Identification of JAK family tyrosine kinases as receptor-associated signaling molecules for growth hormone (GH) receptors (GHR) and the many other members of the cytokine receptor superfamily was a major step forward in our understanding of the cellular basis for GH actions. Upon GH binding, GHR-associated JAK2 is activated and phosphorylates tyrosines within JAK2 itself and GHR. These phosphotyrosines form binding sites for a variety of signaling molecules whose recruitment to and/or activation by JAK2-GHR complexes initiates the signaling pathways that lead to the diverse physiological responses to GH. While some signaling molecules bind to phosphotyrosines within GHR, little is known about the signaling molecules that bind to phosphotyrosines within JAK2, despite the presence of 49 tyrosines within JAK2. SH2-B is one of the few known JAK2 binding proteins. In response to GH, SH2-B binds to JAK2, is phosphorylated by JAK2 and enhances GH-induced activation of JAK2. Independent of its ability to activate JAK2, SH2-B enhances GH regulation of the actin cytoskeleton and cell motility. Finally, SH2-B shuttles between the cytoplasm and the nucleus, and enhances the nuclear export of forkhead transcription factors.
The aim of this proposal is to test the hypothesis that SH2-B is a multi-functional adapter/scaffolding protein that enhances GH activation of JAK2, enhances GH regulation of the actin cytoskeleton and regulates GH responses in the nucleus.
Aim 1 lexamines the mechanism by which SH2-B activates JAK2.
Aim 2 will further define the role of SH2-B in GH-mediated changes in the actin cytoskeleton. Multiple proteins implicated in the regulation of the actin cytoskeleton have been tentatively identified as SH2-Bbeta binding proteins. Their binding to SH2-B will be confirmed and the effect of SH2-B on their function and subcellular location determined.
Aim 3 will examine whether SH2-B regulates transcription by shuttling proteins into or out of the nucleus. Finally, Aim 4 will use a variety of protein depletion techniques to identify the physiological role for SH2-B in GH action, including siRNA, SH2-B"""""""" mice, and murine embryo fibroblasts and tissues from SH2-B -/- mice. These studies will provide insight into the role of SH2-B in the function of GH and other cytokines, hormones and growth factors (e.g. interferon-gamma, insulin, nerve growth factor) that also utilize SH2-B as a signaling protein. Such insight is relevant to understanding the mechanism(s) by which GH regulates body growth and metabolism, and other SH2-B activating ligands contribute to, prevent and/or alleviate symptoms of a variety of diseases including various cancers, diabetes, multiple sclerosis, and diseases of the immune system.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
2R01DK054222-06
Application #
6826387
Study Section
Endocrinology Study Section (END)
Program Officer
Blondel, Olivier
Project Start
1999-07-01
Project End
2009-05-31
Budget Start
2004-07-05
Budget End
2005-05-31
Support Year
6
Fiscal Year
2004
Total Cost
$367,298
Indirect Cost
Name
University of Michigan Ann Arbor
Department
Physiology
Type
Schools of Medicine
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Joe, Ray M; Flores, Anabel; Doche, Michael E et al. (2017) Phosphorylation of the Unique C-terminal Tail of the Alpha Isoform of the Scaffold Protein SH2B1 Controls the Ability of SH2B1alpha to Enhance Nerve Growth Factor Function. Mol Cell Biol :
Carter-Su, Christin; Schwartz, Jessica; Argetsinger, Lawrence S (2016) Growth hormone signaling pathways. Growth Horm IGF Res 28:11-5
Xue, Xiang; Ramakrishnan, Sadeesh K; Weisz, Kevin et al. (2016) Iron Uptake via DMT1 Integrates Cell Cycle with JAK-STAT3 Signaling to Promote Colorectal Tumorigenesis. Cell Metab 24:447-461
Pearce, Laura R; Joe, Ray; Doche, Michael E et al. (2014) Functional characterization of obesity-associated variants involving the ? and ? isoforms of human SH2B1. Endocrinology 155:3219-26
Su, Hsiao-Wen; Lanning, Nathan J; Morris, David L et al. (2013) Phosphorylation of the adaptor protein SH2B1? regulates its ability to enhance growth hormone-dependent macrophage motility. J Cell Sci 126:1733-43
Javadi, Mojib; Hofstätter, Edda; Stickle, Natalie et al. (2012) The SH2B1 adaptor protein associates with a proximal region of the erythropoietin receptor. J Biol Chem 287:26223-34
Doche, Michael E; Bochukova, Elena G; Su, Hsiao-Wen et al. (2012) Human SH2B1 mutations are associated with maladaptive behaviors and obesity. J Clin Invest 122:4732-6
Lanning, Nathan J; Su, Hsiao-Wen; Argetsinger, Lawrence S et al. (2011) Identification of SH2B1? as a focal adhesion protein that regulates focal adhesion size and number. J Cell Sci 124:3095-105
Maures, Travis J; Su, Hsiao-Wen; Argetsinger, Lawrence S et al. (2011) Phosphorylation controls a dual-function polybasic nuclear localization sequence in the adapter protein SH2B1? to regulate its cellular function and distribution. J Cell Sci 124:1542-52
Maures, Travis J; Chen, Linyi; Carter-Su, Christin (2009) Nucleocytoplasmic shuttling of the adapter protein SH2B1beta (SH2-Bbeta) is required for nerve growth factor (NGF)-dependent neurite outgrowth and enhancement of expression of a subset of NGF-responsive genes. Mol Endocrinol 23:1077-91

Showing the most recent 10 out of 26 publications