Network


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

Hotspot


Dive into the research topics where Rosalie C. Sears is active.

Publication


Featured researches published by Rosalie C. Sears.


Nature Cell Biology | 2004

A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells

Elizabeth S. Yeh; Melissa Cunningham; Hugh Arnold; Dawn Chasse; Teresa Monteith; Giovanni Ivaldi; William C. Hahn; P. Todd Stukenberg; Shirish Shenolikar; Takafumi Uchida; Christopher M. Counter; Joseph R. Nevins; Anthony R. Means; Rosalie C. Sears

The stability of c-Myc is regulated by multiple Ras effector pathways. Phosphorylation at Ser 62 stabilizes c-Myc, whereas subsequent phosphorylation at Thr 58 is required for its degradation. Here we show that Ser 62 is dephosphorylated by protein phosphatase 2A (PP2A) before ubiquitination of c-Myc, and that PP2A activity is regulated by the Pin1 prolyl isomerase. Furthermore, the absence of Pin1 or inhibition of PP2A stabilizes c-Myc. A stable c-MycT58A mutant that cannot bind Pin1 or be dephosphorylated by PP2A replaces SV40 small T antigen in human cell transformation and tumorigenesis assays. Therefore, small T antigen, which inactivates PP2A, exerts its oncogenic potential by preventing dephosphorylation of c-Myc, resulting in c-Myc stabilization. Thus, Ras-dependent signalling cascades ensure transient and self-limiting accumulation of c-Myc, disruption of which contributes to human cell oncogenesis.


Journal of Experimental Medicine | 2007

FBW7 mutations in leukemic cells mediate NOTCH pathway activation and resistance to γ-secretase inhibitors

Jennifer O'Neil; Jonathan E. Grim; Peter Strack; Sudhir Rao; Deanne Tibbitts; Christopher Winter; James S. Hardwick; Markus Welcker; Jules P.P. Meijerink; Rob Pieters; Giulio Draetta; Rosalie C. Sears; Bruce E. Clurman; A. Thomas Look

γ-secretase inhibitors (GSIs) can block NOTCH receptor signaling in vitro and therefore offer an attractive targeted therapy for tumors dependent on deregulated NOTCH activity. To clarify the basis for GSI resistance in T cell acute lymphoblastic leukemia (T-ALL), we studied T-ALL cell lines with constitutive expression of the NOTCH intracellular domain (NICD), but that lacked C-terminal truncating mutations in NOTCH1. Each of the seven cell lines examined and 7 of 81 (8.6%) primary T-ALL samples harbored either a mutation or homozygous deletion of the gene FBW7, a ubiquitin ligase implicated in NICD turnover. Indeed, we show that FBW7 mutants cannot bind to the NICD and define the phosphodegron region of the NICD required for FBW7 binding. Although the mutant forms of FBW7 were still able to bind to MYC, they do not target it for degradation, suggesting that stabilization of both NICD and its principle downstream target, MYC, may contribute to transformation in leukemias with FBW7 mutations. In addition, we show that all seven leukemic cell lines with FBW7 mutations were resistant to the MRK-003 GSI. Most of these resistant lines also failed to down-regulate the mRNA levels of the NOTCH targets MYC and DELTEX1 after treatment with MRK-003, implying that residual NOTCH signaling in T-ALLs with FBW7 mutations contributes to GSI resistance.


Cell | 2007

CIP2A Inhibits PP2A in Human Malignancies

Melissa R. Junttila; Pietri Puustinen; Minna Niemelä; Raija Ahola; Hugh Arnold; Trine Bøttzauw; Risto Ala-aho; Christina Nielsen; Johanna Ivaska; Yoichi Taya; Shi-Long Lu; Shujun Lin; Edward K. L. Chan; Xiao-Jing Wang; Reidar Grénman; Juergen Kast; Tuula Kallunki; Rosalie C. Sears; Veli-Matti Kähäri; Jukka Westermarck

Inhibition of protein phosphatase 2A (PP2A) activity has been identified as a prerequisite for the transformation of human cells. However, the molecular mechanisms by which PP2A activity is inhibited in human cancers are currently unclear. In this study, we describe a cellular inhibitor of PP2A with oncogenic activity. The protein, designated Cancerous Inhibitor of PP2A (CIP2A), interacts directly with the oncogenic transcription factor c-Myc, inhibits PP2A activity toward c-Myc serine 62 (S62), and thereby prevents c-Myc proteolytic degradation. In addition to its function in c-Myc stabilization, CIP2A promotes anchorage-independent cell growth and in vivo tumor formation. The oncogenic activity of CIP2A is demonstrated by transformation of human cells by overexpression of CIP2A. Importantly, CIP2A is overexpressed in two common human malignancies, head and neck squamous cell carcinoma (HNSCC) and colon cancer. Thus, our data show that CIP2A is a human oncoprotein that inhibits PP2A and stabilizes c-Myc in human malignancies.


Cell Cycle | 2004

The Life Cycle of C-Myc From Synthesis to Degradation

Rosalie C. Sears

The c-Myc transcription factor is a potent regulator of cellular proliferation and cell fate decision. Precise regulation of c-Myc protein levels is essential to maintain normal cell function. In order to maintain proper levels of c-Myc, its protein stability is tightly controlled. c-Myc is degraded through the ubiquitin-proteasome pathway. This perspective discusses a sophisticated and complex signaling pathway that controls the life cycle of c-Myc from protein synthesis to ubiquitin-mediated degradation. The pathway involves Ras-activated kinases, the Pin1 prolyl isomerase, the PP2A phosphatase and a series of c-Myc phosphorylation and dephosphorylation events that control its stability.


Molecular and Cellular Biology | 2006

Protein phosphatase 2A regulatory subunit B56α associates with c-Myc and negatively regulates c-Myc accumulation

Hugh Arnold; Rosalie C. Sears

ABSTRACT Protein phosphatase 2A (PP2A) plays a prominent role in controlling accumulation of the proto-oncoprotein c-Myc. PP2A mediates its effects on c-Myc by dephosphorylating a conserved residue that normally stabilizes c-Myc, and in this way, PP2A enhances c-Myc ubiquitin-mediated degradation. Stringent regulation of c-Myc levels is essential for normal cell function, as c-Myc overexpression can lead to cell transformation. Conversely, PP2A has tumor suppressor activity. Uncovering relevant PP2A holoenzymes for a particular target has been limited by the fact that cellular PP2A represents a large heterogeneous population of trimeric holoenzymes, composed of a conserved catalytic subunit and a structural subunit along with a variable regulatory subunit which directs the holoenzyme to a specific target. We now report the identification of a specific PP2A regulatory subunit, B56α, that selectively associates with the N terminus of c-Myc. B56α directs intact PP2A holoenzymes to c-Myc, resulting in a dramatic reduction in c-Myc levels. Inhibition of PP2A-B56α holoenzymes, using small hairpin RNA to knock down B56α, results in c-Myc overexpression, elevated levels of c-Myc serine 62 phosphorylation, and increased c-Myc function. These results uncover a new protein involved in regulating c-Myc expression and reveal a critical interconnection between a potent oncoprotein, c-Myc, and a well-documented tumor suppressor, PP2A.


The EMBO Journal | 2007

Inhibition of c-Myc activity by ribosomal protein L11.

Mu Shui Dai; Hugh Arnold; Xiao Xin Sun; Rosalie C. Sears; Hua Lu

The c‐Myc oncoprotein promotes cell growth by enhancing ribosomal biogenesis through upregulation of RNA polymerases I‐, II‐, and III‐dependent transcription. Overexpression of c‐Myc and aberrant ribosomal biogenesis leads to deregulated cell growth and tumorigenesis. Hence, c‐Myc activity and ribosomal biogenesis must be regulated in cells. Here, we show that ribosomal protein L11, a component of the large subunit of the ribosome, controls c‐Myc function through a negative feedback mechanism. L11 is transcriptionally induced by c‐Myc, and overexpression of L11 inhibits c‐Myc‐induced transcription and cell proliferation. Conversely, reduction of endogenous L11 by siRNA increases these c‐Myc activities. Mechanistically, L11 binds to the Myc box II (MB II), inhibits the recruitment of the coactivator TRRAP, and reduces histone H4 acetylation at c‐Myc target gene promoters. In response to serum stimulation or serum starvation, L11 and TRRAP display inverse promoter‐binding profiles. In addition, L11 regulates c‐Myc levels. These results identify L11 as a feedback inhibitor of c‐Myc and suggest a novel role for L11 in regulating c‐Myc‐enhanced ribosomal biogenesis.


The FASEB Journal | 2009

Direct interaction between the inhibitor 2 and ceramide via sphingolipid-protein binding is involved in the regulation of protein phosphatase 2A activity and signaling

Archana Mukhopadhyay; Sahar A. Saddoughi; Pengfei Song; Iyad Sultan; Suriyan Ponnusamy; Can E. Senkal; Christopher F. Snook; Hugh Arnold; Rosalie C. Sears; Yusuf A. Hannun; Besim Ogretmen

In this study, the inhibitor 2 of protein phosphatase 2A (I2PP2A) was identified in vitro and in situ as a ceramide‐binding protein, which exhibits stereoisomer specificity and fatty acid chain length preference. Site‐directed mutagenesis coupled with structural details of I2PP2A suggested that VIK 207‐209 residues localized on helix 7 are important for ceramide binding and single mutation of K209D altered this interaction. Notably, I2PP2A‐ceramide binding decreased the association between PP2A and the inhibitor, preventing the inhibition of PP2A activity in vitro. In addition, studies in A549 human lung cancer cells revealed that ceramide mediates c‐Myc degradation via its PP2A‐dependent dephosphorylation at S62, and treatment with okadaic acid and expression of c‐Myc mutants with S62A or S62D conversions resulted in resistance to ceramidemediated degradation. Importantly, whereas down‐regulation of I2PP2A enhanced PP2A‐mediated c‐Myc degradation in response to ceramide, ectopic expression of wild‐type I2PP2A but not of its K209D mutant protected this degradation in A549 cells. Moreover, expression of wild‐type I2PP2A prevented the growth‐inhibitory effects of ceramide both against A549 cells and xenograft‐driven tumors in situ and in vivo compared with that in controls. Thus, these results suggest that direct interaction of I2PP2A with ceramide plays important biological roles via the regulation of PP2A activity and signaling, which in turn control ceramide‐mediated degradation of c‐Myc and antiproliferation.— Mukhopadhyay, A., Saddoughi, S. A., Song, P., Sultan, I., Ponnusamy, S., Senkal, C. E., Snook, C. F., Arnold, H. K., Sears, R. C., Hannun, Y. A., Ogretmen, B. Direct interaction between the inhibitor 2 and ceramide via sphingolipid‐protein binding is involved in the regulation of protein phosphatase 2A activity and signaling. FASEB J. 23, 751–763 (2009)


Developmental Cell | 2010

Focal Adhesion Kinase Is Required for Intestinal Regeneration and Tumorigenesis Downstream of Wnt/c-Myc Signaling

Gabrielle H. Ashton; Jennifer P. Morton; Kevin Myant; Toby J. Phesse; Rachel A. Ridgway; Victoria Marsh; Julie A. Wilkins; Dimitris Athineos; Vanesa Muncan; Richard Kemp; Kristi L. Neufeld; Hans Clevers; Valerie G. Brunton; Douglas J. Winton; Xiaoyan Wang; Rosalie C. Sears; Alan Richard Clarke; Margaret C. Frame; Owen J. Sansom

The intestinal epithelium has a remarkable capacity to regenerate after injury and DNA damage. Here, we show that the integrin effector protein Focal Adhesion Kinase (FAK) is dispensable for normal intestinal homeostasis and DNA damage signaling, but is essential for intestinal regeneration following DNA damage. Given Wnt/c-Myc signaling is activated following intestinal regeneration, we investigated the functional importance of FAK following deletion of the Apc tumor suppressor protein within the intestinal epithelium. Following Apc loss, FAK expression increased in a c-Myc-dependent manner. Codeletion of Apc and Fak strongly reduced proliferation normally induced following Apc loss, and this was associated with reduced levels of phospho-Akt and suppression of intestinal tumorigenesis in Apc heterozygous mice. Thus, FAK is required downstream of Wnt Signaling, for Akt/mTOR activation, intestinal regeneration, and tumorigenesis. Importantly, this work suggests that FAK inhibitors may suppress tumorigenesis in patients at high risk of developing colorectal cancer.


Cancer Research | 2011

Phosphorylation Regulates c-Myc's Oncogenic Activity in the Mammary Gland

Xiaoyan Wang; Melissa Cunningham; Xiaoli Zhang; Sara Tokarz; Bryan Laraway; Megan L. Troxell; Rosalie C. Sears

Expression of the c-Myc oncoprotein is affected by conserved threonine 58 (T58) and serine 62 (S62) phosphorylation sites that help to regulate c-Myc protein stability, and altered ratios of T58 and S62 phosphorylation have been observed in human cancer. Here, we report the development of 3 unique c-myc knock-in mice that conditionally express either c-Myc(WT) or the c-Myc(T58A) or c-Myc(S62A) phosphorylation mutant from the constitutively active ROSA26 locus in response to Cre recombinase to study the role of these phosphorylation sites in vivo. Using a mammary-specific Cre model, we found that expression of c-Myc(WT) resulted in increased mammary gland density, but normal morphology and no tumors at the level expressed from the ROSA promoter. In contrast, c-Myc(T58A) expression yielded enhanced mammary gland density, hyperplastic foci, cellular dysplasia, and mammary carcinoma, associated with increased genomic instability and suppressed apoptosis relative to c-Myc(WT). Alternatively, c-Myc(S62A) expression reduced mammary gland density relative to control glands, and this was associated with increased genomic instability and normal apoptotic function. Our results indicate that specific activities of c-Myc are differentially affected by T58 and S62 phosphorylation. This model provides a robust platform to interrogate the role that these phosphorylation sites play in c-Myc function during development and tumorigenesis.


The EMBO Journal | 2009

The Axin1 scaffold protein promotes formation of a degradation complex for c‐Myc

Hugh Arnold; Xiaoli Zhang; Colin J. Daniel; Deanne Tibbitts; Julie Escamilla-Powers; Amy S. Farrell; Sara A. Tokarz; Charlie Morgan; Rosalie C. Sears

Expression of the c‐Myc proto‐oncoprotein is tightly regulated in normal cells. Phosphorylation at two conserved residues, threonine58 (T58) and serine62 (S62), regulates c‐Myc protein stability. In cancer cells, c‐Myc can become aberrantly stabilized associated with altered T58 and S62 phosphorylation. A complex signalling cascade involving GSK3β kinase, the Pin1 prolyl isomerase, and the PP2A‐B56α phosphatase controls phosphorylation at these sites. We report here a novel role for the tumour suppressor scaffold protein Axin1 in facilitating the formation of a degradation complex for c‐Myc containing GSK3β, Pin1, and PP2A‐B56α. Although knockdown of Axin1 decreases the association of c‐Myc with these proteins, reduces T58 and enhances S62 phosphorylation, and increases c‐Myc stability, acute expression of Axin1 reduces c‐Myc levels and suppresses c‐Myc transcriptional activity. Moreover, the regulation of c‐Myc by Axin1 is impaired in several tested cancer cell lines with known stabilization of c‐Myc or loss of Axin1. This study provides critical insight into the regulation of c‐Myc expression, how this can be disrupted in three cancer types, and adds to our knowledge of the tumour suppressor activity of Axin1.

Collaboration


Dive into the Rosalie C. Sears'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge