Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(1): 105509, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042493

ABSTRACT

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.


Subject(s)
Acetylcysteine , Cyclic AMP , Guanine Nucleotide Exchange Factors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Thionucleotides , Animals , Child , Humans , Mice , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP/therapeutic use , DNA/drug effects , Guanine Nucleotide Exchange Factors/agonists , Mice, Inbred NOD , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Male , Female , Child, Preschool , Thionucleotides/pharmacology , Thionucleotides/therapeutic use , DNA Damage , Drug Therapy, Combination
2.
Mol Cancer Res ; 20(3): 400-411, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34880123

ABSTRACT

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.


Subject(s)
NAD , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Autophagy , Cell Survival , Child , Humans , Mice , Mice, Inbred NOD , Poly (ADP-Ribose) Polymerase-1/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Reactive Oxygen Species
3.
J Immunol ; 204(8): 2133-2142, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32188759

ABSTRACT

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.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Reactive Oxygen Species/metabolism , Toll-Like Receptors/immunology , Acetylcysteine/pharmacology , Benzoxazoles/pharmacology , Humans , Reactive Oxygen Species/antagonists & inhibitors , Triazoles/pharmacology
4.
Blood Adv ; 3(21): 3181-3190, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31698450

ABSTRACT

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.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Indomethacin/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Biomarkers , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Child , Child, Preschool , Dinoprostone/blood , Disease Models, Animal , Disease Progression , Humans , Immunophenotyping , Male , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
5.
Oncotarget ; 9(54): 30434-30449, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30100998

ABSTRACT

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.

6.
PLoS One ; 12(10): e0185708, 2017.
Article in English | MEDLINE | ID: mdl-28973009

ABSTRACT

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.


Subject(s)
B-Lymphocytes/physiology , Common Variable Immunodeficiency/genetics , DNA Damage , Infrared Rays , Toll-Like Receptor 9/physiology , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/genetics , Case-Control Studies , Humans
7.
Cell Cycle ; 16(10): 947-956, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28379780

ABSTRACT

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.


Subject(s)
A Kinase Anchor Proteins/genetics , M Phase Cell Cycle Checkpoints/genetics , Nuclear Pore Complex Proteins/genetics , Proteomics , Proto-Oncogene Proteins/genetics , Chromosome Segregation/genetics , HeLa Cells , Humans , Mitosis/genetics , Nuclear Pore/genetics , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
8.
J Nutr Biochem ; 27: 153-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26419686

ABSTRACT

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.


Subject(s)
Coffee , NF-kappa B/metabolism , Prostatic Neoplasms/pathology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Heterografts , Humans , Male , Mice
9.
Sci Rep ; 5: 13001, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26306624

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Colforsin/administration & dosage , Dexamethasone/administration & dosage , Membrane Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Proto-Oncogene Proteins/metabolism , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bcl-2-Like Protein 11 , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Multiple Myeloma/pathology , Treatment Outcome
10.
Autophagy ; 11(3): 460-71, 2015.
Article in English | MEDLINE | ID: mdl-25749095

ABSTRACT

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.


Subject(s)
Autophagy , B-Lymphocytes/metabolism , Immunoglobulin G/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptors/metabolism , Tretinoin/chemistry , Antigens, CD/metabolism , Antigens, CD19/metabolism , Autophagy-Related Protein-1 Homolog , B-Lymphocytes/immunology , Cell Differentiation/drug effects , CpG Islands , Humans , Immune System , Lymphocyte Activation/immunology , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Oligonucleotides/chemistry , RNA, Small Interfering/chemistry , Receptors, Retinoic Acid/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 9/metabolism , Transcription, Genetic
11.
J Neuroimmunol ; 278: 11-8, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25595247

ABSTRACT

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.


Subject(s)
Antigens, CD/pharmacology , B-Lymphocytes/drug effects , Interleukin-10/metabolism , Keratolytic Agents/pharmacology , Multiple Sclerosis, Relapsing-Remitting/pathology , Tretinoin/pharmacology , Adult , Aged , Antigens, CD19/metabolism , B-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Glatiramer Acetate , Humans , Immunosuppressive Agents/pharmacology , Middle Aged , Peptides/pharmacology , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Mol Cancer ; 14: 14, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25623255

ABSTRACT

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.


Subject(s)
DNA Damage , Dinoprostone/metabolism , Mesenchymal Stem Cells/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Death , Cell Line, Tumor , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Models, Biological , Signal Transduction
13.
Cancer Lett ; 335(2): 323-31, 2013 Jul 28.
Article in English | MEDLINE | ID: mdl-23454584

ABSTRACT

There is a continuous search for new therapeutic targets for treatment of multiple myeloma (MM). Here we investigated the mechanisms involved in cAMP-induced apoptosis of human MM cells. cAMP-increasing agents rapidly inhibited activation of JAK1 and its substrate STAT3. In line with STAT3 being a regulator of Mcl-1 transcription, the expression of this pro-survival factor was rapidly and selectively reduced. Notably, exogenous interleukin-6 neither prevented the inhibition of JAK1/STAT3 nor the death of MM cells induced by cAMP. Our results suggest that cAMP-mediated killing of MM cells involves inhibition of the JAK/STAT pathway, making the cAMP-pathway a promising target for treatment of MM.


Subject(s)
Colforsin/pharmacology , Cyclic AMP/metabolism , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis , Cell Line, Tumor , Down-Regulation , Humans , Interleukin-6/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Multiple Myeloma/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Syndecan-1/metabolism
14.
Int J Oncol ; 42(5): 1815-21, 2013 May.
Article in English | MEDLINE | ID: mdl-23483263

ABSTRACT

Activation of cAMP signalling potently inhibits DNA damage-induced apoptosis in acute lymphoblastic leukemia cells by promoting the turnover of p53 protein. Recently, we showed that the cAMP-induced destabilization of p53 in DNA-damaged cells occurs as a result of enhanced interaction between p53 and HDM2. In this report, we present results showing that increased levels of cAMP in cells with DNA damage enhances the deacetylation of p53, an event that facilitates the interaction of p53 with HDM2, thus annulling the stabilizing effect of DNA damage on p53. The combined inhibition of the HDAC and SIRT1 deacetylases abolished the cAMP-mediated deacetylation of p53, implying that cAMP-mediated deacetylation of p53 is dependent on the activity of these two classes of histone deacetylases. Importantly, diminishing the activity of HDACs and SIRT1 was also found to reverse the inhibitory effect of cAMP on the DNA damage-induced p53 stabilization and apoptosis, suggesting the involvement of the p53 acetylation pathway in the anti-apoptotic effect of cAMP signalling.


Subject(s)
Apoptosis/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Cell Line, Tumor , Cyclic AMP/metabolism , DNA Damage/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylases/metabolism , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction , Sirtuin 1/biosynthesis , Tumor Suppressor Protein p53/biosynthesis
15.
Blood ; 121(10): 1805-13, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23299313

ABSTRACT

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.


Subject(s)
Apoptosis , Blast Crisis/pathology , Cyclic AMP/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cells, B-Lymphoid/cytology , Tumor Suppressor Protein p53/metabolism , Adolescent , Adult , Blast Crisis/drug therapy , Blast Crisis/metabolism , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Colforsin/pharmacology , DNA Damage/physiology , DNA Damage/radiation effects , Dinoprostone/pharmacology , Female , Humans , Infant , Male , Oxytocics/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/metabolism , Tumor Suppressor Protein p53/genetics , Young Adult
16.
Cell Immunol ; 279(1): 87-95, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23103284

ABSTRACT

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.


Subject(s)
Antigens, CD/metabolism , B-Lymphocytes/drug effects , Cell Proliferation/drug effects , Immunoglobulin G/biosynthesis , Toll-Like Receptor 9/metabolism , Tretinoin/pharmacology , Antibodies/pharmacology , Antigens, CD/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunologic Memory/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Oligodeoxyribonucleotides/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
17.
Autophagy ; 7(10): 1199-211, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21750416

ABSTRACT

Autophagy plays an important role in cellular remodelling during differentiation and development, however little is known about its regulation in stem cells. Here we show that cAMP, a well-known differentiation factor for mesenchymal stem cells (MSCs), is also a potent inducer of autophagy in these cells. We have previously shown that activation of the cAMP-signaling pathway inhibits proliferation of MSCs despite induction of the cell cycle component cyclin E. Here, we demonstrate a critical role of cyclin E in the induction of autophagy. Our data suggest a model in which cAMP-signaling via ERK-mediated induction of cyclin E leads to enhanced perinuclear recruitment of Beclin 1 and formation of autophagosomes. Given the roles of deregulated autophagy in neurodegenerative disorders and cAMP as a neurogenic inducer, identification of this novel autophagocytic pathway may provide new targets for intervention against neurological disorders.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy , Cyclic AMP/metabolism , Cyclin E/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Membrane Proteins/metabolism , Adipose Tissue/cytology , Beclin-1 , Cell Differentiation , Cell Proliferation , Dose-Response Relationship, Drug , Humans , Nervous System Diseases/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors
18.
Neoplasia ; 13(7): 653-63, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21750659

ABSTRACT

The tumor suppressor p53 provides an important barrier to the initiation and maintenance of cancers. As a consequence, p53 function must be inactivated for a tumor to develop. This is achieved by mutation in approximately 50% of cases and probably by functional inactivation in the remaining cases. We have previously shown that the second messenger cAMP can inhibit DNA damage-induced wild-type p53 accumulation in acute lymphoblastic leukemia cells, leading to a profound reduction of their apoptotic response. In the present article, we provide a mechanistic insight into the regulation of p53 levels by cAMP. We show that increased levels of cAMP augment the binding of p53 to its negative regulator HDM2, overriding the DNA damage-induced dissociation of p53 from HDM2. This results in maintained levels of p53 ubiquitination and proteasomal degradation, which in turn counteracts the DNA damage-induced stabilization of the p53 protein. The apoptosis inhibitory effect of cAMP is further shown to depend on this effect on p53 levels. These findings potentially implicate deregulation of cAMP signaling as a candidate mechanism used by transformed cells to quench the p53 response while retaining wild-type p53.


Subject(s)
Cyclic AMP/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Stress, Physiological/physiology , Tumor Suppressor Protein p53/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , DNA Damage/drug effects , DNA Damage/physiology , HCT116 Cells , Humans , Models, Biological , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Stress, Physiological/drug effects , Stress, Physiological/genetics , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Ubiquitination/drug effects , Ubiquitination/genetics
19.
Blood ; 114(3): 608-18, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19451550

ABSTRACT

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.


Subject(s)
Apoptosis , Cyclic AMP/metabolism , DNA Damage , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/biosynthesis , Cell Cycle , Cell Line, Tumor , Humans , Radiation, Ionizing
20.
Immunology ; 126(4): 514-22, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18778286

ABSTRACT

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.


Subject(s)
Interleukin-2/biosynthesis , Lymphocyte Activation/drug effects , T-Lymphocytes/drug effects , Tretinoin/pharmacology , Animals , Benzoates/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Chromans/pharmacology , Female , Gene Expression Regulation/drug effects , Interleukin-2/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/immunology , Spleen/immunology , T-Lymphocytes/immunology , Vitamin A Deficiency/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...