Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Breast Cancer Res ; 19(1): 115, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-29047390

ABSTRACT

BACKGROUND: Dysregulated DNA repair and cell proliferation controls are essential driving forces in mammary tumorigenesis. BCCIP was originally identified as a BRCA2 and CDKN1A interacting protein that has been implicated in maintenance of genomic stability, cell cycle regulation, and microtubule dynamics. The aims of this study were to determine whether BCCIP deficiency contributes to mammary tumorigenesis, especially for a subset of breast cancers with 53BP1 abnormality, and to reveal the mechanistic implications of BCCIP in breast cancer interventions. METHODS: We analyzed the BCCIP protein level in 470 cases of human breast cancer to determine the associations between BCCIP and 53BP1, p53, and subtypes of breast cancer. We further constructed a unique BCCIP knockdown mouse model to determine whether a partial BCCIP deficiency leads to spontaneous breast cancer formation. RESULTS: We found that the BCCIP protein level is downregulated in 49% of triple-negative breast cancer and 25% of nontriple-negative breast cancer. The downregulation of BCCIP is mutually exclusive with p53 mutations but concurrent with 53BP1 loss in triple-negative breast cancer. In a K14-Cre-mediated conditional BCCIP knockdown mouse model, we found that BCCIP downregulation causes a formation of benign modules in the mammary glands, resembling the epidermal inclusion cyst of the breast. However, the majority of these benign lesions remain indolent, and only ~ 10% of them evolve into malignant tumors after a long latency. This tumor progression is associated with a loss of 53BP1 and p16 expression. BCCIP knockdown did not alter the latency of mammary tumor formation induced by conditional Trp53 deletion. CONCLUSIONS: Our data suggest a confounding role of BCCIP deficiency in modulating breast cancer development by enhancing tumor initiation but hindering progression. Furthermore, secondary genetic alternations may overcome the progression suppression imposed by BCCIP deficiency through a synthetic viability mechanism.


Subject(s)
Calcium-Binding Proteins/genetics , Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Mammary Glands, Human/pathology , Nuclear Proteins/genetics , Animals , BRCA2 Protein/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Triple Negative Breast Neoplasms , Tumor Suppressor Protein p53/genetics , Tumor Suppressor p53-Binding Protein 1/genetics
2.
Cancer Res ; 73(23): 7122-33, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24145349

ABSTRACT

Dysfunctions of genome caretaker genes contribute to genomic instability and tumor initiation. Because many of the caretaker genes are also essential for cell viability, permanent loss of function of these genes would prohibit further tumor progression. How essential caretaker genes contribute to tumorigenesis is not fully understood. Here, we report a "hit-and-run" mode of action for an essential caretaker gene in tumorigenesis. Using a BRCA2-interacting protein BCCIP as the platform, we found that a conditional BCCIP knockdown and concomitant p53 deletion caused rapid development of medulloblastomas, which bear a wide spectrum of alterations involving the Sonic Hedgehog (Shh) pathway, consistent with a caretaker responsibility of BCCIP on genomic integrity. Surprisingly, the progressed tumors have spontaneously lost the transgenic BCCIP knockdown cassette and restored BCCIP expression. Thus, a transient downregulation of BCCIP, but not necessarily a permanent mutation, is sufficient to initiate tumorigenesis. After the malignant transformation has been accomplished and autonomous cancer growth has been established, BCCIP reverses its role from a tumor-initiation suppressor to become a requisite for progression. This exemplifies a new type of tumor suppressor, which is distinct from the classical tumor suppressors that are often permanently abrogated during tumorigenesis. It has major implications on how a nonmutagenic or transient regulation of essential caretaker gene contributes to tumorigenesis. We further suggest that BCCIP represents a paradoxical class of modulators for tumorigenesis as a suppressor for initiation but a requisite for progression (SIRP).


Subject(s)
Calcium-Binding Proteins/physiology , Carcinogenesis/genetics , Cell Cycle Proteins/physiology , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/physiology , Animals , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Disease Progression , Genes, Tumor Suppressor/physiology , Hedgehog Proteins/physiology , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Signal Transduction/physiology
3.
PLoS One ; 7(1): e30638, 2012.
Article in English | MEDLINE | ID: mdl-22292003

ABSTRACT

Multiple DNA repair pathways are involved in the orderly development of neural systems at distinct stages. The homologous recombination (HR) pathway is required to resolve stalled replication forks and critical for the proliferation of progenitor cells during neural development. BCCIP is a BRCA2 and CDKN1A interacting protein implicated in HR and inhibition of DNA replication stress. In this study, we determined the role of BCCIP in neural development using a conditional BCCIP knock-down mouse model. BCCIP deficiency impaired embryonic and postnatal neural development, causing severe ataxia, cerebral and cerebellar defects, and microcephaly. These development defects are associated with spontaneous DNA damage and subsequent cell death in the proliferative cell populations of the neural system during embryogenesis. With in vitro neural spheroid cultures, BCCIP deficiency impaired neural progenitor's self-renewal capability, and spontaneously activated p53. These data suggest that BCCIP and its anti-replication stress functions are essential for normal neural development by maintaining an orderly proliferation of neural progenitors.


Subject(s)
Cell Cycle Proteins/physiology , Cell Proliferation , Neural Stem Cells/physiology , Neurogenesis/genetics , Neurons/physiology , Animals , Ataxia/complications , Ataxia/congenital , Ataxia/genetics , Ataxia/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Embryo, Mammalian , Glial Fibrillary Acidic Protein/genetics , Growth Disorders/complications , Growth Disorders/congenital , Growth Disorders/genetics , Growth Disorders/pathology , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Models, Biological , Neural Stem Cells/metabolism , Neurons/metabolism , Organ Specificity/genetics , Postural Balance/genetics , Promoter Regions, Genetic/genetics , Sensation Disorders/complications , Sensation Disorders/congenital , Sensation Disorders/genetics , Sensation Disorders/pathology , Stem Cells/metabolism , Stem Cells/physiology
4.
PLoS Genet ; 7(9): e1002291, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21966279

ABSTRACT

BCCIP is a BRCA2- and CDKN1A(p21)-interacting protein that has been implicated in the maintenance of genomic integrity. To understand the in vivo functions of BCCIP, we generated a conditional BCCIP knockdown transgenic mouse model using Cre-LoxP mediated RNA interference. The BCCIP knockdown embryos displayed impaired cellular proliferation and apoptosis at day E7.5. Consistent with these results, the in vitro proliferation of blastocysts and mouse embryonic fibroblasts (MEFs) of BCCIP knockdown mice were impaired considerably. The BCCIP deficient mouse embryos die before E11.5 day. Deletion of the p53 gene could not rescue the embryonic lethality due to BCCIP deficiency, but partially rescues the growth delay of mouse embryonic fibroblasts in vitro. To further understand the cause of development and proliferation defects in BCCIP-deficient mice, MEFs were subjected to chromosome stability analysis. The BCCIP-deficient MEFs displayed significant spontaneous chromosome structural alterations associated with replication stress, including a 3.5-fold induction of chromatid breaks. Remarkably, the BCCIP-deficient MEFs had a ∼20-fold increase in sister chromatid union (SCU), yet the induction of sister chromatid exchanges (SCE) was modestly at 1.5 fold. SCU is a unique type of chromatid aberration that may give rise to chromatin bridges between daughter nuclei in anaphase. In addition, the BCCIP-deficient MEFs have reduced repair of irradiation-induced DNA damage and reductions of Rad51 protein and nuclear foci. Our data suggest a unique function of BCCIP, not only in repair of DNA damage, but also in resolving stalled replication forks and prevention of replication stress. In addition, BCCIP deficiency causes excessive spontaneous chromatin bridges via the formation of SCU, which can subsequently impair chromosome segregations in mitosis and cell division.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Instability/genetics , Embryonic Development/genetics , Animals , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Cycle Proteins/genetics , Chromosome Segregation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Transgenic , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Recombination, Genetic , Sister Chromatid Exchange
SELECTION OF CITATIONS
SEARCH DETAIL
...