38 research outputs found

    Cyclin B1/Cdk1 phosphorylation of mitochondrial p53 induces anti-apoptotic response.

    Get PDF
    The pro-apoptotic function of p53 has been well defined in preventing genomic instability and cell transformation. However, the intriguing fact that p53 contributes to a pro-survival advantage of tumor cells under DNA damage conditions raises a critical question in radiation therapy for the 50% human cancers with intact p53 function. Herein, we reveal an anti-apoptotic role of mitochondrial p53 regulated by the cell cycle complex cyclin B1/Cdk1 in irradiated human colon cancer HCT116 cells with p53(+/+) status. Steady-state levels of p53 and cyclin B1/Cdk1 were identified in the mitochondria of many human and mouse cells, and their mitochondrial influx was significantly enhanced by radiation. The mitochondrial kinase activity of cyclin B1/Cdk1 was found to specifically phosphorylate p53 at Ser-315 residue, leading to enhanced mitochondrial ATP production and reduced mitochondrial apoptosis. The improved mitochondrial function can be blocked by transfection of mutant p53 Ser-315-Ala, or by siRNA knockdown of cyclin B1 and Cdk1 genes. Enforced translocation of cyclin B1 and Cdk1 into mitochondria with a mitochondrial-targeting-peptide increased levels of Ser-315 phosphorylation on mitochondrial p53, improved ATP production and decreased apoptosis by sequestering p53 from binding to Bcl-2 and Bcl-xL. Furthermore, reconstitution of wild-type p53 in p53-deficient HCT116 p53(-/-) cells resulted in an increased mitochondrial ATP production and suppression of apoptosis. Such phenomena were absent in the p53-deficient HCT116 p53(-/-) cells reconstituted with the mutant p53. These results demonstrate a unique anti-apoptotic function of mitochondrial p53 regulated by cyclin B1/Cdk1-mediated Ser-315 phosphorylation in p53-wild-type tumor cells, which may provide insights for improving the efficacy of anti-cancer therapy, especially for tumors that retain p53

    Breast cancer adaptive resistance: HER2 and cancer stem cell repopulation in a heterogeneous tumor society

    Get PDF

    HER2 serves as the marker for identifying radiation-resistant breast cancer stem cells

    No full text
    Cancer stem cells (CSCs) are shown to be able to self-renew and be resistant to radiation therapy. Elucidation of the role of CSCs in radiation-mediated repopulation may provide critical insights for development of new approaches to resensitize resistant tumor cells. Identification of biomarkers of therapy-resistant CSCs will provide approaches to target the CSCs especially in recurrent and metastatic tumors. This thesis explores to establish a new marker for breast CSCs and offers a possible mechanism that might be responsible for their radio-resistant phenotypes. Results demonstrated that 45% of cells with the feature of CD44 +/CD24-/low are HER2 positive in the MCF7 breast cancer cells survived from therapeutic irradiation due to HER2 overexpression. The radiation-surviving cells with HER2+/CD44+/CD24 -/low feature showed a more aggressive growth with radioresistant phenotype than the HER2−/CD44+/CD24 -/low counterparts. Proteomics analysis revealed unique protein expression profiles for HER2+/CD44+/CD24-/low and HER2−/CD44+/CD24-/low fractions. Co-expression of HER2 and CD44 was increased in the recurrent tumors compared to the primary tumors from 40 patients diagnosed with breast cancers and the HER2+/CD44+/CD24-/low MCF7 cells showed an enhanced tumorigenesis in NSG mice. Thus, HER2 +/CD44+/CD24-/low shall be considered as a CSC marker for therapy-resistant breast tumors and current treatment designs should be reassessed incorporating HER2’s herein demonstrated role in radioresistant breast CSCs

    Early-Onset Progressive Myoclonic Epilepsy With Dystonia Mapping to 16pter-p13.3

    No full text
    The authors present three patients from a consanguineous family afflicted with novel recessive myoclonic epilepsy characterized by very early onset and a steadily progressive course. The onset is in early infancy, and death occurs in the first decade. In addition to various types of myoclonic seizures, episodic phenomena such as dystonias, postictal enduring hemipareses, autonomic involvements, and periods of obtundation and lethargy were also observed. Developmental and neurological retardation, coupled with systemic infections, leads to a full deterioration. The authors designated the disease progressive myoclonic epilepsy with dystonia (PMED). A genome scan for the family and subsequent fine mapping localized the gene responsible for the disease to the most telomeric 6.73 mega base pairs at the p-terminus of chromosome 16, with a maximum multipoint logarithm-of-odds score of 7.83 and a maximum two-point score of 4.25. A candidate gene was analyzed for mutations in patients, but no mutation was found

    Cyclin B1/Cdk1 phosphorylation of mitochondrial p53 induces anti-apoptotic response.

    Get PDF
    The pro-apoptotic function of p53 has been well defined in preventing genomic instability and cell transformation. However, the intriguing fact that p53 contributes to a pro-survival advantage of tumor cells under DNA damage conditions raises a critical question in radiation therapy for the 50% human cancers with intact p53 function. Herein, we reveal an anti-apoptotic role of mitochondrial p53 regulated by the cell cycle complex cyclin B1/Cdk1 in irradiated human colon cancer HCT116 cells with p53(+/+) status. Steady-state levels of p53 and cyclin B1/Cdk1 were identified in the mitochondria of many human and mouse cells, and their mitochondrial influx was significantly enhanced by radiation. The mitochondrial kinase activity of cyclin B1/Cdk1 was found to specifically phosphorylate p53 at Ser-315 residue, leading to enhanced mitochondrial ATP production and reduced mitochondrial apoptosis. The improved mitochondrial function can be blocked by transfection of mutant p53 Ser-315-Ala, or by siRNA knockdown of cyclin B1 and Cdk1 genes. Enforced translocation of cyclin B1 and Cdk1 into mitochondria with a mitochondrial-targeting-peptide increased levels of Ser-315 phosphorylation on mitochondrial p53, improved ATP production and decreased apoptosis by sequestering p53 from binding to Bcl-2 and Bcl-xL. Furthermore, reconstitution of wild-type p53 in p53-deficient HCT116 p53(-/-) cells resulted in an increased mitochondrial ATP production and suppression of apoptosis. Such phenomena were absent in the p53-deficient HCT116 p53(-/-) cells reconstituted with the mutant p53. These results demonstrate a unique anti-apoptotic function of mitochondrial p53 regulated by cyclin B1/Cdk1-mediated Ser-315 phosphorylation in p53-wild-type tumor cells, which may provide insights for improving the efficacy of anti-cancer therapy, especially for tumors that retain p53
    corecore