Name | SMAD specific E3 ubiquitin protein ligase 1 |
Description | This gene encodes a ubiquitin ligase that is specific for receptor-regulated SMAD proteins in the bone morphogenetic protein (BMP) pathway. This protein plays a key roll in the regulation of cell motility, cell signalling, and cell polarity. Alternative splicing results in multiple transcript variants encoding different isoforms.[provided by RefSeq, Dec 2010] |
Summary |
{"type": "root", "children": [{"type": "p", "children": [{"type": "t", "text": "\nSMURF1 is a HECT‐domain E3 ubiquitin ligase that plays a central role in controlling cytoskeletal dynamics and cell motility. It is recruited to cellular protrusions by polarity complexes and mediates localized degradation of RhoA, thereby regulating actin cytoskeleton reorganization and tumor cell migration. In addition, through its interaction with inhibitory Smad7, SMURF1 modulates the turnover of TGF‑β receptors and influences directional cell movement and polarity. These activities have been documented in diverse cellular contexts—from the regulation of lamellipodia formation in transformed cells to the modulation of migratory behavior during pathological conditions—supporting SMURF1’s pivotal function in governing cellular motility and invasive potential."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "1", "end_ref": "9"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nIn the realm of TGF‑β/BMP and osteogenic signaling, SMURF1 orchestrates receptor turnover and transcription factor degradation to finely tune cellular responsiveness and differentiation. By mediating ubiquitination and proteasomal degradation of critical effectors such as Runx2, and by promoting the nuclear export of inhibitory Smad7, SMURF1 acts as a negative regulator of the BMP–Smad–Runx2 cascade. Moreover, its activity can be modulated by upstream factors including Cdh1 and specific miRNAs, thus impacting osteoblast function, autophagosome maturation via substrates like UVRAG, and overall BMP signaling sensitivity in both normal bone formation and fibrotic conditions."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "10", "end_ref": "15"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nBeyond its roles in cell polarity and differentiation, SMURF1 contributes to diverse aspects of disease pathogenesis, particularly in cancer and stress responses. In several malignancies—including pancreatic, gastric, lung, ovarian, and colon cancers—SMURF1 is frequently amplified or upregulated, where it accelerates tumor progression by promoting the degradation of substrates that negatively regulate cell growth, adhesion, and immune responses. For example, SMURF1 targets proteins involved in antiviral signaling (such as STAT1 and MAVS), modulates endoplasmic reticulum homeostasis by degrading WFS1, and influences axon growth and vascular integrity through interactions with factors like CCM2 and ARHGAP26. Its activity is further regulated by interacting partners such as FKBP12, USP9X, Ndfip1, and transcription factors like USF2, while being targeted by several microRNAs (e.g. miR‑1254) and kinases (e.g. p70S6K) that affect its stability and substrate specificity. These multifaceted roles underscore SMURF1 as an important nodal regulator whose dysregulation impacts signaling pathways including the IFN‑γ, PI3K/Akt/mTOR, and ATR/Chk1 axes, ultimately affecting tumor cell invasion, metastasis, and cellular responses to DNA damage and developmental cues."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "16", "end_ref": "36"}]}, {"type": "t", "text": "\n"}]}, {"type": "rg", "children": [{"type": "r", "ref": 1, "children": [{"type": "t", "text": "Chie Suzuki, Gyo Murakami, Minoru Fukuchi, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Smurf1 regulates the inhibitory activity of Smad7 by targeting Smad7 to the plasma membrane."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2002)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M201901200"}], "href": "https://doi.org/10.1074/jbc.M201901200"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "12151385"}], "href": "https://pubmed.ncbi.nlm.nih.gov/12151385"}]}, {"type": "r", "ref": 2, "children": [{"type": "t", "text": "Hong-Rui Wang, Yue Zhang, Barish Ozdamar, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Regulation of cell polarity and protrusion formation by targeting RhoA for degradation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Science (2003)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1126/science.1090772"}], "href": "https://doi.org/10.1126/science.1090772"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "14657501"}], "href": "https://pubmed.ncbi.nlm.nih.gov/14657501"}]}, {"type": "r", "ref": 3, "children": [{"type": "t", "text": "Yoshihide Asano, Hironobu Ihn, Kenichi Yamane, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Impaired Smad7-Smurf-mediated negative regulation of TGF-beta signaling in scleroderma fibroblasts."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Clin Invest (2004)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1172/JCI16269"}], "href": "https://doi.org/10.1172/JCI16269"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "14722617"}], "href": "https://pubmed.ncbi.nlm.nih.gov/14722617"}]}, {"type": "r", "ref": 4, "children": [{"type": "t", "text": "Laurent Boyer, Laurent Turchi, Benoit Desnues, et al. "}, {"type": "b", "children": [{"type": "t", "text": "CNF1-induced ubiquitylation and proteasome destruction of activated RhoA is impaired in Smurf1-/- cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Biol Cell (2006)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1091/mbc.e05-09-0876"}], "href": "https://doi.org/10.1091/mbc.e05-09-0876"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "16540523"}], "href": "https://pubmed.ncbi.nlm.nih.gov/16540523"}]}, {"type": "r", "ref": 5, "children": [{"type": "t", "text": "Erik Sahai, Raquel Garcia-Medina, Jacques Pouysségur, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Smurf1 regulates tumor cell plasticity and motility through degradation of RhoA leading to localized inhibition of contractility."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Cell Biol (2007)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1083/jcb.200605135"}], "href": "https://doi.org/10.1083/jcb.200605135"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "17190792"}], "href": "https://pubmed.ncbi.nlm.nih.gov/17190792"}]}, {"type": "r", "ref": 6, "children": [{"type": "t", "text": "Erina Fukunaga, Yasumichi Inoue, Setsuro Komiya, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Smurf2 induces ubiquitin-dependent degradation of Smurf1 to prevent migration of breast cancer cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M710496200"}], "href": "https://doi.org/10.1074/jbc.M710496200"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18927080"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18927080"}]}, {"type": "r", "ref": 7, "children": [{"type": "t", "text": "Maoyuan Tian, Chunmei Bai, Qi Lin, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Binding of RhoA by the C2 domain of E3 ligase Smurf1 is essential for Smurf1-regulated RhoA ubiquitination and cell protrusive activity."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "FEBS Lett (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.febslet.2011.06.016"}], "href": "https://doi.org/10.1016/j.febslet.2011.06.016"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21708152"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21708152"}]}, {"type": "r", "ref": 8, "children": [{"type": "t", "text": "Arang Kwon, Hye-Lim Lee, Kyung Mi Woo, et al. "}, {"type": "b", "children": [{"type": "t", "text": "SMURF1 plays a role in EGF-induced breast cancer cell migration and invasion."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Cells (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1007/s10059-013-0233-4"}], "href": "https://doi.org/10.1007/s10059-013-0233-4"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24241683"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24241683"}]}, {"type": "r", "ref": 9, "children": [{"type": "t", "text": "Lin Yu, Xin Liu, Kang Cui, et al. "}, {"type": "b", "children": [{"type": "t", "text": "SND1 Acts Downstream of TGFβ1 and Upstream of Smurf1 to Promote Breast Cancer Metastasis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Res (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/0008-5472.CAN-14-2387"}], "href": "https://doi.org/10.1158/0008-5472.CAN-14-2387"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25596283"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25596283"}]}, {"type": "r", "ref": 10, "children": [{"type": "t", "text": "Yoshitaka Tajima, Kouichiro Goto, Minoru Yoshida, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Chromosomal region maintenance 1 (CRM1)-dependent nuclear export of Smad ubiquitin regulatory factor 1 (Smurf1) is essential for negative regulation of transforming growth factor-beta signaling by Smad7."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2003)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M212663200"}], "href": "https://doi.org/10.1074/jbc.M212663200"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "12519765"}], "href": "https://pubmed.ncbi.nlm.nih.gov/12519765"}]}, {"type": "r", "ref": 11, "children": [{"type": "t", "text": "Run Shen, Mo Chen, Yong-Jun Wang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Smad6 interacts with Runx2 and mediates Smad ubiquitin regulatory factor 1-induced Runx2 degradation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2006)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M506761200"}], "href": "https://doi.org/10.1074/jbc.M506761200"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "16299379"}], "href": "https://pubmed.ncbi.nlm.nih.gov/16299379"}]}, {"type": "r", "ref": 12, "children": [{"type": "t", "text": "Yali Liu, Wenjia Liu, Chenghu Hu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "MiR-17 modulates osteogenic differentiation through a coherent feed-forward loop in mesenchymal stem cells isolated from periodontal ligaments of patients with periodontitis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Stem Cells (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/stem.728"}], "href": "https://doi.org/10.1002/stem.728"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21898695"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21898695"}]}, {"type": "r", "ref": 13, "children": [{"type": "t", "text": "Lixin Wan, Weiguo Zou, Daming Gao, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Cdh1 regulates osteoblast function through an APC/C-independent modulation of Smurf1."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Cell (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.molcel.2011.09.024"}], "href": "https://doi.org/10.1016/j.molcel.2011.09.024"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22152476"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22152476"}]}, {"type": "r", "ref": 14, "children": [{"type": "t", "text": "Xing Feng, Yanyan Jia, Yuyu Zhang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Ubiquitination of UVRAG by SMURF1 promotes autophagosome maturation and inhibits hepatocellular carcinoma growth."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Autophagy (2019)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1080/15548627.2019.1570063"}], "href": "https://doi.org/10.1080/15548627.2019.1570063"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "30686098"}], "href": "https://pubmed.ncbi.nlm.nih.gov/30686098"}]}, {"type": "r", "ref": 15, "children": [{"type": "t", "text": "Lin Fu, Chun-Ping Cui, Xueli Zhang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The functions and regulation of Smurfs in cancers."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Semin Cancer Biol (2020)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.semcancer.2019.12.023"}], "href": "https://doi.org/10.1016/j.semcancer.2019.12.023"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "31899247"}], "href": "https://pubmed.ncbi.nlm.nih.gov/31899247"}]}, {"type": "r", "ref": 16, "children": [{"type": "t", "text": "Sreedhara Sangadala, Scott D Boden, Manjula Viggeswarapu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "LIM mineralization protein-1 potentiates bone morphogenetic protein responsiveness via a novel interaction with Smurf1 resulting in decreased ubiquitination of Smads."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2006)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M511013200"}], "href": "https://doi.org/10.1074/jbc.M511013200"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "16611643"}], "href": "https://pubmed.ncbi.nlm.nih.gov/16611643"}]}, {"type": "r", "ref": 17, "children": [{"type": "t", "text": "Ayako Suzuki, Tatsuhiro Shibata, Yutaka Shimada, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Identification of SMURF1 as a possible target for 7q21.3-22.1 amplification detected in a pancreatic cancer cell line by in-house array-based comparative genomic hybridization."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Sci (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1111/j.1349-7006.2008.00779.x"}], "href": "https://doi.org/10.1111/j.1349-7006.2008.00779.x"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18380791"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18380791"}]}, {"type": "r", "ref": 18, "children": [{"type": "t", "text": "Lisa E S Crose, Thomas L Hilder, Noah Sciaky, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Cerebral cavernous malformation 2 protein promotes smad ubiquitin regulatory factor 1-mediated RhoA degradation in endothelial cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2009)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.C900009200"}], "href": "https://doi.org/10.1074/jbc.C900009200"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19318350"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19318350"}]}, {"type": "r", "ref": 19, "children": [{"type": "t", "text": "Xing Guo, Shan Shen, Shanshan Song, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The E3 ligase Smurf1 regulates Wolfram syndrome protein stability at the endoplasmic reticulum."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M111.225615"}], "href": "https://doi.org/10.1074/jbc.M111.225615"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21454619"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21454619"}]}, {"type": "r", "ref": 20, "children": [{"type": "t", "text": "Chao Yuan, Jianni Qi, Xueying Zhao, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Smurf1 protein negatively regulates interferon-γ signaling through promoting STAT1 protein ubiquitination and degradation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M112.341198"}], "href": "https://doi.org/10.1074/jbc.M112.341198"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22474288"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22474288"}]}, {"type": "r", "ref": 21, "children": [{"type": "t", "text": "Vincent J Dalbo, Michael D Roberts, Scott Hassell, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Effects of pre-exercise feeding on serum hormone concentrations and biomarkers of myostatin and ubiquitin proteasome pathway activity."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Eur J Nutr (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1007/s00394-012-0349-x"}], "href": "https://doi.org/10.1007/s00394-012-0349-x"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22476926"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22476926"}]}, {"type": "r", "ref": 22, "children": [{"type": "t", "text": "Madhuvanthi Kannan, Shih-Ju Lee, Nicola Schwedhelm-Domeyer, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The E3 ligase Cdh1-anaphase promoting complex operates upstream of the E3 ligase Smurf1 in the control of axon growth."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Development (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1242/dev.081786"}], "href": "https://doi.org/10.1242/dev.081786"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22949615"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22949615"}]}, {"type": "r", "ref": 23, "children": [{"type": "t", "text": "Yetao Wang, Xiaomei Tong, Xin Ye "}, {"type": "b", "children": [{"type": "t", "text": "Ndfip1 negatively regulates RIG-I-dependent immune signaling by enhancing E3 ligase Smurf1-mediated MAVS degradation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Immunol (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.4049/jimmunol.1201445"}], "href": "https://doi.org/10.4049/jimmunol.1201445"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23087404"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23087404"}]}, {"type": "r", "ref": 24, "children": [{"type": "t", "text": "Yang Xie, Monika Avello, Markus Schirle, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Deubiquitinase FAM/USP9X interacts with the E3 ubiquitin ligase SMURF1 protein and protects it from ligase activity-dependent self-degradation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M112.430066"}], "href": "https://doi.org/10.1074/jbc.M112.430066"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23184937"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23184937"}]}, {"type": "r", "ref": 25, "children": [{"type": "t", "text": "J Nie, L Liu, G Xing, et al. "}, {"type": "b", "children": [{"type": "t", "text": "CKIP-1 acts as a colonic tumor suppressor by repressing oncogenic Smurf1 synthesis and promoting Smurf1 autodegradation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Oncogene (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/onc.2013.340"}], "href": "https://doi.org/10.1038/onc.2013.340"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23995790"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23995790"}]}, {"type": "r", "ref": 26, "children": [{"type": "t", "text": "Jiayi Wang, Yue Zhang, Wenhao Weng, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Impaired phosphorylation and ubiquitination by p70 S6 kinase (p70S6K) and Smad ubiquitination regulatory factor 1 (Smurf1) promote tribbles homolog 2 (TRIB2) stability and carcinogenic property in liver cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M113.503292"}], "href": "https://doi.org/10.1074/jbc.M113.503292"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24089522"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24089522"}]}, {"type": "r", "ref": 27, "children": [{"type": "t", "text": "Yu Cao, Cheng Wang, Xueli Zhang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Selective small molecule compounds increase BMP-2 responsiveness by inhibiting Smurf1-mediated Smad1/5 degradation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Sci Rep (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/srep04965"}], "href": "https://doi.org/10.1038/srep04965"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24828823"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24828823"}]}, {"type": "r", "ref": 28, "children": [{"type": "t", "text": "Meilin Wang, Lei Guo, Qingang Wu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "ATR/Chk1/Smurf1 pathway determines cell fate after DNA damage by controlling RhoB abundance."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Commun (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/ncomms5901"}], "href": "https://doi.org/10.1038/ncomms5901"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25249323"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25249323"}]}, {"type": "r", "ref": 29, "children": [{"type": "t", "text": "Ali Khammanivong, Raj Gopalakrishnan, Erin B Dickerson "}, {"type": "b", "children": [{"type": "t", "text": "SMURF1 silencing diminishes a CD44-high cancer stem cell-like population in head and neck squamous cell carcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Cancer (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1186/1476-4598-13-260"}], "href": "https://doi.org/10.1186/1476-4598-13-260"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25471937"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25471937"}]}, {"type": "r", "ref": 30, "children": [{"type": "t", "text": "Xun Tang, Xianzhen Chen, Yanfeng Xu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "CD166 positively regulates MCAM via inhibition to ubiquitin E3 ligases Smurf1 and βTrCP through PI3K/AKT and c-Raf/MEK/ERK signaling in Bel-7402 hepatocellular carcinoma cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cell Signal (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.cellsig.2015.05.006"}], "href": "https://doi.org/10.1016/j.cellsig.2015.05.006"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "26004137"}], "href": "https://pubmed.ncbi.nlm.nih.gov/26004137"}]}, {"type": "r", "ref": 31, "children": [{"type": "t", "text": "Alexander M K Rothman, Nadine D Arnold, Josephine A Pickworth, et al. "}, {"type": "b", "children": [{"type": "t", "text": "MicroRNA-140-5p and SMURF1 regulate pulmonary arterial hypertension."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Clin Invest (2016)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1172/JCI83361"}], "href": "https://doi.org/10.1172/JCI83361"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "27214554"}], "href": "https://pubmed.ncbi.nlm.nih.gov/27214554"}]}, {"type": "r", "ref": 32, "children": [{"type": "t", "text": "H Li, N Xiao, Y Wang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Smurf1 regulates lung cancer cell growth and migration through interaction with and ubiquitination of PIPKIγ."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Oncogene (2017)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/onc.2017.166"}], "href": "https://doi.org/10.1038/onc.2017.166"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "28581524"}], "href": "https://pubmed.ncbi.nlm.nih.gov/28581524"}]}, {"type": "r", "ref": 33, "children": [{"type": "t", "text": "Youmao Tao, Caixia Sun, Tao Zhang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "SMURF1 promotes the proliferation, migration and invasion of gastric cancer cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Oncol Rep (2017)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.3892/or.2017.5825"}], "href": "https://doi.org/10.3892/or.2017.5825"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "28731194"}], "href": "https://pubmed.ncbi.nlm.nih.gov/28731194"}]}, {"type": "r", "ref": 34, "children": [{"type": "t", "text": "Rui Baptista, Carla Marques, Steve Catarino, et al. "}, {"type": "b", "children": [{"type": "t", "text": "MicroRNA-424(322) as a new marker of disease progression in pulmonary arterial hypertension and its role in right ventricular hypertrophy by targeting SMURF1."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cardiovasc Res (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1093/cvr/cvx187"}], "href": "https://doi.org/10.1093/cvr/cvx187"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "29016730"}], "href": "https://pubmed.ncbi.nlm.nih.gov/29016730"}]}, {"type": "r", "ref": 35, "children": [{"type": "t", "text": "Yawen Tan, Yujiao Chen, Mengge Du, et al. "}, {"type": "b", "children": [{"type": "t", "text": "USF2 inhibits the transcriptional activity of Smurf1 and Smurf2 to promote breast cancer tumorigenesis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cell Signal (2019)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.cellsig.2018.09.013"}], "href": "https://doi.org/10.1016/j.cellsig.2018.09.013"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "30244169"}], "href": "https://pubmed.ncbi.nlm.nih.gov/30244169"}]}, {"type": "r", "ref": 36, "children": [{"type": "t", "text": "Mingkun Jiang, Liang Shi, Chao Yang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "miR-1254 inhibits cell proliferation, migration, and invasion by down-regulating Smurf1 in gastric cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cell Death Dis (2019)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/s41419-018-1262-x"}], "href": "https://doi.org/10.1038/s41419-018-1262-x"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "30631050"}], "href": "https://pubmed.ncbi.nlm.nih.gov/30631050"}]}]}]}
|
Proteins | SMUF1_HUMAN |
NCBI Gene ID | 57154 |
API | |
Download Associations | |
Predicted Functions |
![]() |
Co-expressed Genes |
![]() |
Expression in Tissues and Cell Lines |
![]() |
SMURF1 has 7,538 functional associations with biological entities spanning 9 categories (molecular profile, organism, functional term, phrase or reference, chemical, disease, phenotype or trait, structural feature, cell line, cell type or tissue, gene, protein or microRNA, sequence feature) extracted from 118 datasets.
Click the + buttons to view associations for SMURF1 from the datasets below.
If available, associations are ranked by standardized value
Dataset | Summary | |
---|---|---|
Allen Brain Atlas Adult Human Brain Tissue Gene Expression Profiles | tissues with high or low expression of SMURF1 gene relative to other tissues from the Allen Brain Atlas Adult Human Brain Tissue Gene Expression Profiles dataset. | |
Allen Brain Atlas Adult Mouse Brain Tissue Gene Expression Profiles | tissues with high or low expression of SMURF1 gene relative to other tissues from the Allen Brain Atlas Adult Mouse Brain Tissue Gene Expression Profiles dataset. | |
Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by Microarray | tissue samples with high or low expression of SMURF1 gene relative to other tissue samples from the Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by Microarray dataset. | |
Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by RNA-seq | tissue samples with high or low expression of SMURF1 gene relative to other tissue samples from the Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by RNA-seq dataset. | |
Allen Brain Atlas Prenatal Human Brain Tissue Gene Expression Profiles | tissues with high or low expression of SMURF1 gene relative to other tissues from the Allen Brain Atlas Prenatal Human Brain Tissue Gene Expression Profiles dataset. | |
Biocarta Pathways | pathways involving SMURF1 protein from the Biocarta Pathways dataset. | |
BioGPS Cell Line Gene Expression Profiles | cell lines with high or low expression of SMURF1 gene relative to other cell lines from the BioGPS Cell Line Gene Expression Profiles dataset. | |
BioGPS Human Cell Type and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of SMURF1 gene relative to other cell types and tissues from the BioGPS Human Cell Type and Tissue Gene Expression Profiles dataset. | |
BioGPS Mouse Cell Type and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of SMURF1 gene relative to other cell types and tissues from the BioGPS Mouse Cell Type and Tissue Gene Expression Profiles dataset. | |
CCLE Cell Line Gene CNV Profiles | cell lines with high or low copy number of SMURF1 gene relative to other cell lines from the CCLE Cell Line Gene CNV Profiles dataset. | |
CCLE Cell Line Gene Expression Profiles | cell lines with high or low expression of SMURF1 gene relative to other cell lines from the CCLE Cell Line Gene Expression Profiles dataset. | |
CCLE Cell Line Gene Mutation Profiles | cell lines with SMURF1 gene mutations from the CCLE Cell Line Gene Mutation Profiles dataset. | |
CCLE Cell Line Proteomics | Cell lines associated with SMURF1 protein from the CCLE Cell Line Proteomics dataset. | |
ChEA Transcription Factor Binding Site Profiles | transcription factor binding site profiles with transcription factor binding evidence at the promoter of SMURF1 gene from the CHEA Transcription Factor Binding Site Profiles dataset. | |
ChEA Transcription Factor Targets | transcription factors binding the promoter of SMURF1 gene in low- or high-throughput transcription factor functional studies from the CHEA Transcription Factor Targets dataset. | |
ChEA Transcription Factor Targets 2022 | transcription factors binding the promoter of SMURF1 gene in low- or high-throughput transcription factor functional studies from the CHEA Transcription Factor Targets 2022 dataset. | |
CMAP Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of SMURF1 gene from the CMAP Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
COMPARTMENTS Curated Protein Localization Evidence Scores | cellular components containing SMURF1 protein from the COMPARTMENTS Curated Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Text-mining Protein Localization Evidence Scores | cellular components co-occuring with SMURF1 protein in abstracts of biomedical publications from the COMPARTMENTS Text-mining Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Text-mining Protein Localization Evidence Scores 2025 | cellular components co-occuring with SMURF1 protein in abstracts of biomedical publications from the COMPARTMENTS Text-mining Protein Localization Evidence Scores 2025 dataset. | |
CORUM Protein Complexes | protein complexs containing SMURF1 protein from the CORUM Protein Complexes dataset. | |
COSMIC Cell Line Gene CNV Profiles | cell lines with high or low copy number of SMURF1 gene relative to other cell lines from the COSMIC Cell Line Gene CNV Profiles dataset. | |
COSMIC Cell Line Gene Mutation Profiles | cell lines with SMURF1 gene mutations from the COSMIC Cell Line Gene Mutation Profiles dataset. | |
CTD Gene-Chemical Interactions | chemicals interacting with SMURF1 gene/protein from the curated CTD Gene-Chemical Interactions dataset. | |
CTD Gene-Disease Associations | diseases associated with SMURF1 gene/protein from the curated CTD Gene-Disease Associations dataset. | |
dbGAP Gene-Trait Associations | traits associated with SMURF1 gene in GWAS and other genetic association datasets from the dbGAP Gene-Trait Associations dataset. | |
DeepCoverMOA Drug Mechanisms of Action | small molecule perturbations with high or low expression of SMURF1 protein relative to other small molecule perturbations from the DeepCoverMOA Drug Mechanisms of Action dataset. | |
DepMap CRISPR Gene Dependency | cell lines with fitness changed by SMURF1 gene knockdown relative to other cell lines from the DepMap CRISPR Gene Dependency dataset. | |
DISEASES Experimental Gene-Disease Association Evidence Scores 2025 | diseases associated with SMURF1 gene in GWAS datasets from the DISEASES Experimental Gene-Disease Assocation Evidence Scores 2025 dataset. | |
DISEASES Text-mining Gene-Disease Association Evidence Scores | diseases co-occuring with SMURF1 gene in abstracts of biomedical publications from the DISEASES Text-mining Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Text-mining Gene-Disease Association Evidence Scores 2025 | diseases co-occuring with SMURF1 gene in abstracts of biomedical publications from the DISEASES Text-mining Gene-Disease Assocation Evidence Scores 2025 dataset. | |
DisGeNET Gene-Disease Associations | diseases associated with SMURF1 gene in GWAS and other genetic association datasets from the DisGeNET Gene-Disease Associations dataset. | |
DisGeNET Gene-Phenotype Associations | phenotypes associated with SMURF1 gene in GWAS and other genetic association datasets from the DisGeNET Gene-Phenoptype Associations dataset. | |
ENCODE Histone Modification Site Profiles | histone modification site profiles with high histone modification abundance at SMURF1 gene from the ENCODE Histone Modification Site Profiles dataset. | |
ENCODE Transcription Factor Binding Site Profiles | transcription factor binding site profiles with transcription factor binding evidence at the promoter of SMURF1 gene from the ENCODE Transcription Factor Binding Site Profiles dataset. | |
ENCODE Transcription Factor Targets | transcription factors binding the promoter of SMURF1 gene in ChIP-seq datasets from the ENCODE Transcription Factor Targets dataset. | |
ESCAPE Omics Signatures of Genes and Proteins for Stem Cells | PubMedIDs of publications reporting gene signatures containing SMURF1 from the ESCAPE Omics Signatures of Genes and Proteins for Stem Cells dataset. | |
GAD Gene-Disease Associations | diseases associated with SMURF1 gene in GWAS and other genetic association datasets from the GAD Gene-Disease Associations dataset. | |
GAD High Level Gene-Disease Associations | diseases associated with SMURF1 gene in GWAS and other genetic association datasets from the GAD High Level Gene-Disease Associations dataset. | |
GDSC Cell Line Gene Expression Profiles | cell lines with high or low expression of SMURF1 gene relative to other cell lines from the GDSC Cell Line Gene Expression Profiles dataset. | |
GeneRIF Biological Term Annotations | biological terms co-occuring with SMURF1 gene in literature-supported statements describing functions of genes from the GeneRIF Biological Term Annotations dataset. | |
GeneSigDB Published Gene Signatures | PubMedIDs of publications reporting gene signatures containing SMURF1 from the GeneSigDB Published Gene Signatures dataset. | |
GEO Signatures of Differentially Expressed Genes for Diseases | disease perturbations changing expression of SMURF1 gene from the GEO Signatures of Differentially Expressed Genes for Diseases dataset. | |
GEO Signatures of Differentially Expressed Genes for Gene Perturbations | gene perturbations changing expression of SMURF1 gene from the GEO Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Kinase Perturbations | kinase perturbations changing expression of SMURF1 gene from the GEO Signatures of Differentially Expressed Genes for Kinase Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of SMURF1 gene from the GEO Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
GEO Signatures of Differentially Expressed Genes for Transcription Factor Perturbations | transcription factor perturbations changing expression of SMURF1 gene from the GEO Signatures of Differentially Expressed Genes for Transcription Factor Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Viral Infections | virus perturbations changing expression of SMURF1 gene from the GEO Signatures of Differentially Expressed Genes for Viral Infections dataset. | |
GO Biological Process Annotations 2015 | biological processes involving SMURF1 gene from the curated GO Biological Process Annotations 2015 dataset. | |
GO Biological Process Annotations 2023 | biological processes involving SMURF1 gene from the curated GO Biological Process Annotations 2023 dataset. | |
GO Biological Process Annotations 2025 | biological processes involving SMURF1 gene from the curated GO Biological Process Annotations2025 dataset. | |
GO Cellular Component Annotations 2015 | cellular components containing SMURF1 protein from the curated GO Cellular Component Annotations 2015 dataset. | |
GO Molecular Function Annotations 2015 | molecular functions performed by SMURF1 gene from the curated GO Molecular Function Annotations 2015 dataset. | |
GO Molecular Function Annotations 2023 | molecular functions performed by SMURF1 gene from the curated GO Molecular Function Annotations 2023 dataset. | |
GO Molecular Function Annotations 2025 | molecular functions performed by SMURF1 gene from the curated GO Molecular Function Annotations 2025 dataset. | |
GTEx eQTL 2025 | SNPs regulating expression of SMURF1 gene from the GTEx eQTL 2025 dataset. | |
GTEx Tissue Gene Expression Profiles | tissues with high or low expression of SMURF1 gene relative to other tissues from the GTEx Tissue Gene Expression Profiles dataset. | |
GTEx Tissue Gene Expression Profiles 2023 | tissues with high or low expression of SMURF1 gene relative to other tissues from the GTEx Tissue Gene Expression Profiles 2023 dataset. | |
GTEx Tissue Sample Gene Expression Profiles | tissue samples with high or low expression of SMURF1 gene relative to other tissue samples from the GTEx Tissue Sample Gene Expression Profiles dataset. | |
GWASdb SNP-Disease Associations | diseases associated with SMURF1 gene in GWAS and other genetic association datasets from the GWASdb SNP-Disease Associations dataset. | |
GWASdb SNP-Phenotype Associations | phenotypes associated with SMURF1 gene in GWAS datasets from the GWASdb SNP-Phenotype Associations dataset. | |
Heiser et al., PNAS, 2011 Cell Line Gene Expression Profiles | cell lines with high or low expression of SMURF1 gene relative to other cell lines from the Heiser et al., PNAS, 2011 Cell Line Gene Expression Profiles dataset. | |
HMDB Metabolites of Enzymes | interacting metabolites for SMURF1 protein from the curated HMDB Metabolites of Enzymes dataset. | |
HPA Cell Line Gene Expression Profiles | cell lines with high or low expression of SMURF1 gene relative to other cell lines from the HPA Cell Line Gene Expression Profiles dataset. | |
HPA Tissue Gene Expression Profiles | tissues with high or low expression of SMURF1 gene relative to other tissues from the HPA Tissue Gene Expression Profiles dataset. | |
HPA Tissue Protein Expression Profiles | tissues with high or low expression of SMURF1 protein relative to other tissues from the HPA Tissue Protein Expression Profiles dataset. | |
HPA Tissue Sample Gene Expression Profiles | tissue samples with high or low expression of SMURF1 gene relative to other tissue samples from the HPA Tissue Sample Gene Expression Profiles dataset. | |
Hub Proteins Protein-Protein Interactions | interacting hub proteins for SMURF1 from the curated Hub Proteins Protein-Protein Interactions dataset. | |
HuGE Navigator Gene-Phenotype Associations | phenotypes associated with SMURF1 gene by text-mining GWAS publications from the HuGE Navigator Gene-Phenotype Associations dataset. | |
IMPC Knockout Mouse Phenotypes | phenotypes of mice caused by SMURF1 gene knockout from the IMPC Knockout Mouse Phenotypes dataset. | |
InterPro Predicted Protein Domain Annotations | protein domains predicted for SMURF1 protein from the InterPro Predicted Protein Domain Annotations dataset. | |
JASPAR Predicted Transcription Factor Targets | transcription factors regulating expression of SMURF1 gene predicted using known transcription factor binding site motifs from the JASPAR Predicted Transcription Factor Targets dataset. | |
KEA Substrates of Kinases | kinases that phosphorylate SMURF1 protein from the curated KEA Substrates of Kinases dataset. | |
KEGG Pathways | pathways involving SMURF1 protein from the KEGG Pathways dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene CNV Profiles | cell lines with high or low copy number of SMURF1 gene relative to other cell lines from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene CNV Profiles dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Expression Profiles | cell lines with high or low expression of SMURF1 gene relative to other cell lines from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Expression Profiles dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Mutation Profiles | cell lines with SMURF1 gene mutations from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Mutation Profiles dataset. | |
LINCS L1000 CMAP Chemical Perturbation Consensus Signatures | small molecule perturbations changing expression of SMURF1 gene from the LINCS L1000 CMAP Chemical Perturbations Consensus Signatures dataset. | |
LINCS L1000 CMAP CRISPR Knockout Consensus Signatures | gene perturbations changing expression of SMURF1 gene from the LINCS L1000 CMAP CRISPR Knockout Consensus Signatures dataset. | |
LINCS L1000 CMAP Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of SMURF1 gene from the LINCS L1000 CMAP Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
LOCATE Curated Protein Localization Annotations | cellular components containing SMURF1 protein in low- or high-throughput protein localization assays from the LOCATE Curated Protein Localization Annotations dataset. | |
LOCATE Predicted Protein Localization Annotations | cellular components predicted to contain SMURF1 protein from the LOCATE Predicted Protein Localization Annotations dataset. | |
MGI Mouse Phenotype Associations 2023 | phenotypes of transgenic mice caused by SMURF1 gene mutations from the MGI Mouse Phenotype Associations 2023 dataset. | |
MiRTarBase microRNA Targets | microRNAs targeting SMURF1 gene in low- or high-throughput microRNA targeting studies from the MiRTarBase microRNA Targets dataset. | |
MPO Gene-Phenotype Associations | phenotypes of transgenic mice caused by SMURF1 gene mutations from the MPO Gene-Phenotype Associations dataset. | |
MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations | gene perturbations changing expression of SMURF1 gene from the MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations dataset. | |
NIBR DRUG-seq U2OS MoA Box Gene Expression Profiles | drug perturbations changing expression of SMURF1 gene from the NIBR DRUG-seq U2OS MoA Box dataset. | |
NURSA Protein Complexes | protein complexs containing SMURF1 protein recovered by IP-MS from the NURSA Protein Complexes dataset. | |
PANTHER Pathways | pathways involving SMURF1 protein from the PANTHER Pathways dataset. | |
Pathway Commons Protein-Protein Interactions | interacting proteins for SMURF1 from the Pathway Commons Protein-Protein Interactions dataset. | |
PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations | gene perturbations changing expression of SMURF1 gene from the PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
PerturbAtlas Signatures of Differentially Expressed Genes for Mouse Gene Perturbations | gene perturbations changing expression of SMURF1 gene from the PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
PFOCR Pathway Figure Associations 2023 | pathways involving SMURF1 protein from the PFOCR Pathway Figure Associations 2023 dataset. | |
PFOCR Pathway Figure Associations 2024 | pathways involving SMURF1 protein from the Wikipathways PFOCR 2024 dataset. | |
Phosphosite Textmining Biological Term Annotations | biological terms co-occuring with SMURF1 protein in abstracts of publications describing phosphosites from the Phosphosite Textmining Biological Term Annotations dataset. | |
PID Pathways | pathways involving SMURF1 protein from the PID Pathways dataset. | |
Reactome Pathways 2014 | pathways involving SMURF1 protein from the Reactome Pathways dataset. | |
Reactome Pathways 2024 | pathways involving SMURF1 protein from the Reactome Pathways 2024 dataset. | |
Replogle et al., Cell, 2022 K562 Essential Perturb-seq Gene Perturbation Signatures | gene perturbations changing expression of SMURF1 gene from the Replogle et al., Cell, 2022 K562 Essential Perturb-seq Gene Perturbation Signatures dataset. | |
Replogle et al., Cell, 2022 K562 Genome-wide Perturb-seq Gene Perturbation Signatures | gene perturbations changing expression of SMURF1 gene from the Replogle et al., Cell, 2022 K562 Genome-wide Perturb-seq Gene Perturbation Signatures dataset. | |
Replogle et al., Cell, 2022 RPE1 Essential Perturb-seq Gene Perturbation Signatures | gene perturbations changing expression of SMURF1 gene from the Replogle et al., Cell, 2022 RPE1 Essential Perturb-seq Gene Perturbation Signatures dataset. | |
Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles | cell types and tissues with high or low DNA methylation of SMURF1 gene relative to other cell types and tissues from the Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles dataset. | |
Roadmap Epigenomics Cell and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of SMURF1 gene relative to other cell types and tissues from the Roadmap Epigenomics Cell and Tissue Gene Expression Profiles dataset. | |
Roadmap Epigenomics Histone Modification Site Profiles | histone modification site profiles with high histone modification abundance at SMURF1 gene from the Roadmap Epigenomics Histone Modification Site Profiles dataset. | |
RummaGEO Drug Perturbation Signatures | drug perturbations changing expression of SMURF1 gene from the RummaGEO Drug Perturbation Signatures dataset. | |
RummaGEO Gene Perturbation Signatures | gene perturbations changing expression of SMURF1 gene from the RummaGEO Gene Perturbation Signatures dataset. | |
Sanger Dependency Map Cancer Cell Line Proteomics | cell lines associated with SMURF1 protein from the Sanger Dependency Map Cancer Cell Line Proteomics dataset. | |
TargetScan Predicted Conserved microRNA Targets | microRNAs regulating expression of SMURF1 gene predicted using conserved miRNA seed sequences from the TargetScan Predicted Conserved microRNA Targets dataset. | |
TargetScan Predicted Nonconserved microRNA Targets | microRNAs regulating expression of SMURF1 gene predicted using nonconserved miRNA seed sequences from the TargetScan Predicted Nonconserved microRNA Targets dataset. | |
TCGA Signatures of Differentially Expressed Genes for Tumors | tissue samples with high or low expression of SMURF1 gene relative to other tissue samples from the TCGA Signatures of Differentially Expressed Genes for Tumors dataset. | |
TISSUES Curated Tissue Protein Expression Evidence Scores | tissues with high expression of SMURF1 protein from the TISSUES Curated Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Curated Tissue Protein Expression Evidence Scores 2025 | tissues with high expression of SMURF1 protein from the TISSUES Curated Tissue Protein Expression Evidence Scores 2025 dataset. | |
TISSUES Experimental Tissue Protein Expression Evidence Scores | tissues with high expression of SMURF1 protein in proteomics datasets from the TISSUES Experimental Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Experimental Tissue Protein Expression Evidence Scores 2025 | tissues with high expression of SMURF1 protein in proteomics datasets from the TISSUES Experimental Tissue Protein Expression Evidence Scores 2025 dataset. | |
TISSUES Text-mining Tissue Protein Expression Evidence Scores | tissues co-occuring with SMURF1 protein in abstracts of biomedical publications from the TISSUES Text-mining Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Text-mining Tissue Protein Expression Evidence Scores 2025 | tissues co-occuring with SMURF1 protein in abstracts of biomedical publications from the TISSUES Text-mining Tissue Protein Expression Evidence Scores 2025 dataset. | |
WikiPathways Pathways 2014 | pathways involving SMURF1 protein from the Wikipathways Pathways 2014 dataset. | |
WikiPathways Pathways 2024 | pathways involving SMURF1 protein from the WikiPathways Pathways 2024 dataset. | |