Network


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

Hotspot


Dive into the research topics where Dipankar Ray is active.

Publication


Featured researches published by Dipankar Ray.


Molecular and Cellular Biology | 2001

The cell cycle-regulatory CDC25A phosphatase inhibits apoptosis signal-regulating kinase 1.

Xianghong Zou; Tateki Tsutsui; Dipankar Ray; James F. Blomquist; Hidenori Ichijo; David S. Ucker; Hiroaki Kiyokawa

ABSTRACT CDC25A phosphatase promotes cell cycle progression by activating G1 cyclin-dependent kinases and has been postulated to be an oncogene because of its ability to cooperate with RAS to transform rodent fibroblasts. In this study, we have identified apoptosis signal-regulating kinase 1 (ASK1) as a CDC25A-interacting protein by yeast two-hybrid screening. ASK1 activates the p38 mitogen-activated protein kinase (MAPK) and c-Jun NH2-terminal protein kinase–stress-activated protein kinase (JNK/SAPK) pathways upon various cellular stresses. Coimmunoprecipitation studies demonstrated that CDC25A physically associates with ASK1 in mammalian cells, and immunocytochemistry with confocal laser-scanning microscopy showed that these two proteins colocalize in the cytoplasm. The carboxyl terminus of CDC25A binds to a domain of ASK1 adjacent to its kinase domain and inhibits the kinase activity of ASK1, independent of and without effect on the phosphatase activity of CDC25A. This inhibitory action of CDC25A on ASK1 activity involves diminished homo-oligomerization of ASK1. Increased cellular expression of wild-type or phosphatase-inactive CDC25A from inducible transgenes suppresses oxidant-dependent activation of ASK1, p38, and JNK1 and reduces specific sensitivity to cell death triggered by oxidative stress, but not other apoptotic stimuli. Thus, increased expression of CDC25A, frequently observed in human cancers, could contribute to reduced cellular responsiveness to oxidative stress under mitogenic or oncogenic conditions, while it promotes cell cycle progression. These observations propose a mechanism of oncogenic transformation by the dual function of CDC25A on cell cycle progression and stress responses.


Cancer Research | 2007

Hemizygous Disruption of Cdc25A Inhibits Cellular Transformation and Mammary Tumorigenesis in Mice

Dipankar Ray; Yasuhisa Terao; Dipali Nimbalkar; Hiroyuki Hirai; Evan C. Osmundson; Xianghong Zou; Roberta Franks; Konstantin Christov; Hiroaki Kiyokawa

CDC25A phosphatase activates multiple cyclin-dependent kinases (CDK) during cell cycle progression. Inactivation of CDC25A by ubiquitin-mediated degradation is a major mechanism of DNA damage-induced S-G(2) checkpoint. Although increased CDC25A expression has been reported in various human cancer tissues, it remains unclear whether CDC25A activation is a critical rate-limiting step of carcinogenesis. To assess the role for CDC25A in cell cycle control and carcinogenesis, we used a Cdc25A-null mouse strain we recently generated. Whereas Cdc25A(-/-) mice exhibit early embryonic lethality, Cdc25A(+/-) mice show no appreciable developmental defect. Cdc25A(+/-) mouse embryonic fibroblasts (MEF) exhibit normal kinetics of cell cycle progression at early passages, modestly enhanced G(2) checkpoint response to DNA damage, and shortened proliferative life span, compared with wild-type MEFs. Importantly, Cdc25A(+/-) MEFs are significantly resistant to malignant transformation induced by coexpression of H-ras(V12) and a dominant negative p53 mutant. The rate-limiting role for CDC25A in transformation is further supported by decreased transformation efficiency in MCF-10A human mammary epithelial cells stably expressing CDC25A small interfering RNA. Consistently, Cdc25A(+/-) mice show substantially prolonged latency in mammary tumorigenesis induced by MMTV-H-ras or MMTV-neu transgene, whereas MMTV-myc-induced tumorigenesis is not significantly affected by Cdc25A heterozygosity. Mammary tissues of Cdc25A(+/-);MMTV-neu mice before tumor development display less proliferative response to the oncogene with increased tyrosine phosphorylation of CDK1/2, but show no significant change in apoptosis. These results suggest that Cdc25A plays a rate-limiting role in transformation and tumor initiation mediated by ras activation.


Cancer Research | 2007

Deregulated CDC25A Expression Promotes Mammary Tumorigenesis with Genomic Instability

Dipankar Ray; Yasuhisa Terao; Peter G. Fuhrken; Zhi Qing Ma; Francesco J. DeMayo; Konstantin Christov; Nyla A. Heerema; Roberta Franks; Sophia Y. Tsai; Eleftherios T. Papoutsakis; Hiroaki Kiyokawa

Checkpoint pathways help cells maintain genomic integrity, delaying cell cycle progression in response to various risks of fidelity, such as genotoxic stresses, compromised DNA replication, and impaired spindle control. Cancer cells frequently exhibit genomic instability, and recent studies showed that checkpoint pathways are likely to serve as a tumor-suppressive barrier in vivo. The cell cycle-promoting phosphatase CDC25A is an activator of cyclin-dependent kinases and one of the downstream targets for the CHK1-mediated checkpoint pathway. Whereas CDC25A overexpression is observed in various human cancer tissues, it has not been determined whether deregulated CDC25A expression triggers or promotes tumorigenesis in vivo. Here, we show that transgenic expression of CDC25A cooperates markedly with oncogenic ras or neu in murine mammary tumorigenesis. MMTV-CDC25A transgenic mice exhibit alveolar hyperplasia in the mammary tissue but do not develop spontaneous mammary tumors. The MMTV-CDC25A transgene markedly shortens latency of tumorigenesis in MMTV-ras mice. The MMTV-CDC25A transgene also accelerates tumor growth in MMTV-neu mice with apparent cell cycle miscoordination. CDC25A-overexpressing tumors, which invade more aggressively, exhibit various chromosomal aberrations on fragile regions, including the mouse counterpart of human 1p31-36, according to array-based comparative genomic hybridization and karyotyping. The chromosomal aberrations account for substantial changes in gene expression profile rendered by transgenic expression of CDC25A, including down-regulation of Trp73. These data indicate that deregulated control of cellular CDC25A levels leads to in vivo genomic instability, which cooperates with the neu-ras oncogenic pathway in mammary tumorigenesis.


Cancer Research | 2008

CDC25A phosphatase: A rate-limiting oncogene that determines genomic stability

Dipankar Ray; Hiroaki Kiyokawa

CDC25A is a critical regulator of cell cycle progression and checkpoint response. Overexpression of this cyclin-dependent kinase phosphatase occurs often in human cancers. Our recent genetic studies in the mouse indicate that restricting CDC25A can limit tumorigenesis induced by the HER2/neu-RAS oncogenic pathway without compromising normal cell division or viability. These findings offer a sound foundation to justify development of CDC25A inhibitors for antitumor therapy.


Anti-cancer Agents in Medicinal Chemistry | 2008

In Vivo Roles of CDC25 Phosphatases: Biological Insight into the Anti-Cancer Therapeutic Targets

Hiroaki Kiyokawa; Dipankar Ray

CDC25 phosphatases are not only rate-limiting activators of cyclin-dependent kinases (CDKs) but also important targets of the CHK1/CHK2-mediated checkpoint pathway. Each isoform of the mammalian CDC25 family seems to exert unique biological functions. CDC25A is a critical regulator for both G1-S and G2-M transitions and essential for embryonic cell proliferation after the blastocyst stage. CDC25B is dispensable for embryogenesis but required for meiotic progression of oocytes in a manner analogous to Drosophila Twine or C. elegans cdc-25.1. Moreover, CDC25A and CDC25B appear to regulate different events or stages of mitosis. CDC25B may mediate the activation of CDK1/Cyclin B at the centrosome during prophase, while CDC25A may be required for the subsequent full activation of nuclear CDK1/Cyclin B. CDC25C is dispensable for both mitotic and meiotic divisions, although it is highly regulated during the processes. Excessive levels of CDC25A and CDC25B are often observed in various human cancer tissues. Deregulated expression of these phosphatases allows cells to overcome DNA damage-induced checkpoint, leading to genomic instability. Studies using mouse models demonstrated that deregulated expression of CDC25A significantly promotes RAS- or NEU-induced mammary tumor development with chromosomal aberrations, whereas decreased CDC25A expression in heterozygous knockout mice delays tumorigenesis. These biological properties of CDC25 phosphatases provide significant insight into the pathobiology of cancer and scientific foundation for anti-CDC25 therapeutic intervention.


Journal of Cell Biology | 2008

The HECT E3 ligase Smurf2 is required for Mad2-dependent spindle assembly checkpoint

Evan C. Osmundson; Dipankar Ray; Finola E. Moore; Qingshen Gao; Gerald H. Thomsen; Hiroaki Kiyokawa

Activation of the anaphase-promoting complex/cyclosome (APC/C) by Cdc20 is critical for the metaphase–anaphase transition. APC/C-Cdc20 is required for polyubiquitination and degradation of securin and cyclin B at anaphase onset. The spindle assembly checkpoint delays APC/C-Cdc20 activation until all kinetochores attach to mitotic spindles. In this study, we demonstrate that a HECT (homologous to the E6-AP carboxyl terminus) ubiquitin ligase, Smurf2, is required for the spindle checkpoint. Smurf2 localizes to the centrosome, mitotic midbody, and centromeres. Smurf2 depletion or the expression of a catalytically inactive Smurf2 results in misaligned and lagging chromosomes, premature anaphase onset, and defective cytokinesis. Smurf2 inactivation prevents nocodazole-treated cells from accumulating cyclin B and securin and prometaphase arrest. The silencing of Cdc20 in Smurf2-depleted cells restores mitotic accumulation of cyclin B and securin. Smurf2 depletion results in enhanced polyubiquitination and degradation of Mad2, a critical checkpoint effector. Mad2 is mislocalized in Smurf2-depleted cells, suggesting that Smurf2 regulates the localization and stability of Mad2. These data indicate that Smurf2 is a novel mitotic regulator.


Cell Cycle | 2007

CDC25A levels determine the balance of proliferation and checkpoint response

Dipankar Ray; Hiroaki Kiyokawa

Current evidence suggests that CDC25A is not only a major regulator of both G1/S and G2/M transition during unperturbed cell cycle progression, but also a critical checkpoint mediator. While CDC25A is overexpressed in a variety of human cancers, a key question remainedunanswered whether such overexpression of this CDK-activating phosphatase was a mechanism or consequence of accelerated proliferation and other malignant phenotypes. Recent studies onthe tumor suppressive roles of checkpoint proteins suggest that overriding checkpoint response leads normal or pre-cancerous cells to genomic instability and cumulative malignant changes. Here we provide our views on the role of CDC25A in cancer development and genomic stability, discussing insights from our recent studies on Cdc25A knockout mice and MMTV-CDC25A transgenic mice.


Science Signaling | 2015

The kinase activity of the Ser/Thr kinase BUB1 promotes TGF-β signaling.

Shyam Nyati; Katrina Schinske-Sebolt; Sethuramasundaram Pitchiaya; Katerina Chekhovskiy; Areeb Chator; Nauman Chaudhry; Joseph Dosch; Marcian E. Van Dort; Sooryanarayana Varambally; Chandan Kumar-Sinha; Mukesh K. Nyati; Dipankar Ray; Nils G. Walter; Hongtao Yu; Brian D. Ross; Alnawaz Rehemtulla

A kinase that controls cell division also promotes the activity of the transforming growth factor–β pathway. Placing BUB1 in the TGF-β Pathway The transforming growth factor–β (TGF-β) pathway regulates cell proliferation and migration, processes involved in development, regeneration, and tumorigenesis. The kinase BUB1, which promotes proper chromosome alignment as cells prepare to divide, also regulates cell proliferation. Nyati et al. connected BUB1 to TGF-β signaling. They found that knocking down BUB1 impaired TGF-β–mediated proliferation of tumor cells—but not by acting at chromosomes. Instead, cytoplasmic BUB1 interacted with TGF-β receptor subunits at the cell surface, promoting the interaction between receptor subunits and between the receptor and downstream signaling proteins. Inhibiting the kinase activity of BUB1 suppressed TGF-β pathway activity in cells in culture and in xenografts. The findings suggest a possible point of crosstalk between the mitotic checkpoint and TGF-β signaling. Transforming growth factor–β (TGF-β) signaling regulates cell proliferation and differentiation, which contributes to development and disease. Upon binding TGF-β, the type I receptor (TGFBRI) binds TGFBRII, leading to the activation of the transcription factors SMAD2 and SMAD3. Using an RNA interference screen of the human kinome and a live-cell reporter for TGFBR activity, we identified the kinase BUB1 (budding uninhibited by benzimidazoles-1) as a key mediator of TGF-β signaling. BUB1 interacted with TGFBRI in the presence of TGF-β and promoted the heterodimerization of TGFBRI and TGFBRII. Additionally, BUB1 interacted with TGFBRII, suggesting the formation of a ternary complex. Knocking down BUB1 prevented the recruitment of SMAD3 to the receptor complex, the phosphorylation of SMAD2 and SMAD3 and their interaction with SMAD4, SMAD-dependent transcription, and TGF-β–mediated changes in cellular phenotype including epithelial-mesenchymal transition (EMT), migration, and invasion. Knockdown of BUB1 also impaired noncanonical TGF-β signaling mediated by the kinases AKT and p38 MAPK (mitogen-activated protein kinase). The ability of BUB1 to promote TGF-β signaling depended on the kinase activity of BUB1. A small-molecule inhibitor of the kinase activity of BUB1 (2OH-BNPP1) and a kinase-deficient mutant of BUB1 suppressed TGF-β signaling and formation of the ternary complex in various normal and cancer cell lines. 2OH-BNPP1 administration to mice bearing lung carcinoma xenografts reduced the amount of phosphorylated SMAD2 in tumor tissue. These findings indicated that BUB1 functions as a kinase in the TGF-β pathway in a role beyond its established function in cell cycle regulation and chromosome cohesion.


Clinical Cancer Research | 2011

Molecular Imaging of TGFβ-Induced Smad2/3 Phosphorylation Reveals a Role for Receptor Tyrosine Kinases in Modulating TGFβ Signaling

Shyam Nyati; Katrina A. Schinske; Dipankar Ray; Mukesh K. Nyati; Brian D. Ross; Alnawaz Rehemtulla

Purpose: The dual modality of TGFβ, both as a potent tumor suppressor and a stimulator of tumor progression, invasion, and metastasis, make it a critical target for therapeutic intervention in human cancers. The ability to carry out real-time, noninvasive imaging of TGFβ-activated Smad signaling in live cells and animal models would significantly improve our understanding of the regulation of this unique signaling cascade. To advance these efforts, we developed a highly sensitive molecular imaging tool that repetitively, noninvasively, and dynamically reports on TGFBR1 kinase activity. Experimental Design: The bioluminescent TGFβR1 reporter construct was developed using a split firefly luciferase gene containing a functional sensor of Smad2 phosphorylation, wherein inhibition of TGFβ receptor1 kinase activity leads to an increase in reporter signaling. The reporter was stably transfected into mammalian cells and used to image in vivo and in vitro bioluminescent activity as a surrogate for monitoring TGFBR1 kinase activity. Results: The reporter was successfully used to monitor direct and indirect inhibition of TGFβ-induced Smad2 and SMAD3 phosphorylation in live cells and tumor xenografts and adapted for high-throughput screening, to identify a role for receptor tyrosine kinase inhibitors as modulators of TGFβ signaling. Conclusion: The reporter is a dynamic, noninvasive imaging modality for monitoring TGFβ-induced Smad2 signaling in live cells and tumor xenografts. It has immense potential for identifying novel effectors of R-Smad phosphorylation, for validating drug–target interaction, and for studying TGFβ signaling in different metastasis models. Clin Cancer Res; 17(23); 7424–39. ©2011 AACR.


Neoplasia | 2014

KRAS protein stability is regulated through SMURF2: UBCH5 complex-mediated β-TrCP1 degradation.

Shirish Shukla; Uday Sankar Allam; Aarif Ahsan; Guoan Chen; Pranathi Meda Krishnamurthy; Katherine Marsh; Matthew Rumschlag; Sunita Shankar; Christopher Whitehead; Matthew Schipper; Venkatesha Basrur; Daniel R. Southworth; Arul M. Chinnaiyan; Alnawaz Rehemtulla; David G. Beer; Theodore S. Lawrence; Mukesh K. Nyati; Dipankar Ray

Attempts to target mutant KRAS have been unsuccessful. Here, we report the identification of Smad ubiquitination regulatory factor 2 (SMURF2) and UBCH5 as a critical E3:E2 complex maintaining KRAS protein stability. Loss of SMURF2 either by small interfering RNA/short hairpin RNA (siRNA/shRNA) or by overexpression of a catalytically inactive mutant causes KRAS degradation, whereas overexpression of wild-type SMURF2 enhances KRAS stability. Importantly, mutant KRAS is more susceptible to SMURF2 loss where protein half-life decreases from >12 hours in control siRNA-treated cells to <3 hours on Smurf2 silencing, whereas only marginal differences were noted for wild-type protein. This loss of mutant KRAS could be rescued by overexpressing a siRNA-resistant wild-type SMURF2. Our data further show that SMURF2 monoubiquitinates UBCH5 at lysine 144 to form an active complex required for efficient degradation of a RAS-family E3, β-transducing repeat containing protein 1 (β-TrCP1). Conversely, β-TrCP1 is accumulated on SMURF2 loss, leading to increased KRAS degradation. Therefore, as expected, β-TrCP1 knockdown following Smurf2 siRNA treatment rescues mutant KRAS loss. Further, we identify two conserved proline (P) residues in UBCH5 critical for SMURF2 interaction; mutation of either of these P to alanine also destabilizes KRAS. As a proof of principle, we demonstrate that Smurf2 silencing reduces the clonogenic survival in vitro and prolongs tumor latency in vivo in cancer cells including mutant KRAS-driven tumors. Taken together, we show that SMURF2:UBCH5 complex is critical in maintaining KRAS protein stability and propose that targeting such complex may be a unique strategy to degrade mutant KRAS to kill cancer cells.

Collaboration


Dive into the Dipankar Ray's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aarif Ahsan

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shyam Nyati

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge