Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 300(1): 105509, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38042493

RESUMEN

Today, the majority of patients with pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL) survive their disease, but many of the survivors suffer from life-limiting late effects of the treatment. ALL develops in the bone marrow, where the cells are exposed to cAMP-generating prostaglandin E2. We have previously identified the cAMP signaling pathway as a putative target for improved efficacy of ALL treatment, based on the ability of cAMP signaling to reduce apoptosis induced by DNA damaging agents. In the present study, we have identified the antioxidant N-acetyl cysteine (NAC) as a powerful modifier of critical events downstream of the cell-permeable cAMP analog 8-(4-chlorophenylthio) adenosine-3', 5'- cyclic monophosphate (8-CPT). Accordingly, we found NAC to turn 8-CPT into a potent killer of ALL cells in vitro both in the presence and absence of DNA damaging treatment. Furthermore, we revealed that NAC in combination with 8-CPT is able to delay the progression of ALL in a xenograft model in NOD-scid IL2Rγnull mice. NAC was shown to rely on the ability of 8-CPT to activate the guanine-nucleotide exchange factor EPAC, and we demonstrated that the ALL cells are killed by apoptosis involving sustained elevated levels of calcium imposed by the combination of the two drugs. Taken together, we propose that 8-CPT in the presence of NAC might be utilized as a novel strategy for treating pediatric ALL patients, and that this powerful combination might be exploited to enhance the therapeutic index of current ALL targeting therapies.


Asunto(s)
Acetilcisteína , AMP Cíclico , Factores de Intercambio de Guanina Nucleótido , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Tionucleótidos , Animales , Niño , Humanos , Ratones , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , AMP Cíclico/uso terapéutico , ADN/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/agonistas , Ratones Endogámicos NOD , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Masculino , Femenino , Preescolar , Tionucleótidos/farmacología , Tionucleótidos/uso terapéutico , Daño del ADN , Quimioterapia Combinada
2.
J Immunol ; 204(8): 2133-2142, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32188759

RESUMEN

It is becoming increasingly evident that reactive oxygen species (ROS) have critical roles as "second messengers" in cell signaling. In B cells, ROS can be generated either as a byproduct of mitochondrial respiration, as a result of the endoplasmic reticulum stress response induced by high production of Igs, or by the activation of NADPH oxidase (NOX) complexes. Having previously shown that costimulation of B cells via TLR 9 and the TLR-related receptor RP105 drives maturation of human peripheral blood B cells into Ig-producing cells, we aimed to study the role of ROS generated during this vital process. To this end, the ROS levels were either reduced by the NOX inhibitor VAS2870 or by the ROS scavenger N-acetyl cysteine (NAC). We revealed that TLR9/RP105-mediated stimulation of human B cells involved a rapid activation of NOX. Moreover, VAS2870 blocked the TLR9/RP105-induced B cell activation and thereby all Ig production. Importantly, we showed that ROS targeted by NAC was selectively required for IgG but not for IgM production. The endoplasmic reticulum stress response in the TLR9/RP105-stimulated cells was higher in IgG+ than in IgG- cells and was reduced by NAC in IgG+ cells only. Of note, we revealed that substantially higher levels of IgG than IgM were produced per cell and that IgG+ cells produced significantly higher ROS levels than IgG- cells. Taken together, our results imply that NAC-targeted ROS may be particularly important for sustaining the high Ig production in IgG+ B cells.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores Toll-Like/inmunología , Acetilcisteína/farmacología , Benzoxazoles/farmacología , Humanos , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Triazoles/farmacología
3.
Mol Cancer ; 14: 14, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25623255

RESUMEN

BACKGROUND: B cell precursor acute lymphoblastic leukaemia (BCP-ALL) is the most common paediatric cancer. BCP-ALL blasts typically retain wild type p53, and are therefore assumed to rely on indirect measures to suppress transformation-induced p53 activity. We have recently demonstrated that the second messenger cyclic adenosine monophosphate (cAMP) through activation of protein kinase A (PKA) has the ability to inhibit DNA damage-induced p53 accumulation and thereby promote survival of the leukaemic blasts. Development of BCP-ALL in the bone marrow (BM) is supported by resident BM-derived mesenchymal stromal cells (MSCs). MSCs are known to produce prostaglandin E(2) (PGE(2)) which upon binding to its receptors is able to elicit a cAMP response in target cells. We hypothesized that PGE(2) produced by stromal cells in the BM microenvironment could stimulate cAMP production and PKA activation in BCP-ALL cells, thereby suppressing p53 accumulation and promoting survival of the malignant cells. METHODS: Primary BCP-ALL cells isolated from BM aspirates at diagnosis were cocultivated with BM-derived MSCs, and effects on DNA damage-induced p53 accumulation and cell death were monitored by SDS-PAGE/immunoblotting and flow cytometry-based methods, respectively. Effects of intervention of signalling along the PGE(2)-cAMP-PKA axis were assessed by inhibition of PGE(2) production or PKA activity. Statistical significance was tested by Wilcoxon signed-rank test or paired samples t test. RESULTS: We demonstrate that BM-derived MSCs produce PGE(2) and protect primary BCP-ALL cells from p53 accumulation and apoptotic cell death. The MSC-mediated protection of DNA damage-mediated cell death is reversible upon inhibition of PGE(2) synthesis or PKA activity. Furthermore our results indicate differences in the sensitivity to variations in p53 levels between common cytogenetic subgroups of BCP-ALL. CONCLUSIONS: Our findings support our hypothesis that BM-derived PGE(2), through activation of cAMP-PKA signalling in BCP-ALL blasts, can inhibit the tumour suppressive activity of wild type p53, thereby promoting leukaemogenesis and protecting against therapy-induced leukaemic cell death. These novel findings identify the PGE(2)-cAMP-PKA signalling pathway as a possible target for pharmacological intervention with potential relevance for treatment of BCP-ALL.


Asunto(s)
Daño del ADN , Dinoprostona/metabolismo , Células Madre Mesenquimatosas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Muerte Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Modelos Biológicos , Transducción de Señal
4.
Blood ; 121(10): 1805-13, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23299313

RESUMEN

B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is the most commonly occurring pediatric cancer. Despite its relatively good prognosis, there is a steady search for strategies to improve treatment effects and prevent the undesired side effects on normal cells. In the present paper, we demonstrate a differential effect of cyclic adenosine monophosphate (cAMP) signaling between normal BCPs and BCP-ALL blasts, pointing to a potential therapeutic window allowing for manipulation of cAMP signaling in the treatment of BCP-ALL. By studying primary cells collected from pediatric BCP-ALL patients and healthy controls, we found that cAMP profoundly decreased basal and DNA damage-induced p53 levels and cell death in malignant cells, whereas normal BCP counterparts displayed slightly augmented cell death when exposed to cAMP-increasing agents. We did not find evidence for a selection process involving generation of increased basal cAMP levels in BCP-ALL cells, but we demonstrate that paracrine signaling involving prostaglandin E2-induced cAMP generation has the potential to suppress p53 activation and cell death induction. The selective inhibitory effect of cAMP signaling on DNA damage-induced cell death in BCP-ALL cells appears to be an acquired trait associated with malignant transformation, potentially allowing the use of inhibitors of this pathway for directed killing of the malignant blasts.


Asunto(s)
Apoptosis , Crisis Blástica/patología , AMP Cíclico/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Células Precursoras de Linfocitos B/citología , Proteína p53 Supresora de Tumor/metabolismo , Adolescente , Adulto , Crisis Blástica/tratamiento farmacológico , Crisis Blástica/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Niño , Preescolar , Colforsina/farmacología , Daño del ADN/fisiología , Daño del ADN/efectos de la radiación , Dinoprostona/farmacología , Femenino , Humanos , Lactante , Masculino , Oxitócicos/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Proteína p53 Supresora de Tumor/genética , Adulto Joven
5.
Cell Immunol ; 279(1): 87-95, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23103284

RESUMEN

The role of vitamin A in the various parts of the immune system remains elusive. Toll-like receptors (TLRs) are involved in innate polyclonal activation of B-cells, and as such they are important for maintaining long-lasting first line defense against pathogens. Here we explore the impact of all-trans retinoic acid (RA) on B cell responses mediated via the TLR homolog RP105 (CD180). We show that RA slightly reduces the proliferation and IgG production in CD27+ memory B cells stimulated by anti-RP105 alone. However, co-stimulation with the TLR9-ligand CpG results in turning RA into a potent stimulator of RP105-induced proliferation and IgG synthesis in memory B cells. The results emphasize the important role of RA in stimulating TLR-mediated polyclonal activation and differentiation of B cells, and reveal the complex interplay between various TLRs that may underlie the ability of RA to fight pathogens.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos B/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inmunoglobulina G/biosíntesis , Receptor Toll-Like 9/metabolismo , Tretinoina/farmacología , Anticuerpos/farmacología , Antígenos CD/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Memoria Inmunológica/inmunología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Oligodesoxirribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 9/agonistas , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
6.
Mol Cancer Res ; 20(3): 400-411, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34880123

RESUMEN

DNA-damaging therapy is the basis for treatment of most cancers, including B-cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL). We have previously shown that cAMP-activating factors present in the bone marrow render ALL cells less sensitive to DNA damage-induced apoptosis, by enhancing autophagy and suppressing p53. To sensitize ALL cells to DNA-damaging therapy, we have searched for novel targets that may counteract the effects induced by cAMP signaling. In the current study, we have identified PARP1 as a potential target. We show that the PARP1 inhibitors olaparib or PJ34 inhibit cAMP-mediated autophagy and thereby potentiate the DNA-damaging treatment. Furthermore, we reveal that cAMP-mediated PARP1 activation is preceded by induction of reactive oxygen species (ROS) and results in depletion of nicotinamide adenine dinucleotide (NAD), both of which are autophagy-promoting events. Accordingly, we demonstrate that scavenging ROS by N-acetylcysteine and repleting NAD independently reduce DNA damage-induced autophagy. In addition, olaparib augmented the effect of DNA-damaging treatment in a human xenograft model of ALL in NOD-scidIL2Rgammanull mice. On the basis of the current findings, we suggest that PARP1 inhibitors may enhance the efficiency of conventional genotoxic therapies and thereby provide a novel treatment strategy for pediatric patients with ALL. IMPLICATIONS: PARP1 inhibitors augment the DNA damage-induced killing of ALL cells by limiting the opposing effects of cAMP-mediated autophagy, which involves ROS-induced PARP1 activation and depletion of cellular NAD levels.


Asunto(s)
NAD , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Autofagia , Supervivencia Celular , Niño , Humanos , Ratones , Ratones Endogámicos NOD , Poli(ADP-Ribosa) Polimerasa-1/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Especies Reactivas de Oxígeno
7.
Blood ; 114(3): 608-18, 2009 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-19451550

RESUMEN

In lymphocytes, the second messenger cyclic adenosine monophosphate (cAMP) plays a well-established antiproliferative role through inhibition of G(1)/S transition and S-phase progression. We have previously demonstrated that, during S-phase arrest, cAMP inhibits the action of S phase-specific cytotoxic compounds, leading to reduction in their apoptotic response. In this report, we provide evidence that cAMP can also inhibit the action of DNA-damaging agents independently of its effect on S phase. Elevation of cAMP in B-cell precursor acute lymphoblastic leukemia cells is shown to profoundly inhibit the apoptotic response to ionizing radiation, anthracyclins, alkylating agents, and platinum compounds. We further demonstrate that this effect depends on the ability of elevated cAMP levels to quench DNA damage-induced p53 accumulation by increasing the p53 turnover, resulting in attenuated Puma and Bax induction, mitochondrial outer membrane depolarization, caspase activation, and poly(ADP-ribose) polymerase cleavage. On the basis of our findings, we suggest that cAMP levels may influence p53 function in malignant cells that retain wild-type p53, potentially affecting p53 both as a tumor suppressor during cancer initiation and maintenance, and as an effector of the apoptotic response to DNA-damaging agents during anticancer treatment.


Asunto(s)
Apoptosis , AMP Cíclico/metabolismo , Daño del ADN , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proteínas Reguladoras de la Apoptosis/biosíntesis , Ciclo Celular , Línea Celular Tumoral , Humanos , Radiación Ionizante
8.
Immunology ; 126(4): 514-22, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18778286

RESUMEN

Interleukin-2 (IL-2) is an essential cytokine for T-lymphocyte homeostasis. We have previously reported that all-trans retinoic acid (atRA) enhances the secretion of IL-2 from human peripheral blood T cells in vitro, followed by increased proliferation and inhibition of spontaneous cell death. In this study we used a transgenic IL-2 gene luciferase reporter model to examine the effects of atRA in vivo. In contrast to the observations in human T cells, we found an overall reduction in luciferase-reported IL-2 gene expression in mice treated with atRA. Whole-body luminescence of anti-CD3-treated and non-treated mice was reduced in mice receiving atRA. Accordingly, after 7 hr, IL-2 gene expression was on average 55% lower in the atRA-treated mice compared with the control mice. Furthermore, mice fed a vitamin A-deficient diet had a significantly higher basal level of luciferase activity compared with control mice, demonstrating that vitamin A modulates IL-2 gene expression in vivo. Importantly, the atRA-mediated inhibition of IL-2 gene expression was accompanied by decreased DNA synthesis in murine T cells, suggesting a physiological relevance of the reduced IL-2 gene expression observed in transgenic reporter mice.


Asunto(s)
Interleucina-2/biosíntesis , Activación de Linfocitos/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Tretinoina/farmacología , Animales , Benzoatos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromanos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-2/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/inmunología , Bazo/inmunología , Linfocitos T/inmunología , Deficiencia de Vitamina A/inmunología
9.
Cell Signal ; 20(6): 1169-78, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18406106

RESUMEN

Cyclic AMP (cAMP) is an important physiological growth inhibitor of lymphoid cells, and the cAMP/protein kinase A (PKA) pathway is disrupted in several immunological disorders and cancers. Epstein Barr virus (EBV) infection of B lymphocytes is responsible for the development of lymphoproliferative disease as well as certain B-lymphoid malignancies. Here we hypothesized that EBV infection might render B lymphocytes resistant to cAMP/PKA-mediated growth inhibition. To test this, we assessed the growth-inhibitory response of cAMP-elevating compounds such as forskolin and isoproterenol, as well as the PKA activator 8-CPT-cAMP in normal B lymphocytes, EBV-infected B cells and in the EBV-negative B lymphoid cell line Reh. We could demonstrate that EBV infection indeed abolished cAMP-mediated growth inhibition of B cells. The defect was pinpointed to defective adenylyl cyclase (AC) activation by forskolin and isoproterenol, resulting in reduced formation of cAMP and lack of PKA activation and CREB phosphorylation. In contrast, 8-CPT-cAMP which directly activates PKA was able to inhibit EBV-infected B cell growth. The physiological implications of these results were underlined by the observation that the ability of forskolin to inhibit camptothecin-induced apoptosis was abolished in EBV-infected B cells. We conclude that EBV infection of B cells abrogates the activation of AC and thereby cAMP formation, and that this dysfunction renders the cells resistant to growth inhibition via the cAMP/PKA pathway.


Asunto(s)
Adenilil Ciclasas/metabolismo , Linfocitos B/virología , Herpesvirus Humano 4/fisiología , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/enzimología , Camptotecina/toxicidad , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colforsina/antagonistas & inhibidores , AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Isoproterenol/farmacología , Transducción de Señal
10.
Blood Adv ; 3(21): 3181-3190, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31698450

RESUMEN

Acute lymphoblastic leukemia (ALL) develops in the bone marrow in the vicinity of stromal cells known to promote tumor development and treatment resistance. We previously showed that the cyclooxygenase (COX) inhibitor indomethacin prevents the ability of stromal cells to diminish p53-mediated killing of cocultured ALL cells in vitro, possibly by blocking the production of prostaglandin E2 (PGE2). Here, we propose that PGE2 released by bone marrow stromal cells might be a target for improved treatment of pediatric ALL. We used a xenograft model of human primary ALL cells in nonobese diabetic-scid IL2rγnull mice to show that indomethacin delivered in the drinking water delayed the progression of ALL in vivo. The progression was monitored by noninvasive in vivo imaging of the engrafted leukemic cells, as well as by analyses of CD19+CD10+ leukemic blasts present in spleen or bone marrow at the termination of the experiments. The indomethacin treatment increased the level of p53 in the leukemic cells, implying that COX inhibition might reduce progression of ALL by attenuating protective paracrine PGE2 signaling from bone marrow stroma to leukemic cells.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacología , Indometacina/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Biomarcadores , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Niño , Preescolar , Dinoprostona/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunofenotipificación , Masculino , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Hum Immunol ; 69(12): 833-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18835311

RESUMEN

Defects in Fas receptor signaling lead to compromised maintenance of lymphocyte homeostasis and peripheral immune tolerance, leading in turn to autoimmune disorders. Therefore, agents that can enhance Fas-mediated apoptosis may be therapeutically useful in management of such disorders. In this study, we focused on the effect of cAMP on Fas-mediated apoptosis in human T cells. We show that elevation of intracellular cAMP levels by forskolin, an activator of adenylyl cyclase, 3-isobutyl-1-methylxanthine, an inhibitor of cyclic nucleotide phosphodiesterases, or prostaglandin E(2) potentiates Fas-induced apoptosis in Jurkat cells. Accordingly, cAMP was found to enhance the cleavage of caspase 8 at death-inducing signaling complex and lead to augmentation of the processing of Fas effector proteins. We also demonstrate that cAMP enahnaces Fas-induced apoptosis in normal human T cells and activation-induced cell death in Jurkat cells. These findings provide a rationale for investigating the feasibility of using cAMP-elevating agents to potentiate apoptosis in T cells with aberrant Fas signaling.


Asunto(s)
Apoptosis/inmunología , Linfocitos T CD4-Positivos/metabolismo , Caspasa 8/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Transducción de Señal/inmunología , Adenilil Ciclasas/inmunología , Adenilil Ciclasas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Caspasa 8/inmunología , Colforsina/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Células Jurkat , Fitohemaglutininas/farmacología , Pirofosfatasas/inmunología , Pirofosfatasas/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Oncotarget ; 9(54): 30434-30449, 2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-30100998

RESUMEN

Autophagy is important in regulating the balance between cell death and survival, with the tumor suppressor p53 as one of the key components in this interplay. We have previously utilized an in vitro model of the most common form of childhood cancer, B cell precursor acute lymphoblastic leukemia (BCP-ALL), to show that activation of the cAMP signaling pathway inhibits p53-mediated apoptosis in response to DNA damage in both cell lines and primary leukemic cells. The present study reveals that cAMP-mediated survival of BCP-ALL cells exposed to DNA damaging agents, involves a critical and p53-independent enhancement of autophagy. Although autophagy generally is regarded as a survival mechanism, DNA damage-induced apoptosis has been linked both to enhanced and reduced levels of autophagy. Here we show that exposure of BCP-ALL cells to irradiation or cytotoxic drugs triggers autophagy and cell death in a p53-dependent manner. Stimulation of the cAMP signaling pathway further augments autophagy and inhibits the DNA damage-induced cell death concomitant with reduced nuclear levels of p53. Knocking-down the levels of p53 reduced the irradiation-induced autophagy and cell death, but had no effect on the cAMP-mediated autophagy. Moreover, prevention of autophagy by bafilomycin A1 or by the ULK-inhibitor MRT68921, diminished the protecting effect of cAMP signaling on DNA damage-induced cell death. Having previously proposed a role of the cAMP signaling pathway in development and treatment of BCP-ALLs, we here suggest that inhibitors of autophagy may improve current DNA damage-based therapy of BCP-ALL - independent of p53.

13.
Cell Cycle ; 16(10): 947-956, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28379780

RESUMEN

Faithful chromosome segregation during mitosis relies on a proofreading mechanism that monitors proper kinetochore-microtubule attachments. The spindle assembly checkpoint (SAC) is based on the concerted action of numerous components that maintain a repressive signal inhibiting transition into anaphase until all chromosomes are attached. Here we show that A-Kinase Anchoring Protein 95 (AKAP95) is necessary for proper SAC function. AKAP95-depleted HeLa cells show micronuclei formed from lagging chromosomes at mitosis. Using a BioID proximity-based proteomic screen, we identify the nuclear pore complex protein TPR as a novel AKAP95 binding partner. We show interaction between AKAP95 and TPR in mitosis, and an AKAP95-dependent enrichment of TPR in the spindle microtubule area in metaphase, then later in the spindle midzone area. AKAP95-depleted cells display faster prometaphase to anaphase transition, escape from nocodazole-induced mitotic arrest and show a partial delocalization from kinetochores of the SAC component MAD1. Our results demonstrate an involvement of AKAP95 in proper SAC function likely through its interaction with TPR.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Proteínas de Complejo Poro Nuclear/genética , Proteómica , Proteínas Proto-Oncogénicas/genética , Segregación Cromosómica/genética , Células HeLa , Humanos , Mitosis/genética , Poro Nuclear/genética , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Unión Proteica , Huso Acromático/genética , Huso Acromático/metabolismo
14.
PLoS One ; 12(10): e0185708, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28973009

RESUMEN

In the present study, we address the important issue of whether B-cells protected from irradiation-induced cell death, may survive with elevated levels of DNA damage. If so, such cells would be at higher risk of gaining mutations and undergoing malignant transformation. We show that stimulation of B-cells with the TLR9 ligands CpG-oligodeoxynucleotides (CpG-ODN) prevents spontaneous and irradiation-induced death of normal peripheral blood B-cells, and of B-cells from patients diagnosed with Common variable immunodeficiency (CVID). The TLR9-mediated survival is enhanced by the vitamin A metabolite retinoic acid (RA). Importantly, neither stimulation of B-cells via TLR9 alone or with RA increases irradiation-induced DNA strand breaks and DNA damage responses such as activation of ATM and DNA-PKcs. We prove that elevated levels of γH2AX imposed by irradiation of stimulated B-cells is not due to induction of DNA double strand breaks, but merely reflects increased levels of total H2AX upon stimulation. Interestingly however, we unexpectedly find that TLR9 stimulation of B-cells induces low amounts of inactive p53, explained by transcriptional induction of TP53. Taken together, we show that enhanced survival of irradiated B-cells is not accompanied by elevated levels of DNA damage. Our results imply that TLR9-mediated activation of B-cells not only promotes cell survival, but may via p53 provide cells with a barrier against harmful consequences of enhanced activation and proliferation. As CVID-derived B-cells are more radiosensitive and prone to undergo apoptosis than normal B-cells, our data support treatment of CVID patients with CpG-ODN and RA.


Asunto(s)
Linfocitos B/fisiología , Inmunodeficiencia Variable Común/genética , Daño del ADN , Rayos Infrarrojos , Receptor Toll-Like 9/fisiología , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/genética , Estudios de Casos y Controles , Humanos
15.
J Nutr Biochem ; 27: 153-63, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26419686

RESUMEN

Chronic inflammation contributes to prostate cancer and the transcription factor Nuclear Factor-kappa B (NF-κB) is constitutively active in most such cancers. We examine the effects of coffee on NF-κB and on the regulation of selected genes in human-derived prostate cancer cells (PC3) and in PC3 xenografts in athymic nude mice. PC3 cells stably transduced with an NF-κB-luciferase reporter were used both in vitro and for xenografts. NF-κB activity was measured by reporter assays, DNA binding and in vivo imaging. Gene expression was measured in PC3 cells, xenografts and tumor microenvironment by low-density arrays. Western blotting of activated caspases was used to quantify apoptosis. Coffee inhibited TNFα-induced NF-κB activity and DNA-binding in PC3 cells. Furthermore, coffee increased apoptosis and modulated expression of a number of inflammation- and cancer-related genes in TNFα-treated PC3 cells. In vivo imaging revealed a 31% lower NF-κB-luciferase activation in the xenografts of the mice receiving 5% coffee compared to control mice. Interestingly, we observed major changes in gene expression in the PC3 cells in xenografts as compared to PC3 cells in vitro. In PC3 xenografts, genes related to inflammation, apoptosis and cytoprotection were down-regulated in mice receiving coffee, and coffee also affected the gene expression in the xenograft microenvironment. Our data demonstrate that coffee inhibits NF-κB activity in PC3 cells in vitro and in xenografts. Furthermore, coffee modulates transcription of genes related to prostate cancer and inflammation. Our results are the first to suggest mechanistic links between coffee consumption and prostate cancer in an experimental mouse model.


Asunto(s)
Café , FN-kappa B/metabolismo , Neoplasias de la Próstata/patología , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Xenoinjertos , Humanos , Masculino , Ratones
16.
Cell Signal ; 15(9): 871-81, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12834812

RESUMEN

The purpose of the present study was to understand the mechanism by which activated protein kinase A (PKA) leads to down-regulation of cyclin D3 in lymphocytes. By using Jurkat cells as a model system, we have been able to demonstrate that cyclin D3 is reduced at the level of translation by inhibition of elongation. One of the important factors involved in translational elongation is the eukaryotic elongation factor 2 (eEF2). eEF2 promotes translation in its unphosphorylated form, and we observed a rapid phosphorylation of the eEF2-protein upon forskolin treatment. When using specific inhibitors of the eEF2-kinase prior to forskolin treatment, we were able to inhibit the increased phosphorylation of eEF2. Furthermore, inhibition of eEF2-kinase prevented the forskolin-mediated down-regulation of cyclin D3. Taken together, it appears that activation of PKA in Jurkat cells reduces the expression of cyclin D3 at the level of translational elongation by increasing the phosphorylation of eEF2 and thereby inhibiting its activity.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , AMP Cíclico/fisiología , Ciclinas/genética , Factor 2 de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas , Linfocitos T/metabolismo , Northern Blotting , Western Blotting , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/efectos de los fármacos , Cromonas/farmacología , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ciclina D3 , Ciclinas/efectos de los fármacos , Ciclinas/metabolismo , Cicloheximida/farmacología , Replicación del ADN/efectos de los fármacos , Regulación hacia Abajo , Quinasa del Factor 2 de Elongación , Citometría de Flujo/métodos , Fase G1/efectos de los fármacos , Fase G1/genética , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Células Jurkat , Morfolinas/farmacología , Compuestos de Organoselenio/farmacología , Extensión de la Cadena Peptídica de Translación/fisiología , Factor 2 de Elongación Peptídica/efectos de los fármacos , Fosforilación/efectos de los fármacos , Polirribosomas/genética , Polirribosomas/metabolismo , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Linfocitos T/efectos de los fármacos , Transfección
17.
Exp Hematol ; 30(9): 990-1000, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12225790

RESUMEN

OBJECTIVE: Several transducing molecules, including JAK2, STAT5, MAP kinases, phosphatidylinositol 3-kinase (PI3K), phospholipase C-gamma1, and PKC are activated by interaction between erythropoietin (EPO) and the EPO receptor. The aim of this was to examine the relative involvement of PI3K in the development of glycophorin A (GPA)(+) erythroid cells from normal hematopoietic progenitor cells. MATERIALS AND METHODS: CD34(+) hematopoietic progenitor cells or subpopulations obtained by FACS sorting were cultured in serum-free medium containing EPO with or without inhibitors for PI3K, p38, MEK, or PKC for various time periods before phenotypic analysis or detection of apoptosis by flow cytometry, cell cycle analysis, high-resolution tracking of cell division, Western blot analysis, or Akt kinase assay were performed. RESULTS: The PI3K inhibitor LY294002 completely counteracted the EPO-induced proliferation of CD34(+) progenitor cells and CD34(+)CD71(+)CD45RA(-) erythroid progenitors. LY294002 also highly suppressed the expanded erythropoiesis induced by the combined action of EPO and stem cell factor. The profound inhibitory effect of LY294002 on proliferation was caused by its induction of cell cycle arrest in the G(0)/G(1) phase of the cell cycle. Some cells acquired GPA expression before they went through cell division. This was completely blocked by LY294002, implying an inhibitory effect on maturation. In addition, LY294002 completely blocked the viability-enhancing effect of EPO in CD34(+)CD71(+)CD45RA(-) erythroid progenitors. LY294002 and various inhibitors of PKC completely suppressed the EPO-induced increase in the activity of Akt kinase, a direct downstream target of PI3K. CONCLUSIONS: Our results point to an important role for PI3K in mediating EPO-induced survival, proliferation, and possibly maturation of early erythroid progenitors.


Asunto(s)
Células Precursoras Eritroides/efectos de los fármacos , Eritropoyesis/fisiología , Eritropoyetina/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Serina-Treonina Quinasas , Adulto , Antígenos CD34/análisis , Antígenos de Diferenciación/análisis , Butadienos/farmacología , Carbazoles/farmacología , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Precursoras Eritroides/citología , Eritropoyesis/efectos de los fármacos , Eritropoyetina/antagonistas & inhibidores , Flavonoides/farmacología , Glicoforinas/análisis , Células Madre Hematopoyéticas/citología , Humanos , Indoles/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/fisiología , Morfolinas/farmacología , Naftalenos/farmacología , Nitrilos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Proteína Quinasa C-alfa , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Estaurosporina/farmacología , Factor de Células Madre/antagonistas & inhibidores , Factor de Células Madre/farmacología
18.
Sci Rep ; 5: 13001, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26306624

RESUMEN

We have previously demonstrated that activation of the cyclic adenosine monophosphate (cAMP) pathway kills multiple myeloma (MM) cells both in vitro and in vivo. In the present study we have investigated the potential of enhancing the killing of MM cell lines and primary MM cells by combining the cAMP-elevating compound forskolin with the commonly used MM therapeutic drugs melphalan, cyclophosphamide, doxorubicin, bortezomib and dexamethasone. We observed that forskolin potentiated the killing induced by all the tested agents as compared to treatment with the single agents alone. In particular, forskolin had a synergistic effect on the dexamethasone-responsive cell lines H929 and OM-2. By knocking down the proapoptotic BCL-2 family member BIM, we proved this protein to be involved in the synergistic induction of apoptosis by dexamethasone and forskolin. The ability of forskolin to maintain the killing of MM cells even at lower concentrations of the conventional agents suggests that forskolin may be used to diminish treatment-associated side effects. Our findings support a potential role of forskolin in combination with current conventional agents in the treatment of MM.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Colforsina/administración & dosificación , Dexametasona/administración & dosificación , Proteínas de la Membrana/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Mieloma Múltiple/patología , Resultado del Tratamiento
19.
J Neuroimmunol ; 278: 11-8, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595247

RESUMEN

We have explored the beneficial effects of retinoic acid (RA) on B cells from multiple sclerosis (MS) patients. When co-stimulated via the toll-like receptors (TLRs) TLR9 and RP105, MS B cells secreted less of the anti-inflammatory cytokine interleukin 10 (IL-10) compared to B cells from healthy controls. Importantly, RA enhanced the secretion of IL-10 by MS-derived B cells without affecting the levels of the pro-inflammatory cytokine TNF-α. RA revealed the same ability to induce IL-10 as did interferon-ß-1b (IFN-ß-1b), and B-cells from patients treated with glatiramer acetate or IFN-ß-1b still displayed the beneficial effects of RA on the IL-10/TNF-α ratio.


Asunto(s)
Antígenos CD/farmacología , Linfocitos B/efectos de los fármacos , Interleucina-10/metabolismo , Queratolíticos/farmacología , Esclerosis Múltiple Recurrente-Remitente/patología , Tretinoina/farmacología , Adulto , Anciano , Antígenos CD19/metabolismo , Linfocitos B/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Acetato de Glatiramer , Humanos , Inmunosupresores/farmacología , Persona de Mediana Edad , Péptidos/farmacología , Receptor Toll-Like 9/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Autophagy ; 11(3): 460-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25749095

RESUMEN

In the present study we have established a vital role of autophagy in retinoic acid (RA)-induced differentiation of toll-like receptor (TLR)-stimulated human B cells into Ig-secreting cells. Thus, RA enhanced autophagy in TLR9- and CD180-stimulated peripheral blood B cells, as revealed by increased levels of the autophagosomal marker LC3B-II, enhanced colocalization between LC3B and the lysosomal marker Lyso-ID, by a larger percentage of cells with more than 5 characteristic LC3B puncta, and by the concomitant reduction in the level of SQSTM1/p62. Furthermore, RA induced expression of the autophagy-inducing protein ULK1 at the transcriptional level, in a process that required the retinoic acid receptor RAR. By inhibiting autophagy with specific inhibitors or by knocking down ULK1 by siRNA, the RA-stimulated IgG production in TLR9- and CD180-mediated cells was markedly reduced. We propose that the identified prominent role of autophagy in RA-mediated IgG-production in normal human B cells provides a novel mechanism whereby vitamin A exerts its important functions in the immune system.


Asunto(s)
Autofagia , Linfocitos B/metabolismo , Inmunoglobulina G/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Toll-Like/metabolismo , Tretinoina/química , Antígenos CD/metabolismo , Antígenos CD19/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia , Linfocitos B/inmunología , Diferenciación Celular/efectos de los fármacos , Islas de CpG , Humanos , Sistema Inmunológico , Activación de Linfocitos/inmunología , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Oligonucleótidos/química , ARN Interferente Pequeño/química , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 9/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA