Network


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

Hotspot


Dive into the research topics where Le Meng is active.

Publication


Featured researches published by Le Meng.


Oncogene | 2010

Heat-shock transcription factor HSF1 has a critical role in human epidermal growth factor receptor-2-induced cellular transformation and tumorigenesis

Le Meng; Vladimir L. Gabai; Michael Y. Sherman

The heat-shock transcription factor HSF1 was recently shown to have a key role in the development of tumors associated with activation of Ras or inactivation of p53. Here, we show that HSF1 is required for the cell transformation and tumorigenesis induced by the human epidermal growth factor receptor-2 (HER2) oncogene responsible for aggressive breast tumors. Upon expression of HER2, untransformed human mammary epithelial MCF-10A cells underwent neoplastic transformation, formed foci in culture and tumors in nude mouse xenografts. However, expression of HER2 in MCF-10A cells with knockdown of HSF1 did not cause either foci formation or tumor growth in xenografts. The antitumorigenic effect of downregulation of HSF1 was associated with HER2-induced accumulation of the cyclin-dependent kinase inhibitor p21 and decrease in the mitotic regulator survivin, which resulted in growth inhibition and cell senescence. In fact, either knockout of p21 or overexpression of survivin alleviated these effects of HSF1 knockdown. The proliferation of certain human HER2-positive breast cancer lines also requires HSF1, as its knockdown led to upregulation of p21 and/or decrease in survivin, precipitating growth arrest. Similar effects were observed with a small-molecular-weight inhibitor of the heat-shock response NZ28. The effects of HSF1 knockdown on the growth arrest and senescence of HER2-expressing cells were associated with downregulation of heat-shock protein (Hsp)72 and Hsp27. Therefore, HSF1 is critical for proliferation of HER2-expressing cells, most likely because it maintains the levels of HSPs, which in turn control regulators of senescence p21 and survivin.


Molecular and Cellular Biology | 2012

Heat Shock Transcription Factor Hsf1 Is Involved in Tumor Progression via Regulation of Hypoxia-Inducible Factor 1 and RNA-Binding Protein HuR

Vladimir L. Gabai; Le Meng; Geunwon Kim; Teresa A. Mills; Ivor J. Benjamin; Michael Y. Sherman

ABSTRACT Previously we demonstrated that the heat shock transcription factor Hsf1 is indispensable for transformation of mammary epithelial cells by the Her2 oncogene. Since Hsf1 affects oncogene-induced senescence (OIS), these findings suggest that Hsf1 affects tumor initiation when OIS plays a role. Indeed, here we report that Hsf1 knockout suppressed mammary hyperplasia in Her2-expressing mice and reduced tumor emergence. On the other hand, Hsf1 expression increases with advanced breast cancer, indicating that there is an additional role of Hsf1 in tumor progression. We studied rare tumors that developed in Hsf1-knockout mice and found that these tumors grew slower than tumors in control animals and showed suppressed angiogenesis. Similarly, in the xenograft model, knockdown of Hsf1 suppressed angiogenesis, which was associated with suppression of the HIF-1 pathway. Suppression of HIF-1 was at the level of translation due to downregulation of the RNA-binding protein HuR. Importantly, besides HIF-1, HuR controls translation of other major regulators of cancer progression, many of which were suppressed in Hsf1-knockdown cells. Therefore, in addition to OIS, Hsf1 regulates the HuR–HIF-1 pathway, thus affecting both cancer initiation and progression.


Oncogene | 2011

Heat shock protein Hsp72 plays an essential role in Her2-induced mammary tumorigenesis

Le Meng; Clayton R. Hunt; Julia A. Yaglom; Vladimir L. Gabai; Michael Y. Sherman

The major heat shock protein Hsp72 is expressed at elevated levels in many human cancers and its expression correlates with tumor progression. Here, we investigated the role of Hsp72 in Her2 oncogene-induced neoplastic transformation and tumorigenesis. Expression of Her2 in untransformed MCF10A mammary epithelial cells caused transformation, as judged by foci formation in culture and tumorigenesis in xenografts. However, expression of Her2 in Hsp72-depleted cells failed to induce transformation. The anti-tumorigenic effects of Hsp72 downregulation were associated with cellular senescence because of accumulation of p21 and depletion of survivin. Accordingly, either knockdown of p21 or expression of survivin reversed this senescence process. Further, we developed an animal model of Hsp72-dependent breast cancer associated with expression of Her2. Knockout (KO) of Hsp72 almost completely suppressed tumorigenesis in the MMTVneu breast cancer mouse model. In young Hsp72 KO mice, expression of Her2 instead of mammary tissue hyperplasia led to suppression of duct development and blocked alveolar budding. These effects were due to massive cell senescence in mammary tissue, which was associated with upregulation of p21 and downregulation of survivin. Therefore, Hsp72 has an essential role in Her2-induced tumorigenesis by regulating oncogene-induced senescence pathways.


Aging Cell | 2011

Oncogenes induce senescence with incomplete growth arrest and suppress the DNA damage response in immortalized cells

Michael Y. Sherman; Le Meng; Martha R. Stampfer; Vladimir L. Gabai; Julia A. Yaglom

Activation of the Her2 (ErbB2) oncogene is implicated in the development of breast, ovary and other cancers. Here, we show that expression of NeuT, a mutant‐activated rodent isoform of Her2, in immortalized breast epithelial cells, while promoting senescence‐associated morphological changes, up‐regulation of senescence‐associated β‐galactosidase activity, and accumulation of the cyclin‐dependent kinase inhibitor p21, failed to trigger the major senescence end‐point, i.e. permanent growth arrest. Similar senescence‐associated phenotype with incomplete growth arrest, which we dubbed senescence with incomplete growth arrest (SWING), could also be triggered by the expression of the Ras oncogene. SWING phenotype was stable, and persisted in tumor xenografts established from NeuT‐transduced cells. Furthermore, a significant population of cells in SWING state was found in tumors in the MMTV/NeuT transgenic mouse model. SWING cells showed downregulation of histone H2AX, critical for repair of double‐stranded DNA breaks, and impaired activation of Chk1 kinase. Overall, SWING cells were characterized by increased DNA instability and hypersensitivity to genotoxic stresses. We propose that the SWING state could be a stage in the process of cancer development.


Oncogene | 2014

ΔNp63α regulates Erk signaling via MKP3 to inhibit cancer metastasis.

Johann Bergholz; Yujun Zhang; Junfeng Wu; Le Meng; Erica M. Walsh; Arun Rai; Michael Y. Sherman; Zhi-Xiong Jim Xiao

Reduced expression of the p53 family member p63 has been suggested to play a causative role in cancer metastasis. Here, we show that ΔNp63α, the predominant p63 isoform, plays a major role in regulation of cell migration, invasion and cancer metastasis. We identified mitogen-activated protein (MAP) kinase phosphatase 3 (MKP3) as a downstream target of ΔNp63α that is required for mediating these effects. We show that ΔNp63α regulates extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) activity via MKP3 in both cancer and non-transformed cells. We further show that exogenous ΔNp63α inhibits cell invasion and is dependent on MKP3 upregulation for repression. Conversely, endogenous pan-p63 ablation results in increased cell migration and invasion, which can be reverted by reintroducing the ΔNp63α isoform alone, but not by other isoforms. Interestingly, these effects require Erk2, but not Erk1 expression, and can be rescued by enforced MKP3 expression. Moreover, MKP3 expression is reduced in invasive cancers, and reduced p63 expression increases metastatic frequency in vivo. Taken together, these results suggest an important role for ΔNp63α in preventing cancer metastasis by inhibition of Erk2 signaling via MKP3.


Cancer Research | 2008

Triggering Senescence Programs Suppresses Chk1 Kinase and Sensitizes Cells To Genotoxic Stresses

Vladimir L. Gabai; Cornelia O'Callaghan-Sunol; Le Meng; Michael Y. Sherman; Julia A. Yaglom

Depletion of the major heat shock protein Hsp72 leads to activation of the senescence program in a variety of tumor cell lines via both p53-dependent and p53-independent pathways. Here, we found that the Hsp72-depleted cells show defect in phosphorylation and activation of the protein kinase Chk1 by genotoxic stresses, such as UVC irradiation or camptothecin. Under these conditions, phosphorylation of Rad17 was also suppressed, whereas phosphorylation of p53 at Ser(15) was not affected, indicating a specific defect in phosphorylation of a subset of the ATR kinase substrates. Similarly, suppression of Chk1 activation was seen when senescence signaling was triggered by direct stimulation of p53, depletion of Cdc2, or overexpression of the cell cycle inhibitors p21 or p16. Thus, defect in Chk1 activation was not a consequence of the chaperone imbalance, but rather a downstream effect of activation of the senescence signaling. Inhibition of Chk1 was associated with inefficient inter-S phase checkpoint, as Hsp72 depleted cells failed to halt cell cycle progression upon UVC irradiation. Accordingly, sensitivity of cells to genotoxic stimuli after Hsp72 depletion was significantly enhanced. Thus, activation of the senescence signaling causes a defect in the DNA damage response manifested in increased sensitivity to genotoxic stresses.


Cancer Research | 2016

Anticancer Effects of Targeting Hsp70 in Tumor Stromal Cells.

Vladimir L. Gabai; Julia A. Yaglom; Yongmei Wang; Le Meng; Hao Shao; Geunwon Kim; Teresa Colvin; Jason E. Gestwicki; Michael Y. Sherman

The stress-induced chaperone protein Hsp70 enables the initiation and progression of many cancers, making it an appealing therapeutic target for development. Here, we show that cancer cells resistant to Hsp70 inhibitors in vitro remain sensitive to them in vivo, revealing the pathogenic significance of Hsp70 in tumor stromal cells rather than tumor cells as widely presumed. Using transgenic mouse models of cancer, we found that expression of Hsp70 in host stromal cells was essential to support tumor growth. Furthermore, genetic ablation or pharmacologic inhibition of Hsp70 suppressed tumor infiltration by macrophages needed to enable tumor growth. Overall, our results illustrate how Hsp70 inhibitors mediate the anticancer effects by targeting both tumor cells and tumor stromal cells, with implications for the broad use of these inhibitors as tools to ablate tumor-associated macrophages that enable malignant progression. Cancer Res; 76(20); 5926-32. ©2016 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Hsp70–Bag3 complex is a hub for proteotoxicity-induced signaling that controls protein aggregation

Anatoli B. Meriin; Arjun Narayanan; Le Meng; Ilya Alexandrov; Xaralabos Varelas; Ibrahim Cisse; Michael Y. Sherman

Significance This work dissects how cells monitor failure of proteasomes and trigger signaling responses defining whether cells survive proteotoxic stress or undergo apoptosis. The monitoring mechanism involves detection of a buildup of abnormal polypeptides released from ribosomes. Accordingly, the system simultaneously monitors effectiveness of several major processes, including protein synthesis, folding, and degradation. A special scaffold complex composed of heat shock protein 70 (Hsp70) and its cofactor Bcl-2–associated athanogene 3 (Bag3) links accumulation of abnormal polypeptide species with a number of protein kinases involved in various signal-transduction pathways. A startling finding is that an Hsp70–Bag3–regulated kinase, LATS1, regulates very early events of formation of protein aggregates; thus protein aggregation appears to be a tightly regulated process rather than the simple collapse of abnormal proteins. Protein abnormalities in cells are the cause of major pathologies, and a number of adaptive responses have evolved to relieve the toxicity of misfolded polypeptides. To trigger these responses, cells must detect the buildup of aberrant proteins which often associate with proteasome failure, but the sensing mechanism is poorly understood. Here we demonstrate that this mechanism involves the heat shock protein 70–Bcl-2–associated athanogene 3 (Hsp70–Bag3) complex, which upon proteasome suppression responds to the accumulation of defective ribosomal products, preferentially recognizing the stalled polypeptides. Components of the ribosome quality control system LTN1 and VCP and the ribosome-associated chaperone NAC are necessary for the interaction of these species with the Hsp70–Bag3 complex. This complex regulates important signaling pathways, including the Hippo pathway effectors LATS1/2 and the p38 and JNK stress kinases. Furthermore, under proteotoxic stress Hsp70–Bag3–LATS1/2 signaling regulates protein aggregation. We established that the regulated step was the emergence and growth of abnormal protein oligomers containing only a few molecules, indicating that aggregation is regulated at very early stages. The Hsp70–Bag3 complex therefore functions as an important signaling node that senses proteotoxicity and triggers multiple pathways that control cell physiology, including activation of protein aggregation.


Molecular and Cellular Biology | 2017

Correction: Heat shock transcription factor Hsf1 is involved in tumor progression via regulation of hypoxia-inducible factor 1 and RNA-binding protein HuR [Molecular and Cellular Biology, 32, 5 (2012) (929-940)] DOI: 10.1128/MCB.05921-11

Vladimir L. Gabai; Le Meng; Geunwon Kim; Teresa A. Mills; Ivor J. Benjamin; Michael Y. Sherman

Vladimir L. Gabai,a Le Meng,a Geunwon Kim,a Teresa A. Mills,a Ivor J. Benjamin,b and Michael Y. Shermana Department of Biochemistry, Boston University Medical School, Boston, Massachusetts, USA,a and Center for Cardiovascular Translational Biomedicine, Division of Cardiology, Department of Internal Medicine, Department of Biochemistry, University of Utah, School of Medicine, Salt Lake City, Utah, USAb


Molecular and Cellular Biology | 2017

Correction for Gabai et al., “Heat Shock Transcription Factor Hsf1 Is Involved in Tumor Progression via Regulation of Hypoxia-Inducible Factor 1 and RNA-Binding Protein HuR”

Vladimir L. Gabai; Le Meng; Geunwon Kim; Teresa A. Mills; Ivor J. Benjamin; Michael Y. Sherman

Collaboration


Dive into the Le Meng's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vladimir L. Gabai

Boston Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ivor J. Benjamin

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arjun Narayanan

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Clayton R. Hunt

Houston Methodist Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge