47 results on '"Y. Rebecca Chin"'
Search Results
2. Suppl. Figure 1 from Upregulation of AKT3 Confers Resistance to the AKT Inhibitor MK2206 in Breast Cancer
- Author
-
Y. Rebecca Chin, Tiffany Tsang, and Casey Stottrup
- Abstract
Upregulation of AKT3, but not AKT1 or AKT2, in AKT inhibitor-resistant MDA-MB-468 breast tumor cells.
- Published
- 2023
3. Supplementary Data from PTEN-Deficient Tumors Depend on AKT2 for Maintenance and Survival
- Author
-
Alex Toker, Steven P. Balk, Xin Yuan, and Y. Rebecca Chin
- Abstract
PDF file 1017K, Figure S1 shows the functions of Akt1 and Akt2 in the proliferation and migration of prostate tumor cells. Figure S2 demonstrates that all three Akt isoforms play a critical role in MDA-MB-468 breast cancer cell proliferation and formation of tumor spheroids. Figure S3 shows that Akt2 does not play a critical role in the survival of tumor cells with wild-type PTEN. Figure S4 shows the expression and phosphorylation levels of Akt1 and Akt2 in prostate tumor cells. Figure S5 demonstrates a critical role of p21 in Akt2-mediated cell cycle progression. Figure S6 shows that Bax does not play a role in Akt2-mediated spheroid survival. Figure S7 shows the volume of individual tumors measured in the xenograft studies for Fig. 6A
- Published
- 2023
4. Suppl. Figure 2 from Upregulation of AKT3 Confers Resistance to the AKT Inhibitor MK2206 in Breast Cancer
- Author
-
Y. Rebecca Chin, Tiffany Tsang, and Casey Stottrup
- Abstract
Knockdown of AKT1 or AKT2 does not sensitizes AKTi-resistant breast tumor cells to MK2206.
- Published
- 2023
5. Data from Upregulation of AKT3 Confers Resistance to the AKT Inhibitor MK2206 in Breast Cancer
- Author
-
Y. Rebecca Chin, Tiffany Tsang, and Casey Stottrup
- Abstract
Acquired resistance to molecular targeted therapy represents a major challenge for the effective treatment of cancer. Hyperactivation of the PI3K/AKT pathway is frequently observed in virtually all human malignancies, and numerous PI3K and AKT inhibitors are currently under clinical evaluation. However, mechanisms of acquired resistance to AKT inhibitors have yet to be described. Here, we use a breast cancer preclinical model to identify resistance mechanisms to a small molecule allosteric AKT inhibitor, MK2206. Using a step-wise and chronic high-dose exposure, breast cancer cell lines harboring oncogenic PI3K resistant to MK2206 were established. Using this model, we reveal that AKT3 expression is markedly upregulated in AKT inhibitor–resistant cells. Induction of AKT3 is regulated epigenetically by the bromodomain and extra terminal domain proteins. Importantly, knockdown of AKT3, but not AKT1 or AKT2, in resistant cells restores sensitivity to MK2206. AKT inhibitor–resistant cells also display an epithelial to mesenchymal transition phenotype as assessed by alterations in the levels of E-Cadherin, N-Cadherin, and vimentin, as well as enhanced invasiveness of tumor spheroids. Notably, the invasive morphology of resistant spheroids is diminished upon AKT3 depletion. We also show that resistance to MK2206 is reversible because upon drug removal resistant cells regain sensitivity to AKT inhibition, accompanied by reexpression of epithelial markers and reduction of AKT3 expression, implying that epigenetic reprogramming contributes to acquisition of resistance. These findings provide a rationale for developing therapeutics targeting AKT3 to circumvent acquired resistance in breast cancer. Mol Cancer Ther; 15(8); 1964–74. ©2016 AACR.
- Published
- 2023
6. Suppl. Figure 3 from Upregulation of AKT3 Confers Resistance to the AKT Inhibitor MK2206 in Breast Cancer
- Author
-
Y. Rebecca Chin, Tiffany Tsang, and Casey Stottrup
- Abstract
Knockdown of AKT3 sensitizes AKTi-resistant breast tumor cells to MK2206.
- Published
- 2023
7. Supplementary Methods, Figures S1 - S7 from PtdIns(3,4,5)P3-Dependent Activation of the mTORC2 Kinase Complex
- Author
-
Wenyi Wei, Bing Su, Alex Toker, Lewis C. Cantley, John Blenis, Bin Wang, Jinfang Zhang, Jianping Guo, Kohei Ogura, Y. Rebecca Chin, Wenjian Gan, and Pengda Liu
- Abstract
Figure S1. The Sin1-PH domain binds the mTOR kinase domain to inhibit mTOR kinase activity, Related to Figure 1. Figure S2. The PH domain is a unique and physiological functional PH domain, Related to Figure 2. Figure S3. PI(3,4,5)P3 binds the Sin1-PH motif to activate mTORC2, Related to Figure 3. Figure S4. The presence of PIP3 species leads to attenuated Sin1-PH domain interaction with mTOR-KD, Related to Figure 4. Figure S5. The Sin1-PH motif mainly interacts with PI(3,4,5)P3 through three critical residues including R393, K428 and K464, Related to Figure 5. Figure S6. The cancerous Sin1-PH mutant lose binding with the mTOR-kinase domain, leading to elevated Akt-S473 phosphorylation and enhanced oncogenic activities, Related to Figure 6. Figure S7. C-terminal tagging of KRas-CAAX to Sin1 could partially rescue the deficiency of the Sin1-CAA mutant in membrane recruitment and mTORC2 activation, Related to Figure 7.
- Published
- 2023
8. Suppl. Table 1 from Upregulation of AKT3 Confers Resistance to the AKT Inhibitor MK2206 in Breast Cancer
- Author
-
Y. Rebecca Chin, Tiffany Tsang, and Casey Stottrup
- Abstract
Table showing differentially expressed genes in T47D MK2206-resistant cells.
- Published
- 2023
9. High Throughput Confined Migration Microfluidic Device for Drug Screening (Small 16/2023)
- Author
-
Zihan Yang, Zhihang Zhou, Tongxu Si, Zhengdong Zhou, Li Zhou, Y. Rebecca Chin, Liang Zhang, Xinyuan Guan, and Mengsu Yang
- Subjects
Biomaterials ,General Materials Science ,General Chemistry ,Biotechnology - Published
- 2023
10. Supplementary Figure 1 from RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Laura E. Benjamin, George C. Prendergast, Alex Toker, Lawrence F. Brown, Olivier N. Kocher, James B. DuHadaway, Stephanie McNamara, Sharon Shechter, Arturo Bravo-Nuevo, Thuy L. Phung, Jacob Pourat, Lee Mangiante, Carole Perruzzi, Rebekah K. O'Donnell, Ningning Zheng, Durga Udayakumar, Y. Rebecca Chin, Minzhou Huang, Damien Gerald, and Shiva Kazerounian
- Abstract
PDF file - 219K, Effects of RhoB deletion on MT-myc induced mammary tumorigenesis in virgin female mice
- Published
- 2023
11. Supplementary Methods and Materials from RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Laura E. Benjamin, George C. Prendergast, Alex Toker, Lawrence F. Brown, Olivier N. Kocher, James B. DuHadaway, Stephanie McNamara, Sharon Shechter, Arturo Bravo-Nuevo, Thuy L. Phung, Jacob Pourat, Lee Mangiante, Carole Perruzzi, Rebekah K. O'Donnell, Ningning Zheng, Durga Udayakumar, Y. Rebecca Chin, Minzhou Huang, Damien Gerald, and Shiva Kazerounian
- Abstract
PDF file - 22K
- Published
- 2023
12. Supplementary Figure 3 from RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Laura E. Benjamin, George C. Prendergast, Alex Toker, Lawrence F. Brown, Olivier N. Kocher, James B. DuHadaway, Stephanie McNamara, Sharon Shechter, Arturo Bravo-Nuevo, Thuy L. Phung, Jacob Pourat, Lee Mangiante, Carole Perruzzi, Rebekah K. O'Donnell, Ningning Zheng, Durga Udayakumar, Y. Rebecca Chin, Minzhou Huang, Damien Gerald, and Shiva Kazerounian
- Abstract
PDF file - 54K, RhoB is downregulated in MCF7 cells by antisense oligonucleotides
- Published
- 2023
13. Supplementary Figure 2 from RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Laura E. Benjamin, George C. Prendergast, Alex Toker, Lawrence F. Brown, Olivier N. Kocher, James B. DuHadaway, Stephanie McNamara, Sharon Shechter, Arturo Bravo-Nuevo, Thuy L. Phung, Jacob Pourat, Lee Mangiante, Carole Perruzzi, Rebekah K. O'Donnell, Ningning Zheng, Durga Udayakumar, Y. Rebecca Chin, Minzhou Huang, Damien Gerald, and Shiva Kazerounian
- Abstract
PDF file - 21K, RhoB is downregulated in MDA-MB-231 cells using lentivirus shRNA
- Published
- 2023
14. Supplementary Information, Methods, Table 1, Figures 1-7 from 3-Phosphoinositide–Dependent Kinase 1 Potentiates Upstream Lesions on the Phosphatidylinositol 3-Kinase Pathway in Breast Carcinoma
- Author
-
Ramon Parsons, Hanina Hibshoosh, Vundavalli V.V.S. Murty, Jean J. Zhao, Gordon B. Mills, Alex Toker, Mary Beth Terry, Jorma Isola, Carlos Cordon-Cardo, Jennifer S. Yu, Tulio Matos, Albert Rojtman, Lorenzo Memeo, Xiaomei Wang, Mervi Laakso, Sofia K. Gruvberger-Saal, Jennifer S. Ferris, Da-In Kim, Y. Rebecca Chin, Yuli Xie, Bhaskar Dutta, Subhadra Nandula, Jiaping Wu, Christina R. Barkley, Benjamin D. Hopkins, Susan Koujak, Lao H. Saal, Tao Su, and Matthew Maurer
- Abstract
Supplementary Information, Methods, Table 1, Figures 1-7 from 3-Phosphoinositide–Dependent Kinase 1 Potentiates Upstream Lesions on the Phosphatidylinositol 3-Kinase Pathway in Breast Carcinoma
- Published
- 2023
15. Supplementary Figure 4 from RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Laura E. Benjamin, George C. Prendergast, Alex Toker, Lawrence F. Brown, Olivier N. Kocher, James B. DuHadaway, Stephanie McNamara, Sharon Shechter, Arturo Bravo-Nuevo, Thuy L. Phung, Jacob Pourat, Lee Mangiante, Carole Perruzzi, Rebekah K. O'Donnell, Ningning Zheng, Durga Udayakumar, Y. Rebecca Chin, Minzhou Huang, Damien Gerald, and Shiva Kazerounian
- Abstract
PDF file - 34K, Level of expression of mRNA of different genes in RhoB+/+ and RhoB-/- mouse tumor cells
- Published
- 2023
16. Data from Targeting Akt3 Signaling in Triple-Negative Breast Cancer
- Author
-
Alex Toker, Kornelia Polyak, Andrew H. Beck, Andriy Marusyk, Taku Yoshida, and Y. Rebecca Chin
- Abstract
Triple-negative breast cancer (TNBC) is currently the only major breast tumor subtype without effective targeted therapy and, as a consequence, in general has a poor outcome. To identify new therapeutic targets in TNBC, we performed a short hairpin RNA (shRNA) screen for protein kinases commonly amplified and overexpressed in breast cancer. Using this approach, we identified AKT3 as a gene preferentially required for the growth of TNBCs. Downregulation of Akt3 significantly inhibits the growth of TNBC lines in three-dimensional (3D) spheroid cultures and in mouse xenograft models, whereas loss of Akt1 or Akt2 have more modest effects. Akt3 silencing markedly upregulates the p27 cell-cycle inhibitor and this is critical for the ability of Akt3 to inhibit spheroid growth. In contrast with Akt1, Akt3 silencing results in only a minor enhancement of migration and does not promote invasion. Depletion of Akt3 in TNBC sensitizes cells to the pan-Akt inhibitor GSK690693. These results imply that Akt3 has a specific function in TNBCs; thus, its therapeutic targeting may provide a new treatment option for this tumor subtype. Cancer Res; 74(3); 964–73. ©2013 AACR.
- Published
- 2023
17. Supplementary Figure 5 from RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Laura E. Benjamin, George C. Prendergast, Alex Toker, Lawrence F. Brown, Olivier N. Kocher, James B. DuHadaway, Stephanie McNamara, Sharon Shechter, Arturo Bravo-Nuevo, Thuy L. Phung, Jacob Pourat, Lee Mangiante, Carole Perruzzi, Rebekah K. O'Donnell, Ningning Zheng, Durga Udayakumar, Y. Rebecca Chin, Minzhou Huang, Damien Gerald, and Shiva Kazerounian
- Abstract
PDF file - 52K, Level of silencing of Akt in RhoB-/- and RhoB+/+ mouse cells
- Published
- 2023
18. Supplementary Figure 6 from RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Laura E. Benjamin, George C. Prendergast, Alex Toker, Lawrence F. Brown, Olivier N. Kocher, James B. DuHadaway, Stephanie McNamara, Sharon Shechter, Arturo Bravo-Nuevo, Thuy L. Phung, Jacob Pourat, Lee Mangiante, Carole Perruzzi, Rebekah K. O'Donnell, Ningning Zheng, Durga Udayakumar, Y. Rebecca Chin, Minzhou Huang, Damien Gerald, and Shiva Kazerounian
- Abstract
PDF file - 26K, Level of EGFR on the surface of human MDA-MB-231 cells
- Published
- 2023
19. Supplementary Figure 1 from Targeting Akt3 Signaling in Triple-Negative Breast Cancer
- Author
-
Alex Toker, Kornelia Polyak, Andrew H. Beck, Andriy Marusyk, Taku Yoshida, and Y. Rebecca Chin
- Abstract
PDF file - 164K, AKT3 Expression and Survival in TNBC.
- Published
- 2023
20. Data from 3-Phosphoinositide–Dependent Kinase 1 Potentiates Upstream Lesions on the Phosphatidylinositol 3-Kinase Pathway in Breast Carcinoma
- Author
-
Ramon Parsons, Hanina Hibshoosh, Vundavalli V.V.S. Murty, Jean J. Zhao, Gordon B. Mills, Alex Toker, Mary Beth Terry, Jorma Isola, Carlos Cordon-Cardo, Jennifer S. Yu, Tulio Matos, Albert Rojtman, Lorenzo Memeo, Xiaomei Wang, Mervi Laakso, Sofia K. Gruvberger-Saal, Jennifer S. Ferris, Da-In Kim, Y. Rebecca Chin, Yuli Xie, Bhaskar Dutta, Subhadra Nandula, Jiaping Wu, Christina R. Barkley, Benjamin D. Hopkins, Susan Koujak, Lao H. Saal, Tao Su, and Matthew Maurer
- Abstract
Lesions of ERBB2, PTEN, and PIK3CA activate the phosphatidylinositol 3-kinase (PI3K) pathway during cancer development by increasing levels of phosphatidylinositol-3,4,5-triphosphate (PIP3). 3-Phosphoinositide-dependent kinase 1 (PDK1) is the first node of the PI3K signal output and is required for activation of AKT. PIP3 recruits PDK1 and AKT to the cell membrane through interactions with their pleckstrin homology domains, allowing PDK1 to activate AKT by phosphorylating it at residue threonine-308. We show that total PDK1 protein and mRNA were overexpressed in a majority of human breast cancers and that 21% of tumors had five or more copies of the gene encoding PDK1, PDPK1. We found that increased PDPK1 copy number was associated with upstream pathway lesions (ERBB2 amplification, PTEN loss, or PIK3CA mutation), as well as patient survival. Examination of an independent set of breast cancers and tumor cell lines derived from multiple forms of human cancers also found increased PDK1 protein levels associated with such upstream pathway lesions. In human mammary cells, PDK1 enhanced the ability of upstream lesions to signal to AKT, stimulate cell growth and migration, and rendered cells more resistant to PDK1 and PI3K inhibition. After orthotopic transplantation, PDK1 overexpression was not oncogenic but dramatically enhanced the ability of ERBB2 to form tumors. Our studies argue that PDK1 overexpression and increased PDPK1 copy number are common occurrences in cancer that potentiate the oncogenic effect of upstream lesions on the PI3K pathway. Therefore, we conclude that alteration of PDK1 is a critical component of oncogenic PI3K signaling in breast cancer. [Cancer Res 2009;69(15):6299–306]
- Published
- 2023
21. High Throughput Confined Migration Microfluidic Device for Drug Screening
- Author
-
Zihan Yang, Zhihang Zhou, Tongxu Si, Zhengdong Zhou, Li Zhou, Y. Rebecca Chin, Liang Zhang, Xinyuan Guan, and Mengsu Yang
- Subjects
Biomaterials ,General Materials Science ,General Chemistry ,Biotechnology - Published
- 2023
22. An Integrated and Multi-Functional Droplet-Based Microfluidic Platform for Digital DNA Amplification
- Author
-
Yuan Wang, Xiaoyu Zhou, Tao Xu, Huayang Fu, Chi-Chun Fong, Y. Rebecca Chin, Liang Zhang, Xinyuan Guan, and Mengsu Yang
- Published
- 2023
23. TCOF1 upregulation in triple-negative breast cancer promotes stemness and tumour growth and correlates with poor prognosis
- Author
-
William C. Cho, C Geoffrey Lau, Kui Ming Chan, Grace Y. K. So, Liang Zhang, Hao Huang, Xin Wang, Saravanan Ramakrishnan, Victor W.S. Ma, Jianyang Hu, Qingling He, Wah Cheuk, Y. Rebecca Chin, Yilin Pan, and Yuni Lai
- Subjects
Cancer Research ,Mice, Nude ,Triple Negative Breast Neoplasms ,Biology ,Article ,Mice ,Breast cancer ,Downregulation and upregulation ,In vivo ,Cell Line, Tumor ,Databases, Genetic ,medicine ,Animals ,Humans ,Triple-negative breast cancer ,Cell Proliferation ,Cancer stem cells ,Effector ,Computational Biology ,Nuclear Proteins ,Cancer ,Phosphoproteins ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Oncology ,Neoplastic Stem Cells ,Cancer research ,Stem cell - Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with poor prognosis. By performing multiomic profiling, we recently uncovered super-enhancer heterogeneity between breast cancer subtypes. Our data also revealed TCOF1 as a putative TNBC-specific super-enhancer-regulated gene. TCOF1 plays a critical role in craniofacial development but its function in cancer remains unclear. Methods Overall survival and multivariant Cox regression analyses were conducted using the METABRIC data set. The effect of TCOF1 knockout on TNBC growth and stemness was evaluated by in vitro and in vivo assays. RNA-seq and rescue experiments were performed to explore the underlying mechanisms. Results TCOF1 is frequently upregulated in TNBC and its elevated expression correlates with shorter overall survival. TCOF1 depletion significantly inhibits the growth and stemness of basal-like TNBC, but not of mesenchymal-like cells, highlighting the distinct molecular dependency in different TNBC subgroups. RNA-seq uncovers several stem cell molecules regulated by TCOF1. We further demonstrate that KIT is a downstream effector of TCOF1 in mediating TNBC stemness. TCOF1 expression in TNBC is regulated by the predicted super-enhancer. Conclusions TCOF1 depletion potently attenuates the growth and stemness of basal-like TNBC. Expression of TCOF1 may serve as a TNBC prognostic marker and a therapeutic target.
- Published
- 2021
24. Cellular uptake, tissue penetration, biodistribution, and biosafety of threose nucleic acids: Assessing in vitro and in vivo delivery
- Author
-
Fei Wang, Ling Sum Liu, Pan Li, Cia Hin Lau, Hoi Man Leung, Y Rebecca Chin, Chung Tin, and Pik Kwan Lo
- Subjects
Biomaterials ,Biomedical Engineering ,Bioengineering ,Cell Biology ,Molecular Biology ,Biotechnology - Abstract
Compared with siRNAs or other antisense oligonucleotides (ASOs), the chemical simplicity, DNA/RNA binding capability, folding ability of tertiary structure, and excellent physiological stability of threose nucleic acid (TNA) motivate scientists to explore it as a novel molecular tool in biomedical applications. Although ASOs reach the target cells/tumors, insufficient tissue penetration and distribution of ASOs result in poor therapeutic efficacy. Therefore, the study of the time course of drug absorption, biodistribution, metabolism, and excretion is of significantly importance. In this work, the pharmacokinetics and biosafety of TNAs in living organisms are investigated. We found that synthetic TNAs exhibited excellent biological stability, low cytotoxicity, and substantial uptake in living cells without transfection. Using U87 three-dimensional (3D) multicellular spheroids to mimic the
- Published
- 2022
25. Cancer stem cells: advances in biology and clinical translation—a Keystone Symposia report
- Author
-
Anna Dubrovska, Lichun Ma, Wei Guo, Dean G. Tang, J. Joseph Melenhorst, Justin D. Lathia, Sonam Bhatia, Jennifer Cable, Panagiotis Karras, Lei Zhou, Wolf R Wiedemeyer, Xin Wei Wang, Shaheen Sikandar, Junfang Ji, Markus Grompe, Duanqing Pei, Y Rebecca Chin, Chunzhang Yang, Klaus H. Kaestner, Ning Zhang, Richard M. White, Lola M. Reid, Clemens A. Schmitt, Michael M. Shen, Calvin J. Kuo, Erwei Song, Stephanie Ma, Irene Oi-Lin Ng, Carmen Chak-Lui Wong, and Irving L. Weissman
- Subjects
Research Report ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,Translational Research, Biomedical ,History and Philosophy of Science ,Cancer stem cell ,Neoplasms ,medicine ,Biomarkers, Tumor ,Tumor Microenvironment ,Animals ,Humans ,Tumor growth ,Progenitor cell ,Tumor microenvironment ,General Neuroscience ,Cancer ,Translation (biology) ,Congresses as Topic ,Tumor control ,medicine.disease ,Cancer research ,Neoplastic Stem Cells ,Carcinogenesis - Abstract
The test for the cancer stem cell (CSC) hypothesis is to find a target expressed on all, and only CSCs in a patient tumor, then eliminate all cells with that target that eliminates the cancer. That test has not yet been achieved, but CSC diagnostics and targets found on CSCs and some other cells have resulted in a few clinically relevant therapies. However, it has become apparent that eliminating the subset of tumor cells characterized by self-renewal properties is essential for long-term tumor control. CSCs are able to regenerate and initiate tumor growth, recapitulating the heterogeneity present in the tumor before treatment. As great progress has been made in identifying and elucidating the biology of CSCs as well as their interactions with the tumor microenvironment, the time seems ripe for novel therapeutic strategies that target CSCs to find clinical applicability. On May 19-21, 2021, researchers in cancer stem cells met virtually for the Keystone eSymposium "Cancer Stem Cells: Advances in Biology and Clinical Translation" to discuss recent advances in the understanding of CSCs as well as clinical efforts to target these populations.
- Published
- 2021
26. Upregulation of Receptor Tyrosine Kinase Activity and Stemness as Resistance Mechanisms to Akt Inhibitors in Breast Cancer
- Author
-
Tiffany Tsang, Qingling He, Emily B. Cohen, Casey Stottrup, Evan C. Lien, Huiqi Zhang, C. Geoffrey Lau, and Y. Rebecca Chin
- Subjects
Cancer Research ,Oncology ,breast cancer ,Akt ,cancer stemness ,drug resistance - Abstract
The PI3K/Akt pathway is frequently deregulated in human cancers, and multiple Akt inhibitors are currently under clinical evaluation. Based on the experience from other molecular targeted therapies, however, it is likely that acquired resistance will be developed in patients treated with Akt inhibitors. We established breast cancer models of acquired resistance by prolonged treatment of cells with allosteric or ATP-competitive Akt inhibitors. Phospho-Receptor tyrosine kinase (Phospho-RTK) arrays revealed hyper-phosphorylation of multiple RTKS, including EGFR, Her2, HFGR, EhpB3 and ROR1, in Akt-inhibitor-resistant cells. Importantly, resistance can be overcome by treatment with an EGFR inhibitor. We further showed that cancer stem cells (CSCs) are enriched in breast tumor cells that have developed resistance to Akt inhibitors. Several candidates of CSC regulators, such as ID4, are identified by RNA sequencing. Cosmic analysis indicated that sensitivity of tumor cells to Akt inhibitors can be predicted by ID4 and stem cell/epithelial–mesenchymal transition pathway targets. These findings indicate the potential of targeting the EGFR pathway and CSC program to circumvent Akt inhibitor resistance in breast cancer.
- Published
- 2022
27. ANLN Enhances Triple-Negative Breast Cancer Stemness Through TWIST1 and BMP2 and Promotes its Spheroid Growth
- Author
-
Alishba Maryam and Y. Rebecca Chin
- Subjects
cancer stem cells ,3D culture ,0301 basic medicine ,QH301-705.5 ,Biology ,medicine.disease_cause ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,ANLN ,03 medical and health sciences ,breast cancer ,0302 clinical medicine ,Breast cancer ,Downregulation and upregulation ,Cancer stem cell ,medicine ,Molecular Biosciences ,Biology (General) ,CRISPR/Cas9 ,Molecular Biology ,Triple-negative breast cancer ,Original Research ,Anilin ,Cell growth ,Spheroid ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Carcinogenesis - Abstract
ANLN is frequently upregulated in triple-negative breast cancer (TNBC) and its high expression in tumors are significantly associated with poor survival and recurrence, thereby it has been proposed to function as a prognostic marker for breast cancer. However, the specific function and molecular mechanisms by which ANLN promotes TNBC tumorigenesis remain elusive. Using multiomic profiling, we recently uncovered ANLN as a TNBC-specific gene driven by super-enhancer. Here, by Crispr/Cas9 editing, we showed that knockout of ANLN inhibits spheroid growth of TNBC. Interestingly, its effect on cell proliferation in 2D cultures is minimal. ANLN depletion inhibits mammosphere formation and clonogenicity potently, suggesting its important function in regulating cancer stem cells (CSCs). We screened a panel of stem cell-related genes and uncovered several CSC genes regulated by ANLN. We further identify TWIST1 and BMP2 as essential genes that mediate ANLN’s function in stemness but not spheroid growth. These findings may contribute to search for effective targeted therapies to treat TNBC.
- Published
- 2021
28. Defining super-enhancer landscape in triple-negative breast cancer by multiomic profiling
- Author
-
Hao Huang, Yuchen Zhang, Wei Wang, William C. Cho, Saravanan Ramakrishnan, Haiying Ma, Alishba Maryam, Jingyu Li, Xin Wang, Qinghua Huang, Grace Y. K. So, Wah Cheuk, Y. Rebecca Chin, Kui Ming Chan, Jianyang Hu, Liang Zhang, and Victor W.S. Ma
- Subjects
0301 basic medicine ,Science ,Mice, Nude ,General Physics and Astronomy ,Triple Negative Breast Neoplasms ,Disease ,Biology ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Gene regulatory networks ,ANLN ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Super-enhancer ,Cell Line, Tumor ,parasitic diseases ,medicine ,Animals ,Humans ,natural sciences ,skin and connective tissue diseases ,Triple-negative breast cancer ,Epigenomics ,Gene Editing ,Multidisciplinary ,Oncogene ,Gene Expression Profiling ,Microfilament Proteins ,technology, industry, and agriculture ,Forkhead Transcription Factors ,General Chemistry ,Proto-Oncogene Proteins c-met ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Enhancer Elements, Genetic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Carcinogenesis - Abstract
Breast cancer is a heterogeneous disease, affecting over 3.5 million women worldwide, yet the functional role of cis-regulatory elements including super-enhancers in different breast cancer subtypes remains poorly characterized. Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with a poor prognosis. Here we apply integrated epigenomic and transcriptomic profiling to uncover super-enhancer heterogeneity between breast cancer subtypes, and provide clinically relevant biological insights towards TNBC. Using CRISPR/Cas9-mediated gene editing, we identify genes that are specifically regulated by TNBC-specific super-enhancers, including FOXC1 and MET, thereby unveiling a mechanism for specific overexpression of the key oncogenes in TNBC. We also identify ANLN as a TNBC-specific gene regulated by super-enhancer. Our studies reveal a TNBC-specific epigenomic landscape, contributing to the dysregulated oncogene expression in breast tumorigenesis., Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with poor prognostic outcomes. Here the authors characterize super-enhancer heterogeneity and they identify genes that are specifically regulated by TNBC-specific super-enhancers, including FOXC1, MET and ANLN.
- Published
- 2021
29. Upregulation of AKT3 Confers Resistance to the AKT Inhibitor MK2206 in Breast Cancer
- Author
-
Casey Stottrup, Y. Rebecca Chin, and Tiffany Tsang
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,medicine.medical_treatment ,AKT1 ,Antineoplastic Agents ,Breast Neoplasms ,AKT2 ,Biology ,Article ,Targeted therapy ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Akt Inhibitor MK2206 ,Humans ,Epithelial–mesenchymal transition ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Dose-Response Relationship, Drug ,Cancer ,Cellular Reprogramming ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Phenotype ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Gene Knockdown Techniques ,Cancer research ,Female ,Heterocyclic Compounds, 3-Ring ,Proto-Oncogene Proteins c-akt - Abstract
Acquired resistance to molecular targeted therapy represents a major challenge for the effective treatment of cancer. Hyperactivation of the PI3K/AKT pathway is frequently observed in virtually all human malignancies, and numerous PI3K and AKT inhibitors are currently under clinical evaluation. However, mechanisms of acquired resistance to AKT inhibitors have yet to be described. Here, we use a breast cancer preclinical model to identify resistance mechanisms to a small molecule allosteric AKT inhibitor, MK2206. Using a step-wise and chronic high-dose exposure, breast cancer cell lines harboring oncogenic PI3K resistant to MK2206 were established. Using this model, we reveal that AKT3 expression is markedly upregulated in AKT inhibitor–resistant cells. Induction of AKT3 is regulated epigenetically by the bromodomain and extra terminal domain proteins. Importantly, knockdown of AKT3, but not AKT1 or AKT2, in resistant cells restores sensitivity to MK2206. AKT inhibitor–resistant cells also display an epithelial to mesenchymal transition phenotype as assessed by alterations in the levels of E-Cadherin, N-Cadherin, and vimentin, as well as enhanced invasiveness of tumor spheroids. Notably, the invasive morphology of resistant spheroids is diminished upon AKT3 depletion. We also show that resistance to MK2206 is reversible because upon drug removal resistant cells regain sensitivity to AKT inhibition, accompanied by reexpression of epithelial markers and reduction of AKT3 expression, implying that epigenetic reprogramming contributes to acquisition of resistance. These findings provide a rationale for developing therapeutics targeting AKT3 to circumvent acquired resistance in breast cancer. Mol Cancer Ther; 15(8); 1964–74. ©2016 AACR.
- Published
- 2016
30. RhoB Differentially Controls Akt Function in Tumor Cells and Stromal Endothelial Cells during Breast Tumorigenesis
- Author
-
Y. Rebecca Chin, Laura E. Benjamin, George C. Prendergast, Damien Gerald, James B. DuHadaway, Shiva Kazerounian, Lawrence F. Brown, Lee E. Mangiante, Olivier Kocher, Thuy L. Phung, Minzhou Huang, Carole A. Perruzzi, Ningning Zheng, Sharon Shechter, Alex Toker, Jacob Pourat, Rebekah K. O'Donnell, Durga Udayakumar, Stephanie L. McNamara, and Arturo Bravo-Nuevo
- Subjects
Cancer Research ,Stromal cell ,Angiogenesis ,RHOB ,Immunoblotting ,Breast Neoplasms ,Mice, Transgenic ,Biology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Article ,Mice ,RhoB GTP-Binding Protein ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunoprecipitation ,rhoB GTP-Binding Protein ,In Situ Hybridization ,Tumor microenvironment ,Neovascularization, Pathologic ,Carcinoma, Ductal, Breast ,Endothelial Cells ,Cancer ,Flow Cytometry ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Oncology ,Cancer cell ,Cancer research ,Female ,Stromal Cells ,Carcinogenesis ,Proto-Oncogene Proteins c-akt - Abstract
Tumors are composed of cancer cells but also a larger number of diverse stromal cells in the tumor microenvironment. Stromal cells provide essential supports to tumor pathophysiology but the distinct characteristics of their signaling networks are not usually considered in developing drugs to target tumors. This oversight potentially confounds proof-of-concept studies and increases drug development risks. Here, we show in established murine and human models of breast cancer how differential regulation of Akt by the small GTPase RhoB in cancer cells or stromal endothelial cells determines their dormancy versus outgrowth when angiogenesis becomes critical. In cancer cells in vitro or in vivo, RhoB functions as a tumor suppressor that restricts EGF receptor (EGFR) cell surface occupancy as well as Akt signaling. However, after activation of the angiogenic switch, RhoB functions as a tumor promoter by sustaining endothelial Akt signaling, growth, and survival of stromal endothelial cells that mediate tumor neoangiogenesis. Altogether, the positive impact of RhoB on angiogenesis and progression supercedes its negative impact in cancer cells themselves. Our findings elucidate the dominant positive role of RhoB in cancer. More generally, they illustrate how differential gene function effects on signaling pathways in the tumor stromal component can complicate the challenge of developing therapeutics to target cancer pathophysiology. Cancer Res; 73(1); 50–61. ©2012 AACR.
- Published
- 2013
31. Acetylation-Dependent Regulation of Skp2 Function
- Author
-
Lydia W.S. Finley, Daming Gao, Zhiwei Wang, Pier Paolo Pandolfi, Lixin Wan, Shavali Shaik, Hidefumi Fukushima, Wen Yang, Alex Toker, Bo Zhai, Wenyi Wei, Keiko Nakayama, Alan W. Lau, Steven P. Gygi, Y. Rebecca Chin, Hiroyuki Inuzuka, Marcia C. Haigis, and Julie Teruya-Feldstein
- Subjects
Male ,Cytoplasm ,SIRT3 ,Molecular Sequence Data ,Breast Neoplasms ,P300-CBP Transcription Factors ,Protein Sorting Signals ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Ubiquitin ,Cell Movement ,Cell Line, Tumor ,Animals ,Humans ,NLS ,p300-CBP Transcription Factors ,Amino Acid Sequence ,S-Phase Kinase-Associated Proteins ,Biochemistry, Genetics and Molecular Biology(all) ,Casein Kinase I ,Cadherin ,Lysine ,Ubiquitination ,Prostatic Neoplasms ,Acetylation ,Cell migration ,Cadherins ,Disease Models, Animal ,Cancer research ,biology.protein ,Protein Processing, Post-Translational ,Sequence Alignment ,Nuclear localization sequence - Abstract
Summary Aberrant Skp2 signaling has been implicated as a driving event in tumorigenesis. Although the underlying molecular mechanisms remain elusive, cytoplasmic Skp2 correlates with more aggressive forms of breast and prostate cancers. Here, we report that Skp2 is acetylated by p300 at K68 and K71, which is a process that can be antagonized by the SIRT3 deacetylase. Inactivation of SIRT3 leads to elevated Skp2 acetylation, which leads to increased Skp2 stability through impairment of the Cdh1-mediated proteolysis pathway. As a result, Skp2 oncogenic function is increased, whereby cells expressing an acetylation-mimetic mutant display enhanced cellular proliferation and tumorigenesis in vivo. Moreover, acetylation of Skp2 in the nuclear localization signal (NLS) promotes its cytoplasmic retention, and cytoplasmic Skp2 enhances cellular migration through ubiquitination and destruction of E-cadherin. Thus, our study identifies an acetylation-dependent regulatory mechanism governing Skp2 oncogenic function and provides insight into how cytoplasmic Skp2 controls cellular migration.
- Published
- 2012
32. NFAT promotes carcinoma invasive migration through glypican-6
- Author
-
Y. Rebecca Chin, Aura Kaunisto, Gary K. Yiu, and Alex Toker
- Subjects
Glypican ,PEI, polyethyleimine ,Biochemistry ,glypican ,p38 Mitogen-Activated Protein Kinases ,COX-2, cyclo-oxygenase-2 ,0302 clinical medicine ,Cell Movement ,10. No inequality ,Wnt Signaling Pathway ,transcription factor ,beta Catenin ,0303 health sciences ,HA, haemagglutinin ,RT, reverse transcription ,Wnt signaling pathway ,NFAT ,TCF, T-cell factor ,3. Good health ,Cell biology ,GAPDH, glyceraldehyde-3-phosphate dehydrogenase ,030220 oncology & carcinogenesis ,shRNA, small-hairpin RNA ,JNK, c-Jun N-terminal kinase ,Female ,cancer invasion ,AP-1, activator protein-1 ,LPA, lysophosphatidic acid ,Research Article ,dox, doxycycline ,Transcriptional Activation ,Beta-catenin ,DYRK, dual-specificity tyrosine-phosphorylated and -regulated kinase ,Breast Neoplasms ,Biology ,EMSA, electrophoretic mobility-shift assay ,Wnt-5a Protein ,03 medical and health sciences ,breast cancer ,Glypicans ,Cell Line, Tumor ,Proto-Oncogene Proteins ,PGE2, prostaglandin E2 ,Gene silencing ,Humans ,Neoplasm Invasiveness ,Molecular Biology ,Transcription factor ,030304 developmental biology ,EGF, epidermal growth factor ,nuclear factor of activated T-cells (NFAT) ,NFATC Transcription Factors ,JNK Mitogen-Activated Protein Kinases ,Cell Biology ,Wnt Proteins ,NFAT, nuclear factor of activated T-cells ,siRNA, small interfering RNA ,Cancer cell ,Cancer research ,biology.protein ,ENPP2, exonucloeotide pyrophosphatase and phosphodiesterase 2 ,GPC, glypican ,MAPK, mitogen-activated protein kinase - Abstract
Invasive migration of carcinoma cells is a prerequisite for the metastatic dissemination of solid tumours. Numerous mechanisms control the ability of cancer cells to acquire a motile and invasive phenotype, and subsequently degrade and invade the basement membrane. Several genes that are up-regulated in breast carcinoma are responsible for mediating the metastatic cascade. Recent studies have revealed that the NFAT (nuclear factor of activated T-cells) is a transcription factor that is highly expressed in aggressive breast cancer cells and tissues, and mediates invasion through transcriptional induction of pro-invasion and migration genes. In the present paper we demonstrate that NFAT promotes breast carcinoma invasion through induction of GPC (glypican) 6, a cell-surface glycoprotein. NFAT transcriptionally regulates GPC6 induction in breast cancer cells and binds to three regulatory elements in the GPC6 proximal promoter. Expression of GPC6 in response to NFAT signalling promotes invasive migration, whereas GPC6 silencing with shRNA (small-hairpin RNA) potently blocks this phenotype. The mechanism by which GPC6 promotes invasive migration involves inhibition of canonical β-catenin and Wnt signalling, and up-regulation of non-canonical Wnt5A signalling leading to the activation of JNK (c-Jun N-terminal kinase) and p38 MAPK (mitogen-activated protein kinase). Thus GPC6 is a novel NFAT target gene in breast cancer cells that promotes invasive migration through Wnt5A signalling.
- Published
- 2011
33. Akt isoform-specific signaling in breast cancer
- Author
-
Alex Toker and Y. Rebecca Chin
- Subjects
Palladin ,AKT1 ,Cancer ,Breast Neoplasms ,AKT2 ,Review ,Cell Biology ,Biology ,Phosphoproteins ,medicine.disease ,medicine.disease_cause ,Metastasis ,Cell biology ,Cytoskeletal Proteins ,Cellular and Molecular Neuroscience ,medicine ,Humans ,Protein Isoforms ,Female ,Carcinogenesis ,Proto-Oncogene Proteins c-akt ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Signal Transduction - Abstract
Numerous studies have shown that Akt isoforms promote tumorigenesis by enhancing cancer cell survival and growth, and it is well established that signaling through the Akt upstream regulator PI 3-K enhances cancer cell migration. Therefore, it is conventionally accepted that PI 3-K/Akt pathway promotes tumor formation and metastasis. A few years ago, studies from several laboratories added a new layer to the pleiotropic effects of Akt function by showing that the Akt1 isoform inhibits breast cancer cell migration and invasion, whereas Akt2 promotes these phenotypes. These studies challenged the dogma and identified non-redundant functions of Akt isoforms in cancer progression. The identification of palladin as an Akt1-specific substrate in our recently published work has exemplified distinct Akt isoform-specific signaling in breast cancer. Here, we review these findings and discuss the implications for the understanding of the mechanistic basis for designing more effective anti-cancer therapeutics targeting the Akt pathway.
- Published
- 2011
34. 3-Phosphoinositide–Dependent Kinase 1 Potentiates Upstream Lesions on the Phosphatidylinositol 3-Kinase Pathway in Breast Carcinoma
- Author
-
Xiaomei Wang, Mervi Laakso, Y. Rebecca Chin, Carlos Cordon-Cardo, Benjamin D. Hopkins, Hanina Hibshoosh, Susan Koujak, Lao H. Saal, Mary Beth Terry, Jorma Isola, Tao Su, Jiaping Wu, Da In Kim, Jennifer S. Ferris, Yuli Xie, Jean J. Zhao, Jennifer S. Yu, Bhaskar Dutta, Albert Rojtman, Gordon B. Mills, Sofia K. Gruvberger-Saal, Christina R. Barkley, Vundavalli V. Murty, Subhadra V. Nandula, Lorenzo Memeo, Alex Toker, Tulio Matos, Matthew Maurer, and Ramon Parsons
- Subjects
Cellular signal transduction ,Cancer Research ,animal structures ,Receptor, ErbB-2 ,Carcinogenesis ,Gene Dosage ,Breast Neoplasms ,Cell Growth Processes ,Mice, SCID ,Protein Serine-Threonine Kinases ,Article ,3-Phosphoinositide-Dependent Protein Kinases ,Mice ,Phosphatidylinositol 3-Kinases ,Protein kinases ,Pathology ,medicine ,Animals ,Humans ,PTEN ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Mice, Inbred BALB C ,biology ,Kinase ,Cancer ,medicine.disease ,Oncogene Protein v-akt ,Pleckstrin homology domain ,Cell Transformation, Neoplastic ,Oncology ,Breast--Cancer ,Cancer research ,biology.protein ,Female ,Signal transduction ,Signal Transduction ,Phosphoinositide-dependent kinase-1 - Abstract
Lesions of ERBB2, PTEN, and PIK3CA activate the phosphatidylinositol 3-kinase (PI3K) pathway during cancer development by increasing levels of phosphatidylinositol-3,4,5-triphosphate (PIP3). 3-Phosphoinositide-dependent kinase 1 (PDK1) is the first node of the PI3K signal output and is required for activation of AKT. PIP3 recruits PDK1 and AKT to the cell membrane through interactions with their pleckstrin homology domains, allowing PDK1 to activate AKT by phosphorylating it at residue threonine-308. We show that total PDK1 protein and mRNA were overexpressed in a majority of human breast cancers and that 21% of tumors had five or more copies of the gene encoding PDK1, PDPK1. We found that increased PDPK1 copy number was associated with upstream pathway lesions (ERBB2 amplification, PTEN loss, or PIK3CA mutation), as well as patient survival. Examination of an independent set of breast cancers and tumor cell lines derived from multiple forms of human cancers also found increased PDK1 protein levels associated with such upstream pathway lesions. In human mammary cells, PDK1 enhanced the ability of upstream lesions to signal to AKT, stimulate cell growth and migration, and rendered cells more resistant to PDK1 and PI3K inhibition. After orthotopic transplantation, PDK1 overexpression was not oncogenic but dramatically enhanced the ability of ERBB2 to form tumors. Our studies argue that PDK1 overexpression and increased PDPK1 copy number are common occurrences in cancer that potentiate the oncogenic effect of upstream lesions on the PI3K pathway. Therefore, we conclude that alteration of PDK1 is a critical component of oncogenic PI3K signaling in breast cancer. [Cancer Res 2009;69(15):6299–306]
- Published
- 2009
35. Inhibition of Rb Phosphorylation Leads to mTORC2-Mediated Activation of Akt
- Author
-
Jianping Guo, Kai Xu, Wenjian Gan, Jinfang Zhang, Christian Berrios, Alex Toker, Piotr Sicinski, Fei Wu, Evan C. Lien, Yan Geng, James A. DeCaprio, Y. Rebecca Chin, Pengda Liu, Wenyi Wei, and Bin Wang
- Subjects
0301 basic medicine ,Time Factors ,Cyclin D ,mTORC1 ,mTORC2 ,Retinoblastoma Protein ,Mice ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Molecular Targeted Therapy ,Phosphorylation ,TOR Serine-Threonine Kinases ,Drug Synergism ,3. Good health ,RNA Interference ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,Protein Binding ,Signal Transduction ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,Transfection ,Article ,03 medical and health sciences ,Enzyme activator ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Molecular Biology ,Protein kinase B ,Transcription factor ,Protein Kinase Inhibitors ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Dose-Response Relationship, Drug ,Cyclin-Dependent Kinase 4 ,Cell Biology ,Cyclin-Dependent Kinase 6 ,HCT116 Cells ,Enzyme Activation ,030104 developmental biology ,HEK293 Cells ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-akt ,HeLa Cells - Abstract
The retinoblastoma (Rb) protein exerts its tumor suppressor function primarily by inhibiting the E2F family of transcription factors that govern cell cycle progression. However, it remains largely elusive whether hyper-phosphorylated, non-E2F1-interacting form, of Rb has any physiological role. Here, we report that hyper-phosphorylated Rb directly binds to, and suppresses the function of mTORC2, but not mTORC1. Mechanistically, Rb, but not p107 nor p130, interacts with Sin1 and blocks the access of Akt to mTORC2, leading to attenuated Akt activation and increased sensitivity to chemotherapeutic drugs. As such, inhibition of Rb phosphorylation by depleting cyclin D, or using CDK4/6 inhibitors, releases Rb-mediated mTORC2 suppression. This, in turn, leads to elevated Akt activation to confer resistance to chemotherapeutic drugs in Rb-proficient cells, which can be attenuated with Akt inhibitors. Therefore, our work provides a molecular basis for the synergistic usage of CDK4/6 and Akt inhibitors in treating Rb-proficient cancer.
- Published
- 2015
36. Adenovirus RID complex enhances degradation of internalized tumour necrosis factor receptor 1 without affecting its rate of endocytosis
- Author
-
Y. Rebecca Chin and Marshall S. Horwitz
- Subjects
Endosome ,medicine.medical_treatment ,education ,Cell ,Biology ,Endocytosis ,medicine.disease_cause ,Receptors, Tumor Necrosis Factor ,Adenoviridae ,Cell Line ,Viral Proteins ,Virology ,Adenovirus E3 Proteins ,medicine ,Humans ,Receptor ,technology, industry, and agriculture ,respiratory system ,Up-Regulation ,Tumor Necrosis Factor Decoy Receptors ,medicine.anatomical_structure ,Cytokine ,Receptors, Tumor Necrosis Factor, Type I ,Multiprotein Complexes ,Biotinylation ,Tumor necrosis factor alpha ,human activities - Abstract
The receptor internalization and degradation (RID) complex of adenovirus plays an important role in modulating the immune response by downregulating the surface levels of tumour necrosis factor receptor 1 (TNFR1), thereby inhibiting NF-κB activation. Total cellular content of TNFR1 is also reduced in the presence of RID, which can be inhibited by treatment with lysosomotropic agents. In this report, surface biotinylation experiments revealed that, although RID and TNFR1 were able to form a complex on the cell surface, the rate of TNFR1 endocytosis was not affected by RID. However, the degradation of internalized TNFR1 was enhanced significantly in the presence of RID. Therefore, these data suggest that RID downregulates TNFR1 levels by altering the fate of internalized TNFR1 that becomes associated with RID at the plasma membrane, probably by promoting its sorting into endosomal/lysosomal degradation compartments.
- Published
- 2006
37. PTEN-deficient tumors depend on AKT2 for maintenance and survival
- Author
-
Xin Yuan, Alex Toker, Steven P. Balk, and Y. Rebecca Chin
- Subjects
Male ,AKT1 ,AKT2 ,Article ,Receptor, IGF Type 1 ,Small hairpin RNA ,Prostate cancer ,medicine ,Gene silencing ,PTEN ,Humans ,Protein Isoforms ,Cell Proliferation ,biology ,Akt/PKB signaling pathway ,PTEN Phosphohydrolase ,Cancer ,Prostatic Neoplasms ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Oncology ,p21-Activated Kinases ,embryonic structures ,Cancer research ,biology.protein ,Heterografts ,Proto-Oncogene Proteins c-akt ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Loss of PTEN is a common event in many cancers and leads to hyperactivation of the PI3K–AKT signaling pathway. The mechanisms by which AKT isoforms mediate signaling to phenotypes associated with PTEN inactivation in cancer have not been defined. Here, we show that AKT2 is exclusively required for PTEN-deficient prostate tumor spheroid maintenance, whereas AKT1 is dispensable. shRNA silencing of AKT2 but not AKT1 promotes regression of prostate cancer xenografts. Mechanistically, we show that AKT2 silencing upregulates p21 and the proapoptotic protein BAX and downregulates the insulin-like growth factor receptor-1. We also show that p21 is an effector of AKT2 in mediating prostate tumor maintenance. Moreover, AKT2 is also exclusively required for the maintenance and survival of other PTEN-deficient solid tumors, including breast cancer and glioblastoma. These findings identify a specific function for AKT2 in mediating survival of PTEN-deficient tumors and provide a rationale for developing therapeutics targeting AKT2. Significance: Depletion of AKT2, but not AKT1, induces potent tumor regression in PTEN-deficient prostate cancer xenografts, concomitant with upregulation of p21, which may serve as a potential biomarker for screening AKT2 activity in clinical samples. The specific role of AKT2 in tumor maintenance provides a rationale for the development of isoform-specific inhibitors for patients with PTEN-deficient cancers. Cancer Discov; 4(8); 942–55. ©2014 AACR. This article is highlighted in the In This Issue feature, p. 855
- Published
- 2014
38. Akt-ing up on SRPK1 – Oncogene or Tumor Suppressor?
- Author
-
Y. Rebecca Chin and Alex Toker
- Subjects
Male ,Mice, 129 Strain ,Carcinogenesis ,AKT1 ,Mice, Nude ,AKT2 ,Biology ,Protein Serine-Threonine Kinases ,Article ,MAP2K7 ,Mice ,Cell Adhesion ,Phosphoprotein Phosphatases ,Animals ,Humans ,c-Raf ,Phosphorylation ,Protein kinase B ,Molecular Biology ,Cells, Cultured ,Cellular Senescence ,Mice, Knockout ,PHLPP ,Akt/PKB signaling pathway ,Nuclear Proteins ,Protein phosphatase 2 ,Cell Biology ,Tumor Burden ,Enzyme Activation ,Colonic Neoplasms ,Cancer research ,Female ,Protein Processing, Post-Translational ,Proto-Oncogene Proteins c-akt ,Neoplasm Transplantation - Abstract
Akt activation is a hallmark of human cancers. Here, we report a critical mechanism for regulation of Akt activity by the splicing kinase SRPK1, a downstream Akt target for transducing growth signals to regulate splicing. Surprisingly, we find that SRPK1 has a tumor suppressor function because ablation of SRPK1 in mouse embryonic fibroblasts induces cell transformation. We link the phenotype to constitutive Akt activation from genome-wide phosphoproteomics analysis and discover that downregulated SRPK1 impairs the recruitment of the Akt phosphatase PHLPP1 (pleckstrin homology (PH) domain leucine-rich repeat protein phosphatase) to Akt. Interestingly, SRPK1 overexpression is also tumorigenic because excess SRPK1 squelches PHLPP1. Thus, aberrant SRPK1 expression in either direction induces constitutive Akt activation, providing a mechanistic basis for previous observations that SRPK1 is downregulated in some cancer contexts and upregulated in others.
- Published
- 2014
39. TARGETING AKT3 SIGNALING IN TRIPLE NEGATIVE BREAST CANCER
- Author
-
Alex Toker, Kornelia Polyak, Taku Yoshida, Y. Rebecca Chin, Andrew H. Beck, and Andriy Marusyk
- Subjects
Cancer Research ,medicine.medical_treatment ,Gene Expression ,AKT2 ,Triple Negative Breast Neoplasms ,Biology ,AKT3 ,Article ,Targeted therapy ,Mice ,Breast cancer ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,RNA Isoforms ,Tumor Cells, Cultured ,Gene silencing ,Animals ,Humans ,Neoplasm Invasiveness ,Protein Kinase Inhibitors ,Triple-negative breast cancer ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Oncology ,Drug Resistance, Neoplasm ,embryonic structures ,Cancer research ,Female ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Triple-negative breast cancer (TNBC) is currently the only major breast tumor subtype without effective targeted therapy and, as a consequence, in general has a poor outcome. To identify new therapeutic targets in TNBC, we performed a short hairpin RNA (shRNA) screen for protein kinases commonly amplified and overexpressed in breast cancer. Using this approach, we identified AKT3 as a gene preferentially required for the growth of TNBCs. Downregulation of Akt3 significantly inhibits the growth of TNBC lines in three-dimensional (3D) spheroid cultures and in mouse xenograft models, whereas loss of Akt1 or Akt2 have more modest effects. Akt3 silencing markedly upregulates the p27 cell-cycle inhibitor and this is critical for the ability of Akt3 to inhibit spheroid growth. In contrast with Akt1, Akt3 silencing results in only a minor enhancement of migration and does not promote invasion. Depletion of Akt3 in TNBC sensitizes cells to the pan-Akt inhibitor GSK690693. These results imply that Akt3 has a specific function in TNBCs; thus, its therapeutic targeting may provide a new treatment option for this tumor subtype. Cancer Res; 74(3); 964–73. ©2013 AACR.
- Published
- 2013
40. PtdIns(3,4,5)P3-Dependent Activation of the mTORC2 Kinase Complex
- Author
-
Lewis C. Cantley, Jinfang Zhang, Y. Rebecca Chin, Jianping Guo, Pengda Liu, John Blenis, Wenyi Wei, Bing Su, Kohei Ogura, Bin Wang, Wenjian Gan, and Alex Toker
- Subjects
Models, Molecular ,Proto-Oncogene Proteins c-akt ,Amino Acid Motifs ,Molecular Conformation ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,mTORC2 ,Catalysis ,Article ,Proto-Oncogene Proteins p21(ras) ,Mice ,Phosphatidylinositol Phosphates ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Phosphorylation ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Binding Sites ,Cell growth ,TOR Serine-Threonine Kinases ,Cell biology ,Pleckstrin homology domain ,Enzyme Activation ,Disease Models, Animal ,Oncology ,Protein kinase domain ,Multiprotein Complexes ,Mutation ,Heterografts ,Female ,Protein Binding - Abstract
mTOR serves as a central regulator of cell growth and metabolism by forming two distinct complexes, mTORC1 and mTORC2. Although mechanisms of mTORC1 activation by growth factors and amino acids have been extensively studied, the upstream regulatory mechanisms leading to mTORC2 activation remain largely elusive. Here, we report that the pleckstrin homology (PH) domain of SIN1, an essential and unique component of mTORC2, interacts with the mTOR kinase domain to suppress mTOR activity. More importantly, PtdIns(3,4,5)P3, but not other PtdInsPn species, interacts with SIN1-PH to release its inhibition on the mTOR kinase domain, thereby triggering mTORC2 activation. Mutating critical SIN1 residues that mediate PtdIns(3,4,5)P3 interaction inactivates mTORC2, whereas mTORC2 activity is pathologically increased by patient-derived mutations in the SIN1-PH domain, promoting cell growth and tumor formation. Together, our study unravels a PI3K-dependent mechanism for mTORC2 activation, allowing mTORC2 to activate AKT in a manner that is regulated temporally and spatially by PtdIns(3,4,5)P3. Significance: The SIN1-PH domain interacts with the mTOR kinase domain to suppress mTOR activity, and PtdIns(3,4,5)P3 binds the SIN1-PH domain to release its inhibition on the mTOR kinase domain, leading to mTORC2 activation. Cancer patient–derived SIN1-PH domain mutations gain oncogenicity by loss of suppressing mTOR activity as a means to facilitate tumorigenesis. Cancer Discov; 5(11); 1194–209. ©2015 AACR. See related commentary by Yuan and Guan, p. 1127. This article is highlighted in the In This Issue feature, p. 1111
- Published
- 2013
41. mTOR drives its own activation via SCF(βTrCP)-dependent degradation of the mTOR inhibitor DEPTOR
- Author
-
J. Wade Harper, John M. Asara, Bo Zhai, Jason W. Locasale, Meng Kwang Marcus Tan, Lewis C. Cantley, Y. Rebecca Chin, Lixin Wan, Costas A. Lyssiotis, Daming Gao, Wenyi Wei, Shavali Shaik, Steven P. Gygi, Hidefumi Fukushima, Alex Toker, Hiroyuki Inuzuka, and Pengda Liu
- Subjects
SKP Cullin F-Box Protein Ligases ,TOR Serine-Threonine Kinases ,Autophagy ,RPTOR ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,mTORC1 ,Biology ,DEPTOR ,Transfection ,mTORC2 ,Article ,Cell biology ,Cancer research ,Humans ,Kinase activity ,Signal transduction ,Phosphorylation ,Molecular Biology ,PI3K/AKT/mTOR pathway ,HeLa Cells ,Signal Transduction - Abstract
Summary The activities of both mTORC1 and mTORC2 are negatively regulated by their endogenous inhibitor, DEPTOR. As such, the abundance of DEPTOR is a critical determinant in the activity status of the mTOR network. DEPTOR stability is governed by the 26S-proteasome through a largely unknown mechanism. Here we describe an mTOR-dependent phosphorylation-driven pathway for DEPTOR destruction via SCF βTrCP . DEPTOR phosphorylation by mTOR in response to growth signals, and in collaboration with casein kinase I (CKI), generates a phosphodegron that binds βTrCP. Failure to degrade DEPTOR through either degron mutation or βTrCP depletion leads to reduced mTOR activity, reduced S6 kinase activity, and activation of autophagy to reduce cell growth. This work expands the current understanding of mTOR regulation by revealing a positive feedback loop involving mTOR and CKI-dependent turnover of its inhibitor, DEPTOR, suggesting that misregulation of the DEPTOR destruction pathway might contribute to aberrant activation of mTOR in disease.
- Published
- 2011
42. THE ACTIN BUNDLING PROTEIN PALLADIN IS AN AKT1-SPECIFIC SUBSTRATE THAT REGULATES BREAST CANCER CELL MIGRATION
- Author
-
Y. Rebecca Chin and Alex Toker
- Subjects
Recombinant Fusion Proteins ,AKT2 ,Breast Neoplasms ,Biology ,Transfection ,Article ,Substrate Specificity ,Breast cancer ,Cell Movement ,Spheroids, Cellular ,medicine ,Serine ,Humans ,Neoplasm Invasiveness ,Phosphorylation ,Molecular Biology ,Cell Shape ,PI3K/AKT/mTOR pathway ,Cytoskeleton ,Palladin ,Cancer ,Cell migration ,Cell Biology ,Actin cytoskeleton ,medicine.disease ,Phosphoproteins ,Actins ,Cell biology ,Enzyme Activation ,Cytoskeletal Proteins ,embryonic structures ,Mutation ,Cancer research ,Female ,RNA Interference ,Signal transduction ,Proto-Oncogene Proteins c-akt ,HeLa Cells ,Signal Transduction - Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway is frequently deregulated in cancer. Downstream of PI3K, Akt1 and Akt2 have opposing roles in breast cancer invasive migration, leading to metastatic dissemination. Here, we identify palladin, an actin-associated protein, as an Akt1-specific substrate that modulates breast cancer cell invasive migration. Akt1, but not Akt2, phosphorylates palladin at Ser507 in a domain that is critical for F-actin bundling. Downregulation of palladin enhances migration and invasion of breast cancer cells and induces abnormal branching morphogenesis in 3D cultures. Palladin phosphorylation at Ser507 is required for Akt1-mediated inhibition of breast cancer cell migration and also for F-actin bundling, leading to the maintenance of an organized actin cytoskeleton. These findings identify palladin as an Akt1-specific substrate that regulates cell motility and provide a molecular mechanism that accounts for the functional distinction between Akt isoforms in breast cancer cell signaling to cell migration.
- Published
- 2010
43. Akt/protein kinase b and glycogen synthase kinase-3beta signaling pathway regulates cell migration through the NFAT1 transcription factor
- Author
-
Christopher K. Hansen, Y. Rebecca Chin, Alex Toker, and Merav Yoeli-Lerner
- Subjects
Cancer Research ,Proteasome Endopeptidase Complex ,Cell Survival ,AKT1 ,AKT2 ,Breast Neoplasms ,Cell Growth Processes ,Biology ,Article ,Glycogen Synthase Kinase 3 ,Mice ,GSK-3 ,Cell Movement ,Cell Line, Tumor ,Animals ,Humans ,Molecular Biology ,Protein kinase B ,1-Phosphatidylinositol 4-Kinase ,PI3K/AKT/mTOR pathway ,Glycogen Synthase Kinase 3 beta ,NFATC Transcription Factors ,Akt/PKB signaling pathway ,NFAT ,Cell biology ,Oncology ,Cancer research ,cardiovascular system ,NIH 3T3 Cells ,Proto-Oncogene Proteins c-akt - Abstract
The phosphoinositide 3-kinase (PI3K) pathway regulates a multitude of cellular processes. Deregulation of PI3K signaling is often observed in human cancers. A major effector of PI3K is Akt/protein kinase B (PKB). Recent studies have pointed to distinct roles of Akt/PKB isoforms in cancer cell signaling. Studies have shown that Akt1 (PKBα) can attenuate breast cancer cell motility, whereas Akt2 (PKBβ) enhances this phenotype. Here, we have evaluated the mechanism by which Akt1 blocks the migration of breast cancer cells through the transcription factor NFAT. A major effector of Akt/PKB is glycogen synthase kinase-3β (GSK-3β), also a NFAT kinase. Inhibition of GSK-3β using short hairpin RNA or a selective inhibitor potently blocks breast cancer cell migration concomitant with a reduction in NFAT activity. GSK-3β-mediated inhibition of NFAT activity is due to proteasomal degradation. Experiments using GSK-3β mutants, which are unresponsive to Akt/PKB, reveal that inhibition of cell migration by Akt/PKB is mediated by GSK-3β. These effects are recapitulated at the levels of NFAT degradation by the proteasome. Our studies show that activation of Akt/PKB leads to inactivation of the effector GSK-3β and the outcome of this signaling event is degradation of NFAT by the proteasome and subsequent inhibition of cell migration. (Mol Cancer Res 2009;7(3):425–32)
- Published
- 2009
44. Function of Akt/PKB Signaling to Cell Motility, Invasion and the Tumor Stroma in Cancer
- Author
-
Y. Rebecca Chin and Alex Toker
- Subjects
Vascular Endothelial Growth Factor A ,Fibrosarcoma ,Breast Neoplasms ,Biology ,Article ,Metastasis ,Mice ,Cell Movement ,Cell Line, Tumor ,medicine ,PTEN ,Animals ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Tumor microenvironment ,Neovascularization, Pathologic ,Akt/PKB signaling pathway ,Cell Biology ,Fibroblasts ,medicine.disease ,Cell biology ,Cell Compartmentation ,Neoplasm Proteins ,Cancer cell ,Cancer research ,biology.protein ,Female ,Signal transduction ,Stromal Cells ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The serine/threonine protein kinase Akt is a major signal transducer of the phosphoinositide 3-kinase (PI 3-K) pathway in all cells and tissues and plays a pivotal role in the maintenance of cellular processes including cell growth, proliferation, survival and metabolism. The frequent aberrant activation of the PI 3-K/Akt pathway in human cancer has made it an attractive therapeutic target. Numerous studies have provided a comprehensive understanding of the specific functions of Akt signaling in cancer cells as well as the surrounding tumor microenvironment and this has informed and enabled the development of therapeutic drugs to target both PI 3-K and Akt. However, recent studies have provided evidence for distinct functions of the three mammalian Akt isoforms, particularly with respect to the regulation of cell motility and metastasis of breast cancer. Here we discuss the mechanisms by which Akt signaling contributes to invasive migration and tumor metastasis, and highlight recent advances in our understanding of the contribution of the Akt pathway in the tumor-associated stroma.
- Published
- 2008
45. Abstract A42: Increased Akt3 expression as a resistance mechanism to targeted therapy
- Author
-
Y. Rebecca Chin and Alex Toker
- Subjects
Cancer Research ,medicine.medical_treatment ,AKT1 ,Cancer ,AKT2 ,Biology ,Pharmacology ,medicine.disease ,Targeted therapy ,Breast cancer ,Oncology ,Cancer research ,medicine ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Triple-negative breast cancer - Abstract
Resistance to targeted therapy poses a major challenge for successful treatment of cancers. Despite a number of drugs targeting the PI 3-K/Akt pathway are in clinical development, mechanisms of resistance to drugs targeting this pathway have not been studied extensively. To determine how breast cancer cells develop resistance to Akt inhibitors (Akti), we generated Akti-resistant lines using dose escalation regimens with pan-Akt inhibitors (ATP-competitive: GDC0068, GSK690693; Allosteric: MK2206). Interestingly, in multiple Akti-resistant breast cancer lines, we observed a robust increase in the protein expression of Akt3, but not Akt1 or Akt2. We were intrigued by this observation, as we have recently demonstrated that Akt3 has a predominant function in regulating growth of triple negative breast cancer cells in three-dimensional spheroids and in xenograft models. The increased expression of Akt3 in Akti-resistant cells occurs via a genetic event, since the levels of Akt3 remain elevated after 3 weeks of Akt inhibitor removal. Moreover, upon Akt inhibitor removal in resistant lines, cells remain resistant to Akt inhibition. The enhanced Akt3 expression is observed in both luminal and basal-like triple-negative breast cancer resistant lines. Importantly, knockdown of Akt3, but not Akt1 or Akt2, results in re-sensitization of resistant cells to Akti. Mechanistically, using anti-phosphotyrosine receptor antibody array (RTK array), we observed increased phosphorylation of several RTKs in resistant cells, including EGFR, Her2, and ROR1. Akt3 knockdown in resistant cells results in reduction of pEGFR, pHer3 and pHer2, suggesting that Akt3 plays a role in the regulation of multiple RTKs in resistant cells. Epithelial-mesenchymal transition (EMT) is implicated in therapeutic resistance of cancers. Interestingly, several of our resistant lines display EMT properties including downregulation of E-Cadherin, and upregulation of N-Cadherin as well as Vimentin. We propose that enhanced Akt3 expression is a general mechanism by which breast cancer cells acquire resistance to Akti targeted therapy. Current efforts are aimed at investigating the mechanistic basis for Akt3-mediated resistance, and demonstrating the physiological relevance of these findings in clinical samples as well as in mouse models. The role of Akt3 in regulating EMT during establishment of resistance will also be explored. These studies will provide insights into molecular mechanisms of therapeutic resistance to drugs targeting the PI 3-K/Akt pathway, as well as information for designing combination strategies and next generation therapy. Citation Format: Y Rebecca Chin, Alex Toker. Increased Akt3 expression as a resistance mechanism to targeted therapy. [abstract]. In: Proceedings of the AACR Special Conference: Targeting the PI3K-mTOR Network in Cancer; Sep 14-17, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(7 Suppl):Abstract nr A42.
- Published
- 2015
46. Mechanism for removal of tumor necrosis factor receptor 1 from the cell surface by the adenovirus RIDalpha/beta complex
- Author
-
Y. Rebecca Chin and Marshall S. Horwitz
- Subjects
Endosome ,Immunology ,Down-Regulation ,Apoptosis ,Endosomes ,Biology ,Virus Replication ,Microbiology ,Receptors, Tumor Necrosis Factor ,Cell Line ,Downregulation and upregulation ,Virology ,Adenovirus E3 Proteins ,Humans ,Tyrosine ,Dynamin ,Adaptor Proteins, Signal Transducing ,Adenoviruses, Human ,Intracellular Signaling Peptides and Proteins ,Signal transducing adaptor protein ,Nuclear Proteins ,respiratory system ,Fas receptor ,Molecular biology ,Virus-Cell Interactions ,Tumor Necrosis Factor Decoy Receptors ,Receptors, Tumor Necrosis Factor, Type I ,Insect Science ,Tumor necrosis factor receptor 1 ,Carrier Proteins ,Lysosomes ,Co-Repressor Proteins ,Molecular Chaperones - Abstract
Proteins encoded in adenovirus early region 3 have important immunoregulatory properties. We have recently shown that the E3-10.4K/14.5K (RIDα/β) complex downregulates tumor necrosis factor receptor 1 (TNFR1) expression at the plasma membrane. To study the role of the RIDβ tyrosine sorting motif in the removal of surface TNFR1, tyrosine 122 on RIDβ was mutated to alanine or phenylalanine. Both RIDβ mutations not only abolished the downregulation of surface TNFR1 but paradoxically increased surface TNFR1 levels. RID also downregulates other death receptors, such as FAS; however, surface FAS expression was not increased by RIDβ mutants, suggesting that regulation of TNFR1 and that of FAS by RID are mechanistically different. In the mixing experiments, the wild-type (WT) RID-mediated TNFR1 downregulation was partially inhibited in the presence of RIDβ mutants, indicating that the mutants compete for TNFR1 access. Indeed, an association between RIDβ and TNFR1 was shown by coimmunoprecipitation. In contrast, the mutants did not affect the WT RID-induced downregulation of FAS. These differential effects support a model in which RID associates with TNFR1 on the plasma membrane, whereas RID probably associates with FAS in a cytoplasmic compartment. By using small interfering RNA against the μ2 subunit of adaptor protein 2, dominant negative dynamin construct K44A, and the lysosomotropic agents bafilomycin A1 and ammonium chloride, we also demonstrated that surface TNFR1 was internalized by RID by a clathrin-dependent process involving μ2 and dynamin, followed by degradation of TNFR1 via an endosomal/lysosomal pathway.
- Published
- 2005
47. Akt2 regulates expression of the actin-bundling protein palladin
- Author
-
Y. Rebecca Chin and Alex Toker
- Subjects
Biophysics ,AKT1 ,AKT2 ,Biology ,Biochemistry ,Article ,Cell Line ,Breast cancer ,Structural Biology ,Palladin ,Protein stability ,Genetics ,Humans ,RNA, Messenger ,Protein Structure, Quaternary ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Actin ,Regulation of gene expression ,Cell Biology ,Actin cytoskeleton ,Phosphoproteins ,Actins ,Up-Regulation ,Isoenzymes ,Cytoskeletal Proteins ,Gene Expression Regulation ,Akt isoform ,embryonic structures ,Cancer research ,Protein Multimerization ,Proto-Oncogene Proteins c-akt - Abstract
The phosphatidylinositol 3-kinase/Akt pathway is responsible for key aspects of tumor progression, and is frequently hyperactivated in cancer. We have recently identified palladin, an actin-bundling protein that functions to control the actin cytoskeleton, as an Akt1-specific substrate that inhibits breast cancer cell migration. Here we have identified a role for Akt isoforms in the regulation of palladin expression. Akt2, but not Akt1, enhances palladin expression by maintaining protein stability and upregulating transcription. These data reveal that Akt signaling regulates the stability of palladin, and further supports the notion that Akt isoforms have distinct and specific roles in tumorigenesis.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.