Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Antonino B. D'Assoro is active.

Publication


Featured researches published by Antonino B. D'Assoro.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Centrosome amplification drives chromosomal instability in breast tumor development

Wilma L. Lingle; Susan L. Barrett; Vivian Negron; Antonino B. D'Assoro; Kelly Boeneman; Wanguo Liu; Clark M. Whitehead; Carol Reynolds; Jeffrey L. Salisbury

Earlier studies of invasive breast tumors have shown that 60–80% are aneuploid and ≈80% exhibit amplified centrosomes. In this study, we investigated the relationship of centrosome amplification with aneuploidy, chromosomal instability, p53 mutation, and loss of differentiation in human breast tumors. Twenty invasive breast tumors and seven normal breast tissues were analyzed by fluorescence in situ hybridization with centromeric probes to chromosomes 3, 7, and 17. We analyzed these tumors for both aneuploidy and unstable karyotypes as determined by chromosomal instability. The results were then tested for correlation with three measures of centrosome amplification: centrosome size, centrosome number, and centrosome microtubule nucleation capacity. Centrosome size and centrosome number both showed a positive, significant, linear correlation with aneuploidy and chromosomal instability. Microtubule nucleation capacity showed no such correlation, but did correlate significantly with loss of tissue differentiation. Centrosome amplification was detected in in situ ductal carcinomas, suggesting that centrosome amplification is an early event in these lesions. Centrosome amplification and chromosomal instability occurred independently of p53 mutation, whereas p53 mutation was associated with a significant increase in centrosome microtubule nucleation capacity. Together, these results demonstrate that independent aspects of centrosome amplification correlate with chromosomal instability and loss of tissue differentiation and may be involved in tumor development and progression. These results further suggest that aspects of centrosome amplification may have clinical diagnostic and/or prognostic value and that the centrosome may be a potential target for cancer therapy.


Oncogene | 2002

Centrosome amplification and the development of cancer.

Antonino B. D'Assoro; Wilma L. Lingle; Jeffrey L. Salisbury

Recent studies have implicated centrosome amplification in the origin of chromosomal instability during tumor development. Theodor Boveri first suggested this notion nearly a century ago (Boveri, 1914). While the role of centrosome amplification in the origin of malignant tumors remains a controversial issue, several emerging lines of evidence suggest that cell cycle pathways converge on the centrosome and implicate this organelle in the control of cell cycle progression in addition to its function as a microtubule organizing center. Here we review the relationship between cell cycle regulation and centrosome amplification in the development of cancer. The centrosome influences cell structure through the nucleation and organization of cytoplasmic microtubules (Kirschner and Mitchison, 1986). Interphase cells contain a single centrosome that is typically located near the nucleus and contains of a pair of centrioles that anchor the recruitment of pericentriolar material including the microtubule nucleating protein g-tubulin (Bobinnec et al., 1998). Centrioles are small organelles (*200 nm diameter and 400 nm in length) consisting of a cylindrical array of nine triplet microtubules (Dutcher, 2001a,b). The centrosome is duplicated once, and only once, during a normal cell cycle to give rise to two centrosomes that function as the spindle poles of the dividing cell (Kellogg, 1989). This process is most clearly illustrated by duplication of the centrioles themselves. In early G1 phase of the cell cycle the two centrioles are typically oriented in a characteristic orthogonal arrangement. As cells pass the G1 restriction point and commit to DNA replication and subsequent cell division, the two centrioles separate a short distance from one another and nascent procentrioles form at the proximal end and orthogonal to each pre-existing centriole (Adams and Kilmartin, 2000; Wheatley, 1982). During G2/M phase centrosome duplication is completed and each new centrosome (i.e. mitotic spindle pole) contains one old and one new centriole. The presence of only two centrosomes in the cell as it enters mitosis ensures the equal segregation of sister chromatids to each daughter cell. Mitotic spindle poles also play a role in determining the position and orientation of the cleavage furrow and in exit from cytokinesis (Khodjakov and Rieder, 2001; Piel et al., 2001).


Breast Cancer Research and Treatment | 2002

Amplified centrosomes in breast cancer: A potential indicator of tumor aggressiveness

Antonino B. D'Assoro; Susan L. Barrett; Christopher Folk; Vivian Negron; Kelly Boeneman; Robert Busby; Clark M. Whitehead; Franca Stivala; Wilma L. Lingle; Jeffrey L. Salisbury

Molecular mechanisms leading to genomic instability and phenotypic variation during tumor development and progression are poorly understood. Such instability represents a major problem in the management of breast cancer because of its contribution to more aggressive phenotypes as well as chemoresistance. In this study we analyzed breast carcinomas and tumor-derived cell lines to determine the relationship between centrosome amplification and established prognostic factors. Our results show that centrosome amplification can arise independent of ER or p53 status and is a common feature of aneuploid breast tumors. Centrosome amplification is associated with mitotic spindle abnormalities in breast carcinomas and thus may contribute to genomic instability and the development of more aggressive phenotypes during tumor progression.


Journal of Mammary Gland Biology and Neoplasia | 2004

Centrosome Amplification and the Origin of Chromosomal Instability in Breast Cancer

Jeffrey L. Salisbury; Antonino B. D'Assoro; Wilma L. Lingle

The development and progression of aggressive breast cancer is characterized by genomic instability leading to multiple genetic defects, phenotypic diversity, chemoresistance, and poor outcome. Centrosome abnormalities have been implicated in the origin of chromosomal instability through the development of multipolar mitotic spindles. Breast tumor centrosomes display characteristic structural abnormalities, termed centrosome amplification, including: increase in centrosome number and volume, accumulation of excess pericentriolar material, supernumerary centrioles, and inappropriate phosphorylation of centrosome proteins. In addition, breast tumor centrosomes also show functional abnormalities characterized by inappropriate centrosome duplication during the cell cycle and nucleation of unusually large microtubule arrays. These observations have important implications for understanding the mechanisms underlying genomic instability and loss of cell polarity in cancer. This review focuses on the coordination of the centrosome, DNA, and cell cycles in normal cells and their deregulation resulting in centrosome amplification and chromosomal instability in the development and progression of breast cancer.


Oncogene | 2004

Genotoxic stress leads to centrosome amplification in breast cancer cell lines that have an inactive G1/S cell cycle checkpoint

Antonino B. D'Assoro; Robert Busby; Kelly M Suino; Emmanuella Delva; Gustavo J Almodovar-Mercado; H. J. Johnson; Christopher Folk; Daniel J. Farrugia; Vlad C. Vasile; Franca Stivala; Jeffrey L. Salisbury

Centrosome amplification plays a key role in the origin of chromosomal instability during cancer development and progression. In this study, breast cancer cell lines with different p53 backgrounds were used to investigate the relationship between genotoxic stress, G1/S cell cycle checkpoint integrity, and the development of centrosome amplification. Introduction of DNA damage in the MCF-7 cell line by treatment with hydroxyurea (HU) or daunorubicin (DR) resulted in the arrest of both G1/S cell cycle progression and centriole duplication. In these cells, which carry functional p53, HU treatment also led to nuclear accumulation of p53 and p21WAF1, retinoblastoma hypophosphorylation, and downregulation of cyclin A. MCF-7 cells carrying a recombinant dominant-negative p53 mutant (vMCF-7DNp53) exhibited a shortened G1 phase of the cell cycle and retained a normal centrosome phenotype. However, these cells developed amplified centrosomes following HU treatment. The MDA-MB 231 cell line, which carries mutant p53 at both alleles, showed amplified centrosomes at the outset, and developed a hyperamplified centrosome phenotype following HU treatment. In cells carrying defective p53, the development of centrosome amplification also occurred following treatment with another DNA damaging agent, DR. Taken together, these findings demonstrate that loss of p53 function alone is not sufficient to drive centrosome amplification, but plays a critical role in this process following DNA damage through abrogation of the G1/S cell cycle checkpoint. Furthermore, these studies have important clinical implications because they suggest that breast cancers with compromised p53 function may develop centrosome amplification and consequent chromosomal instability following treatment with genotoxic anticancer drugs.


Oncogene | 2015

Progesterone receptor-B enhances estrogen responsiveness of breast cancer cells via scaffolding PELP1- and estrogen receptor-containing transcription complexes

Andrea R. Daniel; A. L. Gaviglio; Todd P. Knutson; Julie H. Ostrander; Antonino B. D'Assoro; Preethi Ravindranathan; Yan Peng; Ganesh V. Raj; Douglas Yee; Carol A. Lange

Progesterone and estrogen are important drivers of breast cancer proliferation. Herein, we probed estrogen receptor-α (ER) and progesterone receptor (PR) cross-talk in breast cancer models. Stable expression of PR-B in PR-low/ER+ MCF7 cells increased cellular sensitivity to estradiol and insulin-like growth factor 1 (IGF1), as measured in growth assays performed in the absence of exogenous progestin; similar results were obtained in PR-null/ER+ T47D cells stably expressing PR-B. Genome-wide microarray analyses revealed that unliganded PR-B induced robust expression of a subset of estradiol-responsive ER target genes, including cathepsin-D (CTSD). Estradiol-treated MCF7 cells stably expressing PR-B exhibited enhanced ER Ser167 phosphorylation and recruitment of ER, PR and the proline-, glutamate- and leucine-rich protein 1 (PELP1) to an estrogen response element in the CTSD distal promoter; this complex co-immunoprecipitated with IGF1 receptor (IGFR1) in whole-cell lysates. Importantly, ER/PR/PELP1 complexes were also detected in human breast cancer samples. Inhibition of IGF1R or phosphoinositide 3-kinase blocked PR-B-dependent CTSD mRNA upregulation in response to estradiol. Similarly, inhibition of IGF1R or PR significantly reduced ER recruitment to the CTSD promoter. Stable knockdown of endogenous PR or onapristone treatment of multiple unmodified breast cancer cell lines blocked estradiol-mediated CTSD induction, inhibited growth in soft agar and partially restored tamoxifen sensitivity of resistant cells. Further, combination treatment of breast cancer cells with both onapristone and IGF1R tyrosine kinase inhibitor AEW541 was more effective than either agent alone. In summary, unliganded PR-B enhanced proliferative responses to estradiol and IGF1 via scaffolding of ER-α/PELP1/IGF1R-containing complexes. Our data provide a strong rationale for targeting PR in combination with ER and IGF1R in patients with luminal breast cancer.


Journal of Biological Chemistry | 2010

Pleiotropic Effects of p300-mediated Acetylation on p68 and p72 RNA Helicase

Steven Mooney; Apollina Goel; Antonino B. D'Assoro; Jeffrey L. Salisbury; Ralf Janknecht

Here, we demonstrate that p68 (DDX5) and p72 (DDX17), two homologous RNA helicases and transcriptional cofactors, are substrates for the acetyltransferase p300 in vitro and in vivo. Mutation of acetylation sites affected the binding of p68/p72 to histone deacetylases, but not to p300 or estrogen receptor. Acetylation additionally increased the stability of p68 and p72 RNA helicase and stimulated their ability to coactivate the estrogen receptor, thereby potentially contributing to its aberrant activation in breast tumors. Also, acetylation of p72, but not of p68 RNA helicase, enhanced p53-dependent activation of the MDM2 promoter, pointing at another mechanism of how p72 acetylation may facilitate carcinogenesis by boosting the negative p53-MDM2 feedback loop. Furthermore, blocking p72 acetylation caused cell cycle arrest and apoptosis, revealing an essential role for p72 acetylation. In conclusion, our report has identified for the first time that acetylation modulates RNA helicases and provides multiple mechanisms how acetylation of p68 and p72 may affect normal and tumor cells.


Cancer Research | 2010

Coordination of Centrosome Homeostasis and DNA Repair Is Intact in MCF-7 and Disrupted in MDA-MB 231 Breast Cancer Cells

Ilie D. Acu; Tieju Liu; Kelly Suino-Powell; Steven Mooney; Antonino B. D'Assoro; Nicholas Rowland; Alysson R. Muotri; Ricardo G. Correa; Yun Niu; Rajiv Kumar; Jeffrey L. Salisbury

When cells encounter substantial DNA damage, critical cell cycle events are halted while DNA repair mechanisms are activated to restore genome integrity. Genomic integrity also depends on proper assembly and function of the bipolar mitotic spindle, which is required for equal chromosome segregation. Failure to execute either of these processes leads to genomic instability, aging, and cancer. Here, we show that following DNA damage in the breast cancer cell line MCF-7, the centrosome protein centrin2 moves from the cytoplasm and accumulates in the nucleus in a xeroderma pigmentosum complementation group C protein (XPC)-dependent manner, reducing the available cytoplasmic pool of this key centriole protein and preventing centrosome amplification. MDA-MB 231 cells do not express XPC and fail to move centrin into the nucleus following DNA damage. Reintroduction of XPC expression in MDA-MB 231 cells rescues nuclear centrin2 sequestration and reestablishes control against centrosome amplification, regardless of mutant p53 status. Importantly, the capacity to repair DNA damage was also dependent on the availability of centrin2 in the nucleus. These observations show that centrin and XPC cooperate in a reciprocal mechanism to coordinate centrosome homeostasis and DNA repair and suggest that this process may provide a tractable target to develop treatments to slow progression of cancer and aging.


International Journal of Oncology | 2014

Inhibition of Cdk2 kinase activity selectively targets the CD44+/CD24-/Low stem-like subpopulation and restores chemosensitivity of SUM149PT triple-negative breast cancer cells

Mateusz Opyrchal; Jeffrey L. Salisbury; Ianko Iankov; Mathew P. Goetz; James A. McCubrey; Mario W. Gambino; Lorenzo Malatino; Giuseppe Puccia; James N. Ingle; Evanthia Galanis; Antonino B. D'Assoro

Inflammatory breast cancer (IBC) is an angioinvasive and most aggressive type of advanced breast cancer characterized by rapid proliferation, chemoresistance, early metastatic development and poor prognosis. IBC tumors display a triple-negative breast cancer (TNBC) phenotype characterized by centrosome amplification, high grade of chromosomal instability (CIN) and low levels of expression of estrogen receptor α (ERα), progesterone receptor (PR) and HER-2 tyrosine kinase receptor. Since the TNBC cells lack these receptors necessary to promote tumor growth, common treatments such as endocrine therapy and molecular targeting of HER-2 receptor are ineffective for this subtype of breast cancer. To date, not a single targeted therapy has been approved for non-inflammatory and inflammatory TNBC tumors and combination of conventional cytotoxic chemotherapeutic agents remains the standard therapy. IBC tumors generally display activation of epithelial to mesenchymal transition (EMT) that is functionally linked to a CD44+/CD24−/Low stem-like phenotype. Development of EMT and consequent activation of stemness programming is responsible for invasion, tumor self-renewal and drug resistance leading to breast cancer progression, distant metastases and poor prognosis. In this study, we employed the luminal ER+ MCF-7 and the IBC SUM149PT breast cancer cell lines to establish the extent to which high grade of CIN and chemoresistance were mechanistically linked to the enrichment of CD44+/CD24low/− CSCs. Here, we demonstrate that SUM149PT cells displayed higher CIN than MCF-7 cells characterized by higher percentage of structural and numerical chromosomal aberrations. Moreover, centrosome amplification, cyclin E overexpression and phosphorylation of retinoblastoma (Rb) were restricted to the stem-like CD44+/CD24−/Low subpopulation isolated from SUM149PT cells. Significantly, CD44+/CD24−/Low CSCs displayed resistance to conventional chemotherapy but higher sensitivity to SU9516, a specific cyclin-dependent kinase 2 (Cdk2) inhibitor, demonstrating that aberrant activation of cyclin E/Cdk2 oncogenic signaling is essential for the maintenance and expansion of CD44+/CD24−/Low CSC subpopulation in IBC. In conclusion, our findings propose a novel therapeutic approach to restore chemosensitivity and delay recurrence of IBC tumors based on the combination of conventional chemotherapy with small molecule inhibitors of the Cdk2 cell cycle kinase.


International Journal of Oncology | 2012

Raf-1 oncogenic signaling is linked to activation of mesenchymal to epithelial transition pathway in metastatic breast cancer cells

Alexey A. Leontovich; Shuya Zhang; Cosima Quatraro; Ianko Iankov; Pier Francesco Veroux; Mario W. Gambino; Amy C. Degnim; James A. McCubrey; James N. Ingle; Evanthia Galanis; Antonino B. D'Assoro

Aberrant activation of the Raf/MEK/MAPK pathway plays a key role in breast cancer development and progression. Dysregulation of Raf/MEK/MAPK oncogenic signaling often results from overexpression of the HER-2/Neu tyrosine kinase receptor leading to chemoendocrine resistance, development of distant metastases and ultimately poor prognosis in breast cancer patients. HER-2/Neu overexpression is also linked to activation of the epithelial to mesenchymal transition (EMT) pathway, loss of adhesion molecules and metastasis. Recently, it has been demonstrated that cancer cells that undergo EMT acquire a CD44+/CD24-/low basal cancer stem cell-like phenotype and are characterized by activation of HER-2/Neu and TGFβ oncogenic signaling pathways with increased capacity of self-renewal, drug resistance, invasion and distant metastases. Following metastatic dissemination, cancer cells re-activate certain epithelial properties through mesenchymal to epithelial transition (MET) to establish neoplastic lesions at secondary sites, although the molecular mechanisms regulating MET remain elusive. In this study we demonstrate that constitutive activation of Raf-1 oncogenic signaling induces HER-2/Neu overexpression leading to the development of distant metastases in ERα+ MCF-7 breast cancer xenografts. Importantly, development of distant metastases in xenograft models was linked to activation of the MET pathway characterized by reduced expression of EMT inducer genes (TGFB2, TWIST1 and FOXC1) and overexpression of BMB7, CXCR7 and EGR family of transcription factors. In summary, our results demonstrate for the first time that amplification of Raf/MEK/MAPK oncogenic signaling during tumor growth promotes the genesis of metastatic lesions from primary tumors by activating the mesenchymal epithelial transition.

Collaboration


Dive into the Antonino B. D'Assoro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge