Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 76(18): 5405-14, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27630139

ABSTRACT

p53 is a tumor suppressor that prevents the emergence of transformed cells by inducing apoptosis or senescence, among other responses. Its functions are regulated tightly by posttranslational modifications. Here we show that Bruton's tyrosine kinase (BTK) is a novel modulator of p53. We found that BTK is induced in response to DNA damage and p53 activation. BTK induction leads to p53 phosphorylation, which constitutes a positive feedback loop that increases p53 protein levels and enhances the transactivation of its target genes in response to stress. Inhibiting BTK reduced both p53-dependent senescence and apoptosis. Further, BTK expression also upregulated DNA damage signals and apoptosis. We conclude that despite being involved in oncogenic signals in blood malignancies, BTK has antineoplastic properties in other contexts, such as the enhancement of p53's tumor suppressor responses. Along with evidence that BTK expression correlates with good prognosis in some epithelial tumors, our findings may encourage a reevaluation of the clinical uses of BTK inhibitors in cancer therapy. Cancer Res; 76(18); 5405-14. ©2016 AACR.


Subject(s)
Apoptosis/physiology , Cellular Senescence/physiology , Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Agammaglobulinaemia Tyrosine Kinase , Blotting, Western , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chromatin Immunoprecipitation , Comet Assay , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Neoplasms/mortality , Real-Time Polymerase Chain Reaction
2.
Cancer Res ; 76(21): 6311-6319, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27634759

ABSTRACT

Chronic lymphocytic leukemia (CLL) cells multiply and become more resistant to immunochemotherapy in "proliferation centers" within tissues, whereas apoptosis occurs in the periphery. Various models recapitulate these microenvironments in vitro, such as stimulation with CD154 and IL4. Using this system, we observed a 30- to 40-fold induction of wild-type p53 protein in 50 distinct human CLL specimens tested, without the induction of either cell-cycle arrest or apoptosis. In contrast, the mRNA levels for p53 did not increase, indicating that its elevation occurred posttranscriptionally. Mechanistic investigations revealed that under the conditions studied, p53 was phosphorylated on residues associated with p53 activation and increased half-life. However, p53 protein induced in this manner could transcriptionally activate only a subset of target genes. The addition of a DNA-damaging agent further upregulated p53 protein levels, which led to apoptosis. p53 induction relied on the increase in intracellular reactive oxygen species observed after CD154 and IL4 stimulation. We propose that chronic oxidative stress is a characteristic of the microenvironment in B-cell "proliferation centers" in CLL that are capable of elevating the basal expression of p53, but to levels below the threshold needed to induce arrest or apoptosis. Our findings suggest that reactivation of the full transcriptional activities of p53 in proliferating CLL cells may offer a possible therapeutic strategy. Cancer Res; 76(21); 6311-9. ©2016 AACR.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/physiology , CD40 Ligand/pharmacology , Humans , Interleukin-4/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Transcriptional Activation , Tumor Cells, Cultured , Up-Regulation
3.
Br J Haematol ; 175(4): 641-651, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27469405

ABSTRACT

Dinaciclib is a cyclin-dependent kinase inhibitor with clinical potential in different cancers, including chronic lymphocytic leukaemia (CLL). In order to better understand its cytotoxic action, we characterized its effects on signalling pathways important for the survival of CLL cells. We found that dinaciclib induced apoptosis through the activation of caspases 8 and 9, which was independent of the presence of cytokines to mimic the environment of proliferation centres or IGVH mutation status. Moreover, treatment with dinaciclib led to the inhibition of oncogenic pathways normally activated in stimulated CLL cells, such as STAT3, NF-κB, p38, PI3K/AKT and RAF/MEK/ERK. Dinaciclib was also able to block the expression of anti-apoptotic proteins of the BCL2 family such as MCL1 and BCL-xL (also termed BCL2L1). Finally, we showed that low concentrations of dinaciclib enhanced cell sensitivity to ibrutinib and the BCL2 inhibitor ABT-199, two drugs with known effects on CLL. Taken together, our data show that dinaciclib targets multiple pro-survival signalling pathways in CLL, which provides a mechanistic explanation for its potent induction of apoptosis. They also support a therapeutic application of cyclin-dependent kinase inhibitors in CLL in combination with other relevant targeted therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cyclin-Dependent Kinases/antagonists & inhibitors , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Protein Kinase Inhibitors/therapeutic use , Pyridinium Compounds/therapeutic use , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclic N-Oxides , Humans , Indolizines , Protein Kinase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology
5.
Br J Haematol ; 160(6): 725-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23346939

ABSTRACT

There is now a plethora of new precision medicines for B-cell malignancy including 'classical' kinase inhibitors, rationally designed inhibitors of anti-apoptotic proteins and antibody or antibody drug/toxin conjugates with functional properties. Some are showing spectacular single agent activity in early phase clinical studies and may reduce or, in combination, even obviate the need for chemotherapy. Nevertheless, significant problems remain if these medicines are to be introduced into routine clinical practice in a rational and affordable manner. Firstly, precision medicines must be carefully matched in a mechanistic fashion with specific subtypes of disease. Whilst sensitivity may be predicted by the detection of key mutations or by expression of target molecules, for therapies that depend on intact intracellular signalling pathways, functional assessment on viable primary malignant cells will be necessary using assays that faithfully mimic in vivo conditions. A second, but no less important challenge is to define mechanism-based synergistic combinations associated with minimal toxicities rather than simply adding new precision medicines to existing chemotherapeutic regimens. Finally, a closer, open, two-way interaction between academic medicine and the pharmaceutical industry will be necessary to achieve these aims. Implementing such changes would change radically how and where patients with B-cell malignancies are managed.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, B-Cell/drug therapy , Lymphoma/drug therapy , Molecular Targeted Therapy/methods , Animals , Antineoplastic Agents/therapeutic use , Humans , Leukemia, B-Cell/metabolism , Lymphoma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...