Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 7: e2132, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26962682

ABSTRACT

Evasion of apoptosis is critical for tumorigenesis, and sustained survival of nascent neoplastic cells may depend upon the endogenous levels of pro-survival BCL-2 family members. Indeed, previous studies using gene-targeted mice revealed that BCL-XL, but surprisingly not BCL-2, is critical for the development of c-MYC-induced pre-B/B lymphomas. However, it remains unclear whether another pro-survival BCL-2 relative contributes to their development. MCL-1 is an intriguing candidate, because it is required for cell survival during early B-lymphocyte differentiation. It is expressed abnormally high in several types of human B-cell lymphomas and is implicated in their resistance to chemotherapy. To test the B-cell intrinsic requirement for endogenous MCL-1 in lymphoma development, we conditionally deleted Mcl-1 in B-lymphoid cells of Eµ-Myc transgenic mice. We found that MCL-1 loss in early B-lymphoid progenitors delayed MYC-driven lymphomagenesis. Moreover, the lymphomas that arose when MCL-1 levels were diminished appeared to have been selected for reduced levels of BIM and/or increased levels of BCL-XL. These results underscore the importance of MCL-1 in lymphoma development and show that alterations in the levels of other cell death regulators can compensate for deficiencies in MCL-1 expression.


Subject(s)
Lymphoma, B-Cell/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplastic Stem Cells/metabolism , Precursor Cells, B-Lymphoid/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplastic Stem Cells/pathology , Precursor Cells, B-Lymphoid/pathology , Proto-Oncogene Proteins c-myc/genetics
2.
Oncogene ; 35(29): 3866-71, 2016 07 21.
Article in English | MEDLINE | ID: mdl-26640149

ABSTRACT

The tumor suppressor p53 is mutated in ~50% of human cancers. P53 is activated by a range of stimuli and regulates several cellular processes, including apoptotic cell death, cell cycle arrest, senescence and DNA repair. P53 induces apoptosis via transcriptional induction of the BH3-only proteins PUMA (p53-upregulated modulator of apoptosis) and NOXA, and cell cycle arrest via p21. Induction of these processes was proposed to be critical for p53-mediated tumor suppression. It is therefore surprising that mice lacking PUMA, NOXA and p21, as well as mice bearing mutations in p53 that impair the transcriptional activation of these genes, are not tumor prone, unlike mice lacking p53 function, which spontaneously develop tumors with 100% incidence. These p53 target genes and the processes they regulate may, however, impact differently on tumor development depending on the oncogenic drivers. For example, loss of PUMA enhances c-MYC-driven lymphoma development in mice, but, interestingly, the acceleration was less impressive compared with that caused by the loss of even a single p53 allele. Different studies have reported that loss of p21 can accelerate, delay or have no impact on tumorigenesis. In an attempt to resolve this controversy, we examined whether loss of p21-mediated cell cycle arrest cooperates with PUMA deficiency in accelerating lymphoma development in Eµ-Myc mice (overexpressing c-MYC in B-lymphoid cells). We found that Eµ-Myc mice lacking both p21 and PUMA (Eµ-Myc;Puma(-/-);p21(-/-)) developed lymphoma at a rate comparable to Eµ-Myc;Puma(-/-) animals, notably with considerably longer latency than Eµ-Myc;p53(+/-)mice. Loss of p21 had no impact on the numbers, cycling or survival of pre-leukemic Eµ-Myc B-lymphoid cells, even when PUMA was lost concomitantly. These results demonstrate that even in the context of deregulated c-MYC expression, p53 must suppress tumor development by activating processes apart from, or in addition to, PUMA-mediated apoptosis and p21-induced cell cycle arrest.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , B-Lymphocytes/metabolism , Blotting, Western , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Kaplan-Meier Estimate , Loss of Heterozygosity , Lymphoma/metabolism , Lymphoma/pathology , Mice, Knockout , Mice, Transgenic , Mutation , Precursor Cells, B-Lymphoid/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
3.
Cell Death Dis ; 6: e1938, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26492371

ABSTRACT

It is well established that BAX and BAK play crucial, overlapping roles in the intrinsic pathway of apoptosis. Gene targeted mice lacking both BAX and BAK have previously been generated, but the majority of these animals died perinatally. BOK is a poorly studied relative of BAX and BAK that shares extensive amino acid sequence homology to both proteins, but its function remains largely unclear to date. To determine whether BOK plays an overlapping role with BAX and BAK, we utilized a hematopoietic reconstitution model where lethally irradiated wild type mice were transplanted with Bok(-/-)Bax(-/-)Bak(-/-) triple knockout (TKO) fetal liver cells, and compared alongside mice reconstituted with a Bax(-/-)Bak(-/-) double knockout (DKO) hematopoietic compartment. We report here that mice with a TKO and DKO hematopoietic system died at a similar rate and much earlier than control animals, mostly due to severe autoimmune pathology. Both TKO and DKO reconstituted mice also had altered frequencies of various leukocyte subsets in the thymus, bone marrow and spleen, displayed leukocyte infiltrates and autoimmune pathology in multiple tissues, as well as elevated levels of anti-nuclear autoantibodies. Interestingly, the additional deletion of BOK (on top of BAX and BAK loss) led to a further increase in peripheral blood lymphocytes, as well as enhanced lymphoid infiltration in some organs. These findings suggest that BOK may have some functions that are redundant with BAX and BAK in the hematopoietic system.


Subject(s)
Hematopoietic System/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , Animals , Apoptosis/genetics , Blood Cell Count , Chimera , Gene Deletion , Gene Knockout Techniques , Leukocytes/cytology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
Oncogene ; 34(14): 1872-6, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-24858047

ABSTRACT

Genomic analyses revealed that many cancers have acquired abnormalities in their expression of pro- or anti-apoptotic members of the BCL-2 protein family. It is, however, unknown whether changes in pro- or anti-apoptotic BCL-2 family members have similar impact on tumorigenesis or whether changes in one subgroup have disproportionate impact. We compared the consequences of concomitant loss of anti-apoptotic Bclx and pro-apoptotic Bim on MYC-induced lymphomagenesis. Whereas only loss of both Bclx alleles markedly forestalled tumorigenesis, loss of a single Bim allele overcame this blockade. Conversely, loss of even a single Bim allele sufficed to substantially accelerate lymphomagenesis, and only loss of both but not loss of a single allele of Bclx could attenuate this acceleration. The evidence that modest (two-fold) monoallelic changes in the expression of at least some BH3-only proteins can profoundly impact tumorigenesis suggests that such aberrations, imposed by epigenetic or genetic changes, may expedite tumorigenesis more effectively than elevated expression of pro-survival BCL-2 family members. These findings further our understanding of the mechanisms of lymphomagenesis and possibly also cancer therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Cell Transformation, Neoplastic/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/genetics , bcl-X Protein/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Survival/genetics , Female , Lymphoma/genetics , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism , bcl-X Protein/metabolism
5.
Cell Death Differ ; 20(1): 57-63, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22814621

ABSTRACT

As many oncogenic changes, such as Myc overexpression, promote apoptosis, the survival of emerging neoplastic clones may often initially depend upon endogenous levels of particular pro-survival members of the Bcl-2 protein family. Pertinently, we recently showed that in lymphoma-prone Eµ-myc transgenic mice, which overexpress Myc in all B-lymphoid cells, endogenous Bcl-x(L) is critical for the survival, as well as the expansion of preneoplastic B-lymphoid cells and the development of malignant disease. This discovery raised the possibility that pharmacological blockade of Bcl-x(L) might impede Myc-driven lymphoma development. Indeed, we report here that treatment of preleukaemic Eµ-myc transgenic mice with the Bcl-2 homology (BH)3 mimetic drug ABT-737, which inhibits Bcl-x(L), as well as Bcl-2 and Bcl-w, augmented apoptosis of preneoplastic B-lymphoid cells, reduced their numbers and greatly prolonged lymphoma-free survival. These findings reveal that BH3 mimetic drugs may provide a prophylactic strategy to prevent the development of certain tumours, particularly those driven by deregulated Myc expression. Moreover, such treatment may help in the management of patients with hereditary cancer syndromes and perhaps also in the prevention of tumour relapses.


Subject(s)
Biphenyl Compounds/pharmacology , Lymphoma, B-Cell/prevention & control , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Female , Hematopoiesis/drug effects , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism
6.
Cell Death Differ ; 19(4): 623-32, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21997189

ABSTRACT

The tumour suppressor p53 transcriptionally regulates a range of target genes that control cell growth and survival. Mutations of p53 have been implicated in the development of approximately 50% of human cancers, including those instigated by exposure to mutagens. Although numerically rare, cancers can arise as a consequence of inherited mutations, such as in the Li-Fraumeni syndrome, which is caused by mutation of one p53 allele. Gene-targeted mice deficient for p53 have been generated to study this familial cancer syndrome. On a C57BL/6 background, p53-deficient mice develop primarily thymic lymphoma and more rarely sarcoma. Evasion of apoptosis is considered to be essential for neoplastic transformation. As proteins of the Bcl-2 family are the critical regulators of apoptosis, we investigated the role of the pro-survival members Bcl-2, Bcl-x(L) and Bcl-w in cancer development in p53(+/-) and p53(-/-) mice by testing whether ABT-737, a pharmacological inhibitor of these proteins, could prevent or delay tumourigenesis. Our studies showed that ABT-737 prophylaxis only caused a minor delay and reduction in γ-radiation-induced thymic lymphoma development in p53(-/-) mice, but this was accompanied by a concomitant increase in sarcoma. These data show that, collectively, Bcl-2, Bcl-x(L) and Bcl-w have only minor roles in thymic lymphoma development elicited by defects in p53, and this may indicate that Mcl-1 and/or A1 may feature more prominently in this process.


Subject(s)
Biomimetic Materials/pharmacology , Biphenyl Compounds/pharmacology , Lymphoma/metabolism , Neoplasms, Radiation-Induced/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Nitrophenols/pharmacology , Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Thymus Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/antagonists & inhibitors , Alleles , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins , Humans , Lymphoma/genetics , Lymphoma/prevention & control , Mice , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/prevention & control , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/prevention & control , Piperazines/pharmacology , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Thymus Neoplasms/genetics , Thymus Neoplasms/prevention & control , Tumor Suppressor Protein p53/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL