The mechanistic target of rapamycin complex 1 (mTORC1) is an evolutionarily conserved serine/threonine kinase complex. It is a central regulator of cellular anabolic processes by integrating a variety of upstream inputs, including growth factors, nutrients, energy and oxygen status. It is being increasingly appreciated that downstream targets of mTORC1 can communicate with its upstream regulators, through various feedback loops. These feedback mechanisms play a critical role in stabilizing the entire network. They also have great significance in a variety of human diseases. For example, aberrant mTORC1 activation (e.g. as a result of chronic overfeeding) can cause insulin resistance, a hallmark of type 2 diabetes. The feedback loops are also particularly relevant in cancer biology. mTORC1 is hyperactivated in a major faction of human cancers. Unfortunately, inhibition of mTORC1 using rapamycin analogs (rapalogs) as single agents is cytostatic but not cytotoxic. This is mainly due to the activation of upstream, pro-survival kinases (e.g. Akt) upon mTORC1 inhibition. Understanding how to avoid the relief of feedback loops will be critical to overcome rapamycin resistance for treating human cancer. To this end, we recently used a quantitative phosphoproteomic approach to systematically characterizing the mTOR-regulated phosphoproteome. Detailed biochemical characterization of a novel mTORC1 substrate, Grb10, showed that mTORC1-mediated phosphorylation stabilized this protein, which led to its accumulation, and subsequent inhibition of insulin and IGF1 signaling. Our unpublished results further indicate that cells with hyperactive mTORC1 also secrete a protein(s) that is able to block IGF1 signaling. In this proposal, we will investigate how mTORC1 may inhibit the function of IGF1 through remodeling the secretome.
In Aim 1, we will use a quantitative mass spectrometry approach to comprehensively characterizing the mTORC1-regulated secretome.
In Aim 2, we will elucidate the molecular mechanism by which mTORC1 regulates the expression of this secreted protein, and determine its role in mTORC1-mediated feedback inhibition of IGF1 signaling.
In Aim3, we will investigate the non-cell autonomous function of this secreted protein, and its interplay with Grb10 in vivo. We will accomplish our goals with a multi- disciplinary approach, utilizing tools including proteomics, biochemistry, molecular biology and animal models. The results will greatly facilitate our understanding of mTORC1-mediated feedback loops. Knowledge we garner in this proposal will also be necessary to further explore how we might better target these feedback mechanisms for treating various human diseases.

Public Health Relevance

It is being increasingly appreciated that downstream targets of mTORC1 communicate with its upstream regulators (i.e. insulin and IGF1), through various feedback loops. These feedback mechanisms have great significance in a variety of human diseases, in particular diabetes and cancer. In this proposal, we will develop advanced proteomic technologies, and combine them with biochemical approaches to understand how mTORC1 inhibits IGF1 signaling.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM114160-03
Application #
9341365
Study Section
Molecular and Integrative Signal Transduction Study Section (MIST)
Program Officer
Melillo, Amanda A
Project Start
2015-09-15
Project End
2020-08-31
Budget Start
2017-09-01
Budget End
2018-08-31
Support Year
3
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Texas Sw Medical Center Dallas
Department
Biochemistry
Type
Schools of Medicine
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
Wang, Zhiqiang; Ma, Jing; Miyoshi, Chika et al. (2018) Quantitative phosphoproteomic analysis of the molecular substrates of sleep need. Nature 558:435-439
Yamaguchi, H; Du, Y; Nakai, K et al. (2018) EZH2 contributes to the response to PARP inhibitors through its PARP-mediated poly-ADP ribosylation in breast cancer. Oncogene 37:208-217
Dutchak, Paul A; Estill-Terpack, Sandi J; Plec, Abigail A et al. (2018) Loss of a Negative Regulator of mTORC1 Induces Aerobic Glycolysis and Altered Fiber Composition in Skeletal Muscle. Cell Rep 23:1907-1914
Zhen, Yuanli; Zhang, Yajie; Yu, Yonghao (2017) A Cell-Line-Specific Atlas of PARP-Mediated Protein Asp/Glu-ADP-Ribosylation in Breast Cancer. Cell Rep 21:2326-2337
Zhao, Xiaozheng; Huffman, Kenneth E; Fujimoto, Junya et al. (2017) Quantitative Proteomic Analysis of Optimal Cutting Temperature (OCT) Embedded Core-Needle Biopsy of Lung Cancer. J Am Soc Mass Spectrom 28:2078-2089
Eskiocak, Banu; McMillan, Elizabeth A; Mendiratta, Saurabh et al. (2017) Biomarker Accessible and Chemically Addressable Mechanistic Subtypes of BRAF Melanoma. Cancer Discov 7:832-851
Zhang, Peng; He, Dandan; Xu, Yi et al. (2017) Genome-wide identification and differential analysis of translational initiation. Nat Commun 8:1749
Zhang, Yuanyuan; Zhang, Yajie; Yu, Yonghao (2017) Global Phosphoproteomic Analysis of Insulin/Akt/mTORC1/S6K Signaling in Rat Hepatocytes. J Proteome Res 16:2825-2835
Lee, Gina; Zheng, Yuxiang; Cho, Sungyun et al. (2017) Post-transcriptional Regulation of De Novo Lipogenesis by mTORC1-S6K1-SRPK2 Signaling. Cell 171:1545-1558.e18
Ding, Ming; Bruick, Richard K; Yu, Yonghao (2016) Secreted IGFBP5 mediates mTORC1-dependent feedback inhibition of IGF-1 signalling. Nat Cell Biol 18:319-27

Showing the most recent 10 out of 12 publications