is evidence that, even in relative health, interindividual differences in monocyte subsets exist. It is proposed that the individual's microenvironment could cause long-lasting or irreversible changes to monocyte precursors that echo to monocytes and through to their derived macrophages. Here, we will discuss the types of heterogeneity recognised in monocytes, the implications of these for monocyte research, and most importantly, the relevance of this heterogeneity for health and disease.


Asunto(s)
Macrófagos , Monocitos , Humanos , Monocitos/metabolismo , Macrófagos/metabolismo , Fenotipo , Hematopoyesis , Receptores de IgG/metabolismo , Receptores de Lipopolisacáridos/metabolismo
2.
Blood ; 119(3): 707-16, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22049516

RESUMEN

CXCL12 and VCAM1 retain hematopoietic stem cells (HSCs) in the BM, but the factors mediating HSC egress from the BM to the blood are not known. The sphingosine-1-phosphate receptor 1 (S1P(1)) is expressed on HSCs, and S1P facilitates the egress of committed hematopoietic progenitors from the BM into the blood. In the present study, we show that both the S1P gradient between the BM and the blood and the expression of S1P(1) are essential for optimal HSC mobilization by CXCR4 antagonists, including AMD3100, and for the trafficking of HSCs during steady-state hematopoiesis. We also demonstrate that the S1P(1) agonist SEW2871 increases AMD3100-induced HSC and progenitor cell mobilization. These results suggest that the combination of a CXCR4 antagonist and a S1P(1) agonist may prove to be sufficient for mobilizing HSCs in normal donors for transplantation purposes, potentially providing a single mobilization procedure and eliminating the need to expose normal donors to G-CSF with its associated side effects.


Asunto(s)
Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/farmacología , Lisofosfolípidos/farmacología , Receptores CXCR4/antagonistas & inhibidores , Esfingosina/análogos & derivados , Adulto , Anciano , Animales , Fármacos Anti-VIH/farmacología , Bencilaminas , Western Blotting , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Ciclamas , Citocinas/metabolismo , Combinación de Medicamentos , Sinergismo Farmacológico , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Persona de Mediana Edad , Oligopéptidos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/metabolismo , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato
3.
Blood ; 113(14): 3297-306, 2009 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-19196656

RESUMEN

Despite advances in the treatment of acute lymphoblastic leukemia (ALL), the majority of children who relapse still die of ALL. Therefore, the development of more potent but less toxic drugs for the treatment of ALL is imperative. We investigated the effects of the mammalian target of rapamycin inhibitor, RAD001 (Everolimus), in a nonobese diabetic/severe combined immunodeficiency model of human childhood B-cell progenitor ALL. RAD001 treatment of established disease increased the median survival of mice from 21.3 days to 42.3 days (P < .02). RAD001 together with vincristine significantly increased survival compared with either treatment alone (P < .02). RAD001 induced a cell-cycle arrest in the G(0/1) phase with associated dephosphorylation of the retinoblastoma protein, and reduced levels of cyclin-dependent kinases 4 and 6. Ultrastructure analysis demonstrated the presence of autophagy and limited apoptosis in cells of RAD001-treated animals. In contrast, cleaved poly(ADP-ribose) polymerase suggested apoptosis in cells from animals treated with vincristine or the combination of RAD001 and vincristine, but not in those receiving RAD001 alone. In conclusion, we have demonstrated activity of RAD001 in an in vivo leukemia model supporting further clinical development of target of rapamycin inhibitors for the treatment of patients with ALL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Sirolimus/análogos & derivados , Vincristina/administración & dosificación , Adolescente , Animales , Niño , Preescolar , Sinergismo Farmacológico , Everolimus , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Placebos , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Sirolimus/administración & dosificación , Sirolimus/farmacología , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Br J Haematol ; 145(4): 491-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19344405

RESUMEN

The mechanisms regulating the migration of leukaemic cells between the blood and bone marrow compartments remain obscure, but are of fundamental importance for the dissemination of the disease. This study investigated the in vivo homing of human B cell progenitor acute lymphoblastic leukaemia (ALL) cells to the femoral bone marrow of non-obese diabetic severe combined immunodeficient (NOD/SCID) mice. It was demonstrated that patient ALL cells use the chemokine axis, chemokine (CXC motif) receptor 4 (CXCR4)/ chemokine (CXC motif) ligand 12 (CXCL12), to home to the femoral marrow. CXCL12-mediated signalling through p38 mitogen-activated protein kinase (MAPK) was required for optimal homing. In contrast, the homing of normal peripheral blood CD34(+) cells and the cytokine-dependent CD34(+) cell line Mo7e was independent of p38MAPK, consistent with the dependence of these cells, as well as normal CD34(+) CD19(+) B cell progenitors, on PI-3K/AKT signalling. Altogether, our data provide clarification of the direct role of CXCL12 in the bone marrow homing of ALL cells and demonstrate unique signalling molecule usage that may have therapeutic implications for this disease.


Asunto(s)
Linfocitos B/fisiología , Quimiotaxis de Leucocito/fisiología , Células Madre Neoplásicas/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Receptores CXCR4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Bencilaminas , Médula Ósea/inmunología , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Cromonas/farmacología , Ciclamas , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Compuestos Heterocíclicos/farmacología , Humanos , Imidazoles/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Morfolinas/farmacología , Oligopéptidos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Unión Proteica , Receptores CXCR4/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
5.
Haematologica ; 92(4): 450-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17488655

RESUMEN

BACKGROUND AND OBJECTIVES: The chemokine stroma-derived factor 1a (SDF-1a or CXCL12) is essential for proliferation of B lineage acute lymphoblastic leukemia (ALL) cells in their physiological microenvironment, bone marrow stroma. CXCL12 synergizes with cytokines that stimulate myeloid cells, but its interaction with cytokines affecting lymphoid cells has not been examined. We investigated whether interleukin (IL)-7 and IL-3 interact with CXCL12 to regulate ALL proliferation. DESIGN AND METHODS: The survival of ALL cells in serum-free cultures, with or without stromal support and cytokines, was assessed by flow cytometry, and proliferation by 3H-thymidine incorporation. Signaling mechanisms were assessed by western blotting of phosphorylated forms of signaling molecules and by the use of specific inhibitors. RESULTS: CXCL12, IL-3, and IL-7 had only marginal effects on ALL cell survival under serum-free conditions. However, these molecules individually induced significant proliferative responses in stromal cultures of 11 cases of ALL. The combination of CXCL12 with IL-7 or IL-3 produced a variety of responses, with clear synergistic or additive interactions observed in four cases. Synergistic proliferation in response to CXCL12 plus IL-7 was associated with enhanced phosphorylation of the mitogen-activated protein kinases, ERK-1/2 and p38, and AKT, and was partially inhibited by pretreatment of cells with inhibitors for p38 MAPK and phosphatidylinositol 3-kinase, implicating these pathways in the proliferation in response to IL-7 plus CXCL12. INTERPRETATION AND CONCLUSIONS: These findings indicate a complex interaction between signaling from the CXCR4 receptor on ALL cells with those initiated by the cytokines IL-7 and IL-3, suggesting that CXCL12 may facilitate ALL proliferation by enhancing cytokine-signaling pathways in responsive cases.


Asunto(s)
Linfocitos B/efectos de los fármacos , Quimiocinas CXC/fisiología , Interleucina-3/farmacología , Interleucina-7/farmacología , Proteínas de Neoplasias/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Receptores CXCR4/fisiología , Transducción de Señal/fisiología , Adolescente , Apoptosis , Linfocitos B/patología , Células Sanguíneas/patología , Células de la Médula Ósea/patología , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Niño , Preescolar , Cromonas/farmacología , Técnicas de Cocultivo , Medio de Cultivo Libre de Suero , Sinergismo Farmacológico , Femenino , Humanos , Imidazoles/farmacología , Lactante , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/fisiología , Piridinas/farmacología , Receptores CXCR4/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/patología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
6.
Cancer Res ; 65(8): 3290-8, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15833862

RESUMEN

The chemokine stromal-derived factor-1alpha (SDF-1alpha) regulates leukemic cell motility and proliferation; however, the importance of these functions in the growth and dissemination of leukemia is unclear. We examined SDF-1alpha-mediated responses of cells from 27 cases of acute lymphoblastic leukemia (ALL). Although cells from the majority of cases showed chemotactic and proliferative responses to SDF-1alpha, a subset of cases did not undergo chemotaxis in response to SDF-1alpha, while still demonstrating dependence on SDF-1alpha for proliferation in stroma-supported cultures. This chemotactic defect was associated with an absence of phosphorylation of p38 mitogen-activated protein kinase (MAPK) induced by SDF-1alpha, and of SDF-1alpha-induced augmentation of beta(1) integrin-mediated adhesion. Signaling through phosphoinositide 3-kinase and MEK was not affected. No correlation was observed between CXCR4 expression and chemotactic function, in vitro migration into bone marrow stromal layers, and engraftment of leukemic cells in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. This study suggests that signaling through p38 MAPK is required for ALL cell chemotaxis but not for proliferation, and that the loss of a chemotactic response to SDF-1alpha does not impede engraftment in NOD/SCID mice.


Asunto(s)
Linfoma de Burkitt/enzimología , Linfoma de Burkitt/patología , Quimiocinas CXC/farmacología , Quimiotaxis de Leucocito/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adolescente , Adulto , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Calcio/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Quimiocina CXCL12 , Quimiotaxis de Leucocito/fisiología , Niño , Preescolar , Femenino , Humanos , Lactante , Integrina alfa4beta1/metabolismo , Integrina alfa5beta1/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad
7.
PLoS One ; 9(7): e103383, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25061812

RESUMEN

Resistance to apoptosis remains a significant problem in drug resistance and treatment failure in malignant disease. NO-aspirin is a novel drug that has efficacy against a number of solid tumours, and can inhibit Wnt signaling, and although we have shown Wnt signaling to be important for acute lymphoblastic leukemia (ALL) cell proliferation and survival inhibition of Wnt signaling does not appear to be involved in the induction of ALL cell death. Treatment of B lineage ALL cell lines and patient ALL cells with NO-aspirin induced rapid apoptotic cell death mediated via the extrinsic death pathway. Apoptosis was dependent on caspase-10 in association with the formation of the death-inducing signaling complex (DISC) incorporating pro-caspase-10 and tumor necrosis factor receptor 1 (TNF-R1). There was no measurable increase in TNF-R1 or TNF-α in response to NO-aspirin, suggesting that the process was ligand-independent. Consistent with this, expression of silencer of death domain (SODD) was reduced following NO-aspirin exposure and lentiviral mediated shRNA knockdown of SODD suppressed expansion of transduced cells confirming the importance of SODD for ALL cell survival. Considering that SODD and caspase-10 are frequently over-expressed in ALL, interfering with these proteins may provide a new strategy for the treatment of this and potentially other cancers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Caspasa 10/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Caspasa 10/genética , Células Cultivadas , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Silenciador del Gen , Humanos , Células Jurkat
8.
PLoS One ; 9(7): e102494, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25014496

RESUMEN

Increasingly, anti-cancer medications are being reported to induce cell death mechanisms other than apoptosis. Activating alternate death mechanisms introduces the potential to kill cells that have defects in their apoptotic machinery, as is commonly observed in cancer cells, including in hematological malignancies. We, and others, have previously reported that the mTOR inhibitor everolimus has pre-clinical efficacy and induces caspase-independent cell death in acute lymphoblastic leukemia cells. Furthermore, everolimus is currently in clinical trial for acute lymphoblastic leukemia. Here we characterize the death mechanism activated by everolimus in acute lymphoblastic leukemia cells. We find that cell death is caspase-independent and lacks the morphology associated with apoptosis. Although mitochondrial depolarization is an early event, permeabilization of the outer mitochondrial membrane only occurs after cell death has occurred. While morphological and biochemical evidence shows that autophagy is clearly present it is not responsible for the observed cell death. There are a number of features consistent with paraptosis including morphology, caspase-independence, and the requirement for new protein synthesis. However in contrast to some reports of paraptosis, the activation of JNK signaling was not required for everolimus-induced cell death. Overall in acute lymphoblastic leukemia cells everolimus induces a cell death that resembles paraptosis.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Caspasas/genética , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Niño , Relación Dosis-Respuesta a Droga , Estrés del Retículo Endoplásmico/efectos de los fármacos , Everolimus , Humanos , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
9.
Exp Hematol ; 40(3): 207-215.e1, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22100881

RESUMEN

Although patients with acute lymphoblastic leukemia (ALL) usually achieve complete remission, disease relapse is common and difficult to treat. Para-NO-aspirin (para-NO-ASA) is a novel drug with demonstrated efficacy against a number of solid tumors and most recently chronic lymphocytic leukemia. In this study, we used ALL cell lines to assess the effects on cell viability by flow cytometry and investigated the mechanism of cell death using chemical inhibitors of key molecules and assessed the effects by flow cytometry, electrophoretic mobility shift assay, Western blotting, and quantitative reverse transcription polymerase chain reaction. Para-NO-ASA induced cell death in the pre-B ALL cell lines in association with increased reactive oxygen species, and suppression of nuclear factor-κB (NF-κB) activity. Chemical inhibitors of NF-κB similarly induced apoptosis in ALL cells, suggesting a role for suppression of NF-κB in para-NO-ASA-induced cell death. Modulation of NF-κB was not via regulation of IκB but potentially through suppression of ROCK1 and loss of reduced glutathione. Our results demonstrate that para-NO-ASA potently induces apoptosis in B-lineage ALL cells via a reactive oxygen species-dependent mechanism that is associated with suppression of NF-κB activity.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Aspirina/análogos & derivados , FN-kappa B/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Donantes de Óxido Nítrico/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Aspirina/farmacología , Caspasas/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Glutatión/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Quinasas Asociadas a rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA