Network


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

Hotspot


Dive into the research topics where Manav Korpal is active.

Publication


Featured researches published by Manav Korpal.


Journal of Biological Chemistry | 2008

The miR-200 Family Inhibits Epithelial-Mesenchymal Transition and Cancer Cell Migration by Direct Targeting of E-cadherin Transcriptional Repressors ZEB1 and ZEB2

Manav Korpal; Esther S. Lee; Guohong Hu; Yibin Kang

MicroRNAs are small non-coding RNA molecules that can regulate gene expression by interacting with multiple mRNAs and inducing either translation suppression or degradation of mRNA. Recently, several miRNAs were identified as either promoters or suppressors of metastasis. However, it is unclear in which step(s) of the multistep metastatic cascade these miRNAs play a defined functional role. To study the functional importance of miRNAs in epithelial-mesenchymal transition (EMT), a process thought to initiate metastasis by enhancing the motility of tumor cells, we used a well established in vitro EMT assay: transforming growth factor-β-induced EMT in NMuMG murine mammary epithelial cells. We found that members of the miR-200 family, organized as two clusters in the genome, were repressed during EMT. Overexpression of each miRNA individually or as clusters in NMuMG cells hindered EMT by enhancing E-cadherin expression through direct targeting of ZEB1 and ZEB2, which encode transcriptional repressors of E-cadherin. In the 4TO7 mouse carcinoma cell line, which expresses low levels of endogenous E-cadherin and displays a mesenchymal phenotype, ectopic expression of the miR-200 family miRNAs significantly increased E-cadherin expression and altered cell morphology to an epithelial phenotype. Furthermore, ectopic expression of each miR-200 miRNA cluster significantly reduced the in vitro motility of 4TO7 cells in migration assays. These results suggested that loss of expression of the miR-200 family members may play a critical role in the repression of E-cadherin by ZEB1 and ZEB2 during EMT, thereby enhancing migration and invasion during cancer progression.


Nature Medicine | 2011

Direct targeting of Sec23a by miR-200s influences cancer cell secretome and promotes metastatic colonization

Manav Korpal; Brian Ell; Francesca M. Buffa; Toni Ibrahim; Mario Andres Blanco; Toni Celià-Terrassa; Laura Mercatali; Zia Khan; Hani Goodarzi; Yuling Hua; Yong Wei; Guohong Hu; Benjamin A. Garcia; Jiannis Ragoussis; Dino Amadori; Adrian L. Harris; Yibin Kang

Although the role of miR-200s in regulating E-cadherin expression and epithelial-to-mesenchymal transition is well established, their influence on metastatic colonization remains controversial. Here we have used clinical and experimental models of breast cancer metastasis to discover a pro-metastatic role of miR-200s that goes beyond their regulation of E-cadherin and epithelial phenotype. Overexpression of miR-200s is associated with increased risk of metastasis in breast cancer and promotes metastatic colonization in mouse models, phenotypes that cannot be recapitulated by E-cadherin expression alone. Genomic and proteomic analyses revealed global shifts in gene expression upon miR-200 overexpression toward that of highly metastatic cells. miR-200s promote metastatic colonization partly through direct targeting of Sec23a, which mediates secretion of metastasis-suppressive proteins, including Igfbp4 and Tinagl1, as validated by functional and clinical correlation studies. Overall, these findings suggest a pleiotropic role of miR-200s in promoting metastatic colonization by influencing E-cadherin–dependent epithelial traits and Sec23a-mediated tumor cell secretome.


Nature Medicine | 2009

Imaging transforming growth factor-Β signaling dynamics and therapeutic response in breast cancer bone metastasis

Manav Korpal; Jun Yan; Xin Lu; Shuwa Xu; Dorothy A. Lerit; Yibin Kang

Although the transforming growth factor-β (TGF-β) pathway has been implicated in breast cancer metastasis, its in vivo dynamics and temporal-spatial involvement in organ-specific metastasis have not been investigated. Here we engineered a xenograft model system with a conditional control of the TGF-β–SMAD signaling pathway and a dual-luciferase reporter system for tracing both metastatic burden and TGF-β signaling activity in vivo. Strong TGF-β signaling in osteolytic bone lesions is suppressed directly by genetic and pharmacological disruption of the TGF-β–SMAD pathway and indirectly by inhibition of osteoclast function with bisphosphonates. Notably, disruption of TGF-β signaling early in metastasis can substantially reduce metastasis burden but becomes less effective when bone lesions are well established. Our in vivo system for real-time manipulation and detection of TGF-β signaling provides a proof of principle for using similar strategies to analyze the in vivo dynamics of other metastasis-associated signaling pathways and will expedite the development and characterization of therapeutic agents.


Journal of Translational Medicine | 2011

MiRNA-205 modulates cellular invasion and migration via regulating zinc finger E-box binding homeobox 2 expression in esophageal squamous cell carcinoma cells

Kayoko Matsushima; Hajime Isomoto; Naoyuki Yamaguchi; Naoki Inoue; Haruhisa Machida; Toshiyuki Nakayama; Tomayoshi Hayashi; Masaki Kunizaki; Shigekazu Hidaka; Takeshi Nagayasu; Masahiro Nakashima; Kenta Ujifuku; Norisato Mitsutake; Akira Ohtsuru; Shunichi Yamashita; Manav Korpal; Yibin Kang; Philip A. Gregory; Gregory J. Goodall; Shigeru Kohno; Kazuhiko Nakao

BackgroundEsophageal squamous cell carcinoma (ESCC) is often diagnosed at later stages until they are incurable. MicroRNA (miR) is a small, non-coding RNA that negatively regulates gene expression mainly via translational repression. Accumulating evidence indicates that deregulation of miR is associated with human malignancies including ESCC. The aim of this study was to identify miR that could be specifically expressed and exert distinct biological actions in ESCC.MethodsTotal RNA was extracted from ESCC cell lines, OE21 and TE10, and a non-malignant human esophageal squamous cell line, Het-1A, and subjected to microarray analysis. Expression levels of miR that showed significant differences between the 2 ESCC and Het-1A cells based on the comprehensive analysis were analyzed by the quantitative reverse transcriptase (RT)-PCR method. Then, functional analyses, including cellular proliferation, apoptosis and Matrigel invasion and the wound healing assay, for the specific miR were conducted. Using ESCC tumor samples and paired surrounding non-cancerous tissue obtained endoscopically, the association with histopathological differentiation was examined with quantitative RT-PCR.ResultsBased on the miR microarray analysis, there were 14 miRs that showed significant differences (more than 2-fold) in expression between the 2 ESCC cells and non-malignant Het-1A. Among the significantly altered miRs, miR-205 expression levels were exclusively higher in 5 ESCC cell lines examined than any other types of malignant cell lines and Het-1A. Thus, miR-205 could be a specific miR in ESCC. Modulation of miR-205 expression by transfection with its precursor or anti-miR-205 inhibitor did not affect ESCC cell proliferation and apoptosis, but miR-205 was found to be involved in cell invasion and migration. Western blot revealed that knockdown of miR-205 expression in ESCC cells substantially enhanced expression of zinc finger E-box binding homeobox 2, accompanied by reduction of E-cadherin, a regulator of epithelial mesenchymal transition. The miR-205 expression levels were not associated with histological differentiation of human ESCC.ConclusionsThese results imply that miR-205 is an ESCC-specific miR that exerts tumor-suppressive activities with EMT inhibition by targeting ZEB2.


European Journal of Cancer | 2010

Targeting the transforming growth factor-β signalling pathway in metastatic cancer

Manav Korpal; Yibin Kang

Transforming growth factor (TGF)-beta signalling plays a dichotomous role in tumour progression, acting as a tumour suppressor early and as a pro-metastatic pathway in late-stages. There is accumulating evidence that advanced-stage tumours produce excessive levels of TGF-beta, which acts to promote tumour growth, invasion and colonisation of secondary organs. In light of the pro-metastasis function, many strategies are currently being explored to antagonise the TGF-beta pathway as a treatment for metastatic cancers. Strategies such as using large molecule ligand traps, reducing the translational efficiency of TGF-beta ligands using antisense technology, and antagonising TGF-beta receptor I/II kinase function using small molecule inhibitors are the most prominent methods being explored today. Administration of anti-TGF-beta therapies alone, or in combination with immunosuppressive or cytotoxic therapies, has yielded promising results in the preclinical and clinical settings. Despite these successes, the temporal- and context-dependent roles of TGF-beta signalling in cancer has made it challenging to define patient subgroups that are most likely to respond, and the therapeutic regimens that will be most effective in the clinic. Novel mouse models and diagnostic tools are being developed today to circumvent these issues, which may potentially expedite anti-TGF-beta drug development and clinical application.


Journal of Biological Chemistry | 2008

RanBP10 is a cytoplasmic guanine nucleotide exchange factor that modulates noncentrosomal microtubules.

Harald Schulze; Marei Dose; Manav Korpal; Imke Meyer; Joseph E. Italiano; Ramesh A. Shivdasani

Microtubule spindle assembly in mitosis is stimulated by Ran·GTP, which is generated along condensed chromosomes by the guanine nucleotide exchange factor (GEF) RCC1. This relationship suggests that similar activities might modulate other microtubule structures. Interphase microtubules usually extend from the centrosome, although noncentrosomal microtubules function in some differentiated cells, including megakaryocytes. In these cells, platelet biogenesis requires massive mobilization of microtubules in the cell periphery, where they form proplatelets, the immediate precursors of platelets, in the apparent absence of centrioles. Here we identify a cytoplasmic Ran-binding protein, RanBP10, as a factor that binds β-tubulin and associates with megakaryocyte microtubules. Unexpectedly, RanBP10 harbors GEF activity toward Ran. A point mutation in the candidate GEF domain abolishes exchange activity, and our results implicate RanBP10 as a localized cytoplasmic Ran-GEF. RNA interference-mediated loss of RanBP10 in cultured megakaryocytes disrupts microtubule organization. These results lead us to propose that spatiotemporally restricted generation of cytoplasmic Ran·GTP may influence organization of the specialized microtubules required in thrombopoiesis and that RanBP10 might serve as a molecular link between Ran and noncentrosomal microtubules.


Blood | 2006

Characterization of the megakaryocyte demarcation membrane system and its role in thrombopoiesis.

Harald Schulze; Manav Korpal; Jonathan B. Hurov; Sang We Kim; Jinghang Zhang; Lewis C. Cantley; Thomas Graf; Ramesh A. Shivdasani


Blood | 2004

Interactions between the megakaryocyte/platelet-specific β1 tubulin and the secretory leukocyte protease inhibitor SLPI suggest a role for regulated proteolysis in platelet functions

Harald Schulze; Manav Korpal; Wolfgang Bergmeier; Joseph E. Italiano; Sharon M. Wahl; Ramesh A. Shivdasani


Archive | 2010

regulated proteolysis in platelet functions and the secretory leukocyte protease inhibitor SLPI suggest a role for Interactions between the megakaryocyte/platelet-specific {beta}1 tubulin

Ramesh A. Shivdasani; Harald Schulze; Manav Korpal; Joseph E. Italiano; Sharon M. Wahl


Blood | 2006

Characterization of the megakaryocyte demarcation membrane system and its role in thrombopoiesis. Commentary

Andrew D. Leavitt; Harald Schuize; Manav Korpal; Jonathan B. Hurov; Sang-We Kim; Jinghang Zhang; Lewis C. Cantley; Thomas Graf; Ramesh A. Shivdasani

Collaboration


Dive into the Manav Korpal's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph E. Italiano

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jinghang Zhang

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jonathan B. Hurov

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Graf

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar

Sharon M. Wahl

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Guohong Hu

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge