HGNC Family | Fibronectin type III domain containing, Phosphatases, Immunoglobulin superfamily domain containing |
Name | protein tyrosine phosphatase, receptor type, D |
Description | The protein encoded by this gene is a member of the protein tyrosine phosphatase (PTP) family. PTPs are known to be signaling molecules that regulate a variety of cellular processes including cell growth, differentiation, mitotic cycle, and oncogenic transformation. This PTP contains an extracellular region, a single transmembrane segment and two tandem intracytoplasmic catalytic domains, and thus represents a receptor-type PTP. The extracellular region of this protein is composed of three Ig-like and eight fibronectin type III-like domains. Studies of the similar genes in chicken and fly suggest the role of this PTP is in promoting neurite growth, and regulating neurons axon guidance. Multiple alternatively spliced transcript variants of this gene have been reported. A related pseudogene has been identified on chromosome 5. [provided by RefSeq, Jan 2010] |
Summary |
{"type": "root", "children": [{"type": "p", "children": [{"type": "t", "text": "\nMultiple genetic studies have implicated PTPRD as a pivotal tumor‐suppressor gene whose loss or inactivation contributes to tumor initiation and progression in diverse cancers. In several malignancies – including glioblastoma, lung, head and neck, cutaneous squamous, renal, gastric, lymphoma, neuroblastoma, nasopharyngeal carcinoma, and even melanoma – PTPRD is frequently inactivated by focal deletions, point mutations, aberrant splicing events or epigenetic silencing. Loss‐of‐function of PTPRD deregulates signaling cascades (for example, via failure to dephosphorylate STAT3), resulting in enhanced cell growth, invasion and altered tumor spectrum. These findings suggest that even partial loss of PTPRD (haploinsufficiency) or its cooperative deletion with other tumor suppressors (for example, CDKN2A/p16) accelerates oncogenesis and metastasis, underscoring its critical role in maintaining cell‐cycle and adhesion homeostasis."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "1", "end_ref": "20"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nBeyond its established tumor‐suppressor properties, PTPRD plays critical roles in the nervous system. Several genome‐wide association and functional studies have linked variants in PTPRD to neurodevelopmental and neuropsychiatric conditions – including restless legs syndrome, pediatric asthma with neuroimmune involvement, opioid dependence and tau pathology in Alzheimer’s disease – as well as to synaptogenesis. In neuronal contexts, PTPRD directly interacts with key trans‐synaptic adhesion molecules such as NGL‐3, SALM5 and IL1RAPL1, thereby regulating presynaptic differentiation and dendritic spine formation. These interactions not only highlight a role in proper neural circuit formation but also suggest that dysregulation of PTPRD function may contribute to cognitive impairment and other neurobehavioral disorders."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "21", "end_ref": "30"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nGenetic studies have further revealed an emerging role for PTPRD in metabolic regulation. Genome‐wide association analyses in different populations have associated variations in or near PTPRD with type 2 diabetes susceptibility, altered insulin signaling and differences in therapeutic responses (for example, to pioglitazone). Moreover, altered PTPRD expression – including aberrant DNA methylation patterns – has been linked to impaired adipogenesis and related metabolic dysfunction, and preliminary evidence even suggests a potential contribution of PTPRD to non‐alcoholic fatty liver disease. These metabolic associations point to a multifaceted contribution of PTPRD in both energy homeostasis and disease susceptibility."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "31", "end_ref": "35"}]}, {"type": "t", "text": "\n"}]}, {"type": "rg", "children": [{"type": "r", "ref": 1, "children": [{"type": "t", "text": "Masamitsu Sato, Kenji Takahashi, Kazuhiro Nagayama, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Identification of chromosome arm 9p as the most frequent target of homozygous deletions in lung cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Genes Chromosomes Cancer (2005)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/gcc.20253"}], "href": "https://doi.org/10.1002/gcc.20253"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "16114034"}], "href": "https://pubmed.ncbi.nlm.nih.gov/16114034"}]}, {"type": "r", "ref": 2, "children": [{"type": "t", "text": "Karin J Purdie, Sally R Lambert, Muy-Teck Teh, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Allelic imbalances and microdeletions affecting the PTPRD gene in cutaneous squamous cell carcinomas detected using single nucleotide polymorphism microarray analysis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Genes Chromosomes Cancer (2007)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/gcc.20447"}], "href": "https://doi.org/10.1002/gcc.20447"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "17420988"}], "href": "https://pubmed.ncbi.nlm.nih.gov/17420988"}]}, {"type": "r", "ref": 3, "children": [{"type": "t", "text": "Prakash Nair, Katleen De Preter, Jo Vandesompele, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Aberrant splicing of the PTPRD gene mimics microdeletions identified at this locus in neuroblastomas."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Genes Chromosomes Cancer (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/gcc.20521"}], "href": "https://doi.org/10.1002/gcc.20521"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18050303"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18050303"}]}, {"type": "r", "ref": 4, "children": [{"type": "t", "text": "Selvaraju Veeriah, Cameron Brennan, Shasha Meng, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The tyrosine phosphatase PTPRD is a tumor suppressor that is frequently inactivated and mutated in glioblastoma and other human cancers."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Proc Natl Acad Sci U S A (2009)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1073/pnas.0900571106"}], "href": "https://doi.org/10.1073/pnas.0900571106"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19478061"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19478061"}]}, {"type": "r", "ref": 5, "children": [{"type": "t", "text": "Maciej Giefing, Natalia Zemke, Damian Brauze, et al. "}, {"type": "b", "children": [{"type": "t", "text": "High resolution ArrayCGH and expression profiling identifies PTPRD and PCDH17/PCH68 as tumor suppressor gene candidates in laryngeal squamous cell carcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Genes Chromosomes Cancer (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/gcc.20840"}], "href": "https://doi.org/10.1002/gcc.20840"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21213369"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21213369"}]}, {"type": "r", "ref": 6, "children": [{"type": "t", "text": "Sally R Lambert, Catherine A Harwood, Karin J Purdie, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Metastatic cutaneous squamous cell carcinoma shows frequent deletion in the protein tyrosine phosphatase receptor Type D gene."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Int J Cancer (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/ijc.27333"}], "href": "https://doi.org/10.1002/ijc.27333"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22052591"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22052591"}]}, {"type": "r", "ref": 7, "children": [{"type": "t", "text": "Maria Meehan, Laavanya Parthasarathi, Niamh Moran, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Protein tyrosine phosphatase receptor delta acts as a neuroblastoma tumor suppressor by destabilizing the aurora kinase A oncogene."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Cancer (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1186/1476-4598-11-6"}], "href": "https://doi.org/10.1186/1476-4598-11-6"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22305495"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22305495"}]}, {"type": "r", "ref": 8, "children": [{"type": "t", "text": "Yan Du, Tong Su, Xiaojie Tan, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Polymorphism in protein tyrosine phosphatase receptor delta is associated with the risk of clear cell renal cell carcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Gene (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.gene.2012.09.094"}], "href": "https://doi.org/10.1016/j.gene.2012.09.094"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23069849"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23069849"}]}, {"type": "r", "ref": 9, "children": [{"type": "t", "text": "Berenice Ortiz, Armida W M Fabius, Wei H Wu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Loss of the tyrosine phosphatase PTPRD leads to aberrant STAT3 activation and promotes gliomagenesis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Proc Natl Acad Sci U S A (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1073/pnas.1401952111"}], "href": "https://doi.org/10.1073/pnas.1401952111"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24843164"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24843164"}]}, {"type": "r", "ref": 10, "children": [{"type": "t", "text": "Vijay Walia, Todd D Prickett, Jung-Sik Kim, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Mutational and functional analysis of the tumor-suppressor PTPRD in human melanoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Hum Mutat (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/humu.22630"}], "href": "https://doi.org/10.1002/humu.22630"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25113440"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25113440"}]}, {"type": "r", "ref": 11, "children": [{"type": "t", "text": "Fauzia Chaudhary, Robert Lucito, Nicholas K Tonks "}, {"type": "b", "children": [{"type": "t", "text": "Missing-in-Metastasis regulates cell motility and invasion via PTPδ-mediated changes in SRC activity."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Biochem J (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1042/BJ20140573"}], "href": "https://doi.org/10.1042/BJ20140573"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25287652"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25287652"}]}, {"type": "r", "ref": 12, "children": [{"type": "t", "text": "Dandan Wang, Leilei Wang, Jun Zhou, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Reduced expression of PTPRD correlates with poor prognosis in gastric adenocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS One (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pone.0113754"}], "href": "https://doi.org/10.1371/journal.pone.0113754"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25412184"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25412184"}]}, {"type": "r", "ref": 13, "children": [{"type": "t", "text": "Noah D Peyser, Yu Du, Hua Li, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Loss-of-Function PTPRD Mutations Lead to Increased STAT3 Activation and Sensitivity to STAT3 Inhibition in Head and Neck Cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS One (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pone.0135750"}], "href": "https://doi.org/10.1371/journal.pone.0135750"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "26267899"}], "href": "https://pubmed.ncbi.nlm.nih.gov/26267899"}]}, {"type": "r", "ref": 14, "children": [{"type": "t", "text": "Ikenna C Eze, Medea Imboden, Ashish Kumar, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Air pollution and diabetes association: Modification by type 2 diabetes genetic risk score."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Environ Int (2016)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.envint.2016.04.032"}], "href": "https://doi.org/10.1016/j.envint.2016.04.032"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "27281273"}], "href": "https://pubmed.ncbi.nlm.nih.gov/27281273"}]}, {"type": "r", "ref": 15, "children": [{"type": "t", "text": "Valeria Spina, Hossein Khiabanian, Monica Messina, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The genetics of nodal marginal zone lymphoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Blood (2016)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1182/blood-2016-02-696757"}], "href": "https://doi.org/10.1182/blood-2016-02-696757"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "27335277"}], "href": "https://pubmed.ncbi.nlm.nih.gov/27335277"}]}, {"type": "r", "ref": 16, "children": [{"type": "t", "text": "Worapong Singchat, Ekarat Hitakomate, Budsaba Rerkarmnuaychoke, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Genomic Alteration in Head and Neck Squamous Cell Carcinoma (HNSCC) Cell Lines Inferred from Karyotyping, Molecular Cytogenetics, and Array Comparative Genomic Hybridization."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS One (2016)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pone.0160901"}], "href": "https://doi.org/10.1371/journal.pone.0160901"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "27501229"}], "href": "https://pubmed.ncbi.nlm.nih.gov/27501229"}]}, {"type": "r", "ref": 17, "children": [{"type": "t", "text": "Jodie N Painter, Tracy A O'Mara, Andrew P Morris, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Genetic overlap between endometriosis and endometrial cancer: evidence from cross-disease genetic correlation and GWAS meta-analyses."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Med (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/cam4.1445"}], "href": "https://doi.org/10.1002/cam4.1445"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "29608257"}], "href": "https://pubmed.ncbi.nlm.nih.gov/29608257"}]}, {"type": "r", "ref": 18, "children": [{"type": "t", "text": "Won Jung Bae, Ji Mi Ahn, Hye Eun Byeon, et al. "}, {"type": "b", "children": [{"type": "t", "text": "PTPRD-inactivation-induced CXCL8 promotes angiogenesis and metastasis in gastric cancer and is inhibited by metformin."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Exp Clin Cancer Res (2019)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1186/s13046-019-1469-4"}], "href": "https://doi.org/10.1186/s13046-019-1469-4"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "31805999"}], "href": "https://pubmed.ncbi.nlm.nih.gov/31805999"}]}, {"type": "r", "ref": 19, "children": [{"type": "t", "text": "Yanling Lin, Xiaohan Zhou, Kaifan Yang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Protein tyrosine phosphatase receptor type D gene promotes radiosensitivity via STAT3 dephosphorylation in nasopharyngeal carcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Oncogene (2021)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/s41388-021-01768-8"}], "href": "https://doi.org/10.1038/s41388-021-01768-8"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "33824475"}], "href": "https://pubmed.ncbi.nlm.nih.gov/33824475"}]}, {"type": "r", "ref": 20, "children": [{"type": "t", "text": "Yiting Sun, Jianchun Duan, Wenfeng Fang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Identification and validation of tissue or ctDNA PTPRD phosphatase domain deleterious mutations as prognostic and predictive biomarkers for immune checkpoint inhibitors in non-squamous NSCLC."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "BMC Med (2021)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1186/s12916-021-02075-5"}], "href": "https://doi.org/10.1186/s12916-021-02075-5"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "34615542"}], "href": "https://pubmed.ncbi.nlm.nih.gov/34615542"}]}, {"type": "r", "ref": 21, "children": [{"type": "t", "text": "Shyh-Dar Shyur, Jiu-Yao Wang, Cherry Guan-Ju Lin, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The polymorphisms of protein-tyrosine phosphatase receptor-type delta gene and its association with pediatric asthma in the Taiwanese population."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Eur J Hum Genet (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/ejhg.2008.79"}], "href": "https://doi.org/10.1038/ejhg.2008.79"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18414509"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18414509"}]}, {"type": "r", "ref": 22, "children": [{"type": "t", "text": "Barbara Schormair, David Kemlink, Darina Roeske, et al. "}, {"type": "b", "children": [{"type": "t", "text": "PTPRD (protein tyrosine phosphatase receptor type delta) is associated with restless legs syndrome."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Genet (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/ng.190"}], "href": "https://doi.org/10.1038/ng.190"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18660810"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18660810"}]}, {"type": "r", "ref": 23, "children": [{"type": "t", "text": "Seok-Kyu Kwon, Jooyeon Woo, Soo-Young Kim, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Trans-synaptic adhesions between netrin-G ligand-3 (NGL-3) and receptor tyrosine phosphatases LAR, protein-tyrosine phosphatase delta (PTPdelta), and PTPsigma via specific domains regulate excitatory synapse formation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M109.061127"}], "href": "https://doi.org/10.1074/jbc.M109.061127"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "20139422"}], "href": "https://pubmed.ncbi.nlm.nih.gov/20139422"}]}, {"type": "r", "ref": 24, "children": [{"type": "t", "text": "Qinbo Yang, Lin Li, Rong Yang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Family-based and population-based association studies validate PTPRD as a risk factor for restless legs syndrome."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mov Disord (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/mds.23459"}], "href": "https://doi.org/10.1002/mds.23459"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21264940"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21264940"}]}, {"type": "r", "ref": 25, "children": [{"type": "t", "text": "Pamela Valnegri, Chiara Montrasio, Dario Brambilla, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The X-linked intellectual disability protein IL1RAPL1 regulates excitatory synapse formation by binding PTPδ and RhoGAP2."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Hum Mol Genet (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1093/hmg/ddr418"}], "href": "https://doi.org/10.1093/hmg/ddr418"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21926414"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21926414"}]}, {"type": "r", "ref": 26, "children": [{"type": "t", "text": "Hyatt Moore, Juliane Winkelmann, Ling Lin, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Periodic leg movements during sleep are associated with polymorphisms in BTBD9, TOX3/BC034767, MEIS1, MAP2K5/SKOR1, and PTPRD."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Sleep (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.5665/sleep.4006"}], "href": "https://doi.org/10.5665/sleep.4006"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25142570"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25142570"}]}, {"type": "r", "ref": 27, "children": [{"type": "t", "text": "Dawei Li, Hongyu Zhao, Henry R Kranzler, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Genome-wide association study of copy number variations (CNVs) with opioid dependence."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Neuropsychopharmacology (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/npp.2014.290"}], "href": "https://doi.org/10.1038/npp.2014.290"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25345593"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25345593"}]}, {"type": "r", "ref": 28, "children": [{"type": "t", "text": "L B Chibnik, C C White, S Mukherjee, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Susceptibility to neurofibrillary tangles: role of the PTPRD locus and limited pleiotropy with other neuropathologies."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Psychiatry (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/mp.2017.20"}], "href": "https://doi.org/10.1038/mp.2017.20"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "28322283"}], "href": "https://pubmed.ncbi.nlm.nih.gov/28322283"}]}, {"type": "r", "ref": 29, "children": [{"type": "t", "text": "Sakurako Goto-Ito, Atsushi Yamagata, Yusuke Sato, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Structural basis of trans-synaptic interactions between PTPδ and SALMs for inducing synapse formation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Commun (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/s41467-017-02417-z"}], "href": "https://doi.org/10.1038/s41467-017-02417-z"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "29348429"}], "href": "https://pubmed.ncbi.nlm.nih.gov/29348429"}]}, {"type": "r", "ref": 30, "children": [{"type": "t", "text": "Zhaohan Lin, Jianmei Liu, Huandi Ding, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Structural basis of SALM5-induced PTPδ dimerization for synaptic differentiation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Commun (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/s41467-017-02414-2"}], "href": "https://doi.org/10.1038/s41467-017-02414-2"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "29348579"}], "href": "https://pubmed.ncbi.nlm.nih.gov/29348579"}]}, {"type": "r", "ref": 31, "children": [{"type": "t", "text": "Fuu-Jen Tsai, Chi-Fan Yang, Ching-Chu Chen, et al. "}, {"type": "b", "children": [{"type": "t", "text": "A genome-wide association study identifies susceptibility variants for type 2 diabetes in Han Chinese."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS Genet (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pgen.1000847"}], "href": "https://doi.org/10.1371/journal.pgen.1000847"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "20174558"}], "href": "https://pubmed.ncbi.nlm.nih.gov/20174558"}]}, {"type": "r", "ref": 32, "children": [{"type": "t", "text": "Yi-Cheng Chang, Yen-Feng Chiu, Pi-Hua Liu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Replication of genome-wide association signals of type 2 diabetes in Han Chinese in a prospective cohort."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Endocrinol (Oxf) (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1111/j.1365-2265.2011.04175.x"}], "href": "https://doi.org/10.1111/j.1365-2265.2011.04175.x"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21767287"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21767287"}]}, {"type": "r", "ref": 33, "children": [{"type": "t", "text": "Qi Pei, Qiong Huang, Guo-ping Yang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "PPAR-γ2 and PTPRD gene polymorphisms influence type 2 diabetes patients' response to pioglitazone in China."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Acta Pharmacol Sin (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/aps.2012.144"}], "href": "https://doi.org/10.1038/aps.2012.144"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23147557"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23147557"}]}, {"type": "r", "ref": 34, "children": [{"type": "t", "text": "Shunsuke Nakajima, Hiroki Tanaka, Koji Sawada, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Polymorphism of receptor-type tyrosine-protein phosphatase delta gene in the development of non-alcoholic fatty liver disease."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Gastroenterol Hepatol (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1111/jgh.13820"}], "href": "https://doi.org/10.1111/jgh.13820"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "28497593"}], "href": "https://pubmed.ncbi.nlm.nih.gov/28497593"}]}, {"type": "r", "ref": 35, "children": [{"type": "t", "text": "Luca Parrillo, Rosa Spinelli, Michele Longo, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Altered "}, {"type": "a", "children": [{"type": "t", "text": "i"}], "href": "i"}, {"type": "t", "text": "PTPRD"}, {"type": "a", "children": [{"type": "t", "text": "/i"}], "href": "/i"}, {"type": "t", "text": " DNA methylation associates with restricted adipogenesis in healthy first-degree relatives of Type 2 diabetes subjects."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Epigenomics (2020)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.2217/epi-2019-0267"}], "href": "https://doi.org/10.2217/epi-2019-0267"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "32483983"}], "href": "https://pubmed.ncbi.nlm.nih.gov/32483983"}]}]}]}
|
Synonyms | RPTPDELTA, HPTPD, HPTP, PTPD, HPTPDELTA |
Proteins | PTPRD_HUMAN |
NCBI Gene ID | 5789 |
API | |
Download Associations | |
Predicted Functions |
![]() |
Co-expressed Genes |
![]() |
Expression in Tissues and Cell Lines |
![]() |
PTPRD has 8,024 functional associations with biological entities spanning 9 categories (molecular profile, organism, chemical, functional term, phrase or reference, 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 PTPRD 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 PTPRD gene relative to other tissues from the Allen Brain Atlas Adult Human 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 PTPRD 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 PTPRD 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 PTPRD gene relative to other tissues from the Allen Brain Atlas Prenatal Human Brain Tissue Gene Expression Profiles dataset. | |
BioGPS Cell Line Gene Expression Profiles | cell lines with high or low expression of PTPRD 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 PTPRD 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 PTPRD 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 PTPRD 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 PTPRD gene relative to other cell lines from the CCLE Cell Line Gene Expression Profiles dataset. | |
CCLE Cell Line Gene Mutation Profiles | cell lines with PTPRD gene mutations from the CCLE Cell Line Gene Mutation Profiles dataset. | |
CellMarker Gene-Cell Type Associations | cell types associated with PTPRD gene from the CellMarker Gene-Cell Type Associations dataset. | |
ChEA Transcription Factor Binding Site Profiles | transcription factor binding site profiles with transcription factor binding evidence at the promoter of PTPRD gene from the CHEA Transcription Factor Binding Site Profiles dataset. | |
ChEA Transcription Factor Targets | transcription factors binding the promoter of PTPRD 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 PTPRD 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 PTPRD gene from the CMAP Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
COMPARTMENTS Curated Protein Localization Evidence Scores | cellular components containing PTPRD protein from the COMPARTMENTS Curated Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Curated Protein Localization Evidence Scores 2025 | cellular components containing PTPRD protein from the COMPARTMENTS Curated Protein Localization Evidence Scores 2025 dataset. | |
COMPARTMENTS Text-mining Protein Localization Evidence Scores | cellular components co-occuring with PTPRD 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 PTPRD protein in abstracts of biomedical publications from the COMPARTMENTS Text-mining Protein Localization Evidence Scores 2025 dataset. | |
COSMIC Cell Line Gene CNV Profiles | cell lines with high or low copy number of PTPRD gene relative to other cell lines from the COSMIC Cell Line Gene CNV Profiles dataset. | |
COSMIC Cell Line Gene Mutation Profiles | cell lines with PTPRD gene mutations from the COSMIC Cell Line Gene Mutation Profiles dataset. | |
CTD Gene-Chemical Interactions | chemicals interacting with PTPRD gene/protein from the curated CTD Gene-Chemical Interactions dataset. | |
CTD Gene-Disease Associations | diseases associated with PTPRD gene/protein from the curated CTD Gene-Disease Associations dataset. | |
dbGAP Gene-Trait Associations | traits associated with PTPRD 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 PTPRD 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 PTPRD gene knockdown relative to other cell lines from the DepMap CRISPR Gene Dependency dataset. | |
DISEASES Curated Gene-Disease Association Evidence Scores | diseases involving PTPRD gene from the DISEASES Curated Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Curated Gene-Disease Association Evidence Scores 2025 | diseases involving PTPRD gene from the DISEASES Curated Gene-Disease Association Evidence Scores 2025 dataset. | |
DISEASES Experimental Gene-Disease Association Evidence Scores | diseases associated with PTPRD gene in GWAS datasets from the DISEASES Experimental Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Experimental Gene-Disease Association Evidence Scores 2025 | diseases associated with PTPRD 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 PTPRD 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 PTPRD 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 PTPRD gene in GWAS and other genetic association datasets from the DisGeNET Gene-Disease Associations dataset. | |
DisGeNET Gene-Phenotype Associations | phenotypes associated with PTPRD 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 PTPRD 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 PTPRD gene from the ENCODE Transcription Factor Binding Site Profiles dataset. | |
ENCODE Transcription Factor Targets | transcription factors binding the promoter of PTPRD 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 PTPRD from the ESCAPE Omics Signatures of Genes and Proteins for Stem Cells dataset. | |
GAD Gene-Disease Associations | diseases associated with PTPRD gene in GWAS and other genetic association datasets from the GAD Gene-Disease Associations dataset. | |
GAD High Level Gene-Disease Associations | diseases associated with PTPRD 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 PTPRD gene relative to other cell lines from the GDSC Cell Line Gene Expression Profiles dataset. | |
GeneRIF Biological Term Annotations | biological terms co-occuring with PTPRD 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 PTPRD from the GeneSigDB Published Gene Signatures dataset. | |
GEO Signatures of Differentially Expressed Genes for Diseases | disease perturbations changing expression of PTPRD 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 PTPRD 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 PTPRD 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 PTPRD 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 PTPRD 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 PTPRD gene from the GEO Signatures of Differentially Expressed Genes for Viral Infections dataset. | |
GO Biological Process Annotations 2015 | biological processes involving PTPRD gene from the curated GO Biological Process Annotations 2015 dataset. | |
GO Biological Process Annotations 2023 | biological processes involving PTPRD gene from the curated GO Biological Process Annotations 2023 dataset. | |
GO Biological Process Annotations 2025 | biological processes involving PTPRD gene from the curated GO Biological Process Annotations2025 dataset. | |
GO Cellular Component Annotations 2015 | cellular components containing PTPRD protein from the curated GO Cellular Component Annotations 2015 dataset. | |
GO Cellular Component Annotations 2023 | cellular components containing PTPRD protein from the curated GO Cellular Component Annotations 2023 dataset. | |
GO Cellular Component Annotations 2025 | cellular components containing PTPRD protein from the curated GO Cellular Component Annotations 2025 dataset. | |
GO Molecular Function Annotations 2015 | molecular functions performed by PTPRD gene from the curated GO Molecular Function Annotations 2015 dataset. | |
GO Molecular Function Annotations 2023 | molecular functions performed by PTPRD gene from the curated GO Molecular Function Annotations 2023 dataset. | |
GO Molecular Function Annotations 2025 | molecular functions performed by PTPRD gene from the curated GO Molecular Function Annotations 2025 dataset. | |
GTEx eQTL 2025 | SNPs regulating expression of PTPRD gene from the GTEx eQTL 2025 dataset. | |
GTEx Tissue Gene Expression Profiles | tissues with high or low expression of PTPRD 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 PTPRD 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 PTPRD gene relative to other tissue samples from the GTEx Tissue Sample Gene Expression Profiles dataset. | |
GWAS Catalog SNP-Phenotype Associations | phenotypes associated with PTPRD gene in GWAS datasets from the GWAS Catalog SNP-Phenotype Associations dataset. | |
GWASdb SNP-Disease Associations | diseases associated with PTPRD gene in GWAS and other genetic association datasets from the GWASdb SNP-Disease Associations dataset. | |
GWASdb SNP-Phenotype Associations | phenotypes associated with PTPRD 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 PTPRD 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 PTPRD protein from the curated HMDB Metabolites of Enzymes dataset. | |
HPA Cell Line Gene Expression Profiles | cell lines with high or low expression of PTPRD 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 PTPRD 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 PTPRD 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 PTPRD gene relative to other tissue samples from the HPA Tissue Sample Gene Expression Profiles dataset. | |
Hub Proteins Protein-Protein Interactions | interacting hub proteins for PTPRD from the curated Hub Proteins Protein-Protein Interactions dataset. | |
HuBMAP Azimuth Cell Type Annotations | cell types associated with PTPRD gene from the HuBMAP Azimuth Cell Type Annotations dataset. | |
HuGE Navigator Gene-Phenotype Associations | phenotypes associated with PTPRD gene by text-mining GWAS publications from the HuGE Navigator Gene-Phenotype Associations dataset. | |
IMPC Knockout Mouse Phenotypes | phenotypes of mice caused by PTPRD gene knockout from the IMPC Knockout Mouse Phenotypes dataset. | |
InterPro Predicted Protein Domain Annotations | protein domains predicted for PTPRD protein from the InterPro Predicted Protein Domain Annotations dataset. | |
JASPAR Predicted Transcription Factor Targets | transcription factors regulating expression of PTPRD gene predicted using known transcription factor binding site motifs from the JASPAR Predicted Transcription Factor Targets dataset. | |
Kinase Library Serine Threonine Kinome Atlas | kinases that phosphorylate PTPRD protein from the Kinase Library Serine Threonine Atlas dataset. | |
Kinase Library Tyrosine Kinome Atlas | kinases that phosphorylate PTPRD protein from the Kinase Library Tyrosine Kinome Atlas dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene CNV Profiles | cell lines with high or low copy number of PTPRD 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 Mutation Profiles | cell lines with PTPRD gene mutations from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Mutation Profiles dataset. | |
KnockTF Gene Expression Profiles with Transcription Factor Perturbations | transcription factor perturbations changing expression of PTPRD gene from the KnockTF Gene Expression Profiles with Transcription Factor Perturbations dataset. | |
LINCS L1000 CMAP Chemical Perturbation Consensus Signatures | small molecule perturbations changing expression of PTPRD gene from the LINCS L1000 CMAP Chemical Perturbations Consensus Signatures dataset. | |
LINCS L1000 CMAP CRISPR Knockout Consensus Signatures | gene perturbations changing expression of PTPRD 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 PTPRD gene from the LINCS L1000 CMAP Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
LOCATE Curated Protein Localization Annotations | cellular components containing PTPRD 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 PTPRD protein from the LOCATE Predicted Protein Localization Annotations dataset. | |
MGI Mouse Phenotype Associations 2023 | phenotypes of transgenic mice caused by PTPRD gene mutations from the MGI Mouse Phenotype Associations 2023 dataset. | |
MiRTarBase microRNA Targets | microRNAs targeting PTPRD gene in low- or high-throughput microRNA targeting studies from the MiRTarBase microRNA Targets dataset. | |
MotifMap Predicted Transcription Factor Targets | transcription factors regulating expression of PTPRD gene predicted using known transcription factor binding site motifs from the MotifMap Predicted Transcription Factor Targets dataset. | |
MoTrPAC Rat Endurance Exercise Training | tissue samples with high or low expression of PTPRD gene relative to other tissue samples from the MoTrPAC Rat Endurance Exercise Training dataset. | |
MPO Gene-Phenotype Associations | phenotypes of transgenic mice caused by PTPRD gene mutations from the MPO Gene-Phenotype Associations dataset. | |
MSigDB Cancer Gene Co-expression Modules | co-expressed genes for PTPRD from the MSigDB Cancer Gene Co-expression Modules dataset. | |
MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations | gene perturbations changing expression of PTPRD gene from the MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations dataset. | |
NURSA Protein Complexes | protein complexs containing PTPRD protein recovered by IP-MS from the NURSA Protein Complexes dataset. | |
Pathway Commons Protein-Protein Interactions | interacting proteins for PTPRD from the Pathway Commons Protein-Protein Interactions dataset. | |
PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations | gene perturbations changing expression of PTPRD 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 PTPRD gene from the PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
PFOCR Pathway Figure Associations 2023 | pathways involving PTPRD protein from the PFOCR Pathway Figure Associations 2023 dataset. | |
PFOCR Pathway Figure Associations 2024 | pathways involving PTPRD protein from the Wikipathways PFOCR 2024 dataset. | |
Reactome Pathways 2024 | pathways involving PTPRD protein from the Reactome Pathways 2024 dataset. | |
Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles | cell types and tissues with high or low DNA methylation of PTPRD 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 PTPRD 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 PTPRD gene from the Roadmap Epigenomics Histone Modification Site Profiles dataset. | |
RummaGEO Drug Perturbation Signatures | drug perturbations changing expression of PTPRD gene from the RummaGEO Drug Perturbation Signatures dataset. | |
RummaGEO Gene Perturbation Signatures | gene perturbations changing expression of PTPRD gene from the RummaGEO Gene Perturbation Signatures dataset. | |
SynGO Synaptic Gene Annotations | synaptic terms associated with PTPRD gene from the SynGO Synaptic Gene Annotations dataset. | |
Tabula Sapiens Gene-Cell Associations | cell types with high or low expression of PTPRD gene relative to other cell types from the Tabula Sapiens Gene-Cell Associations dataset. | |
TargetScan Predicted Conserved microRNA Targets | microRNAs regulating expression of PTPRD gene predicted using conserved miRNA seed sequences from the TargetScan Predicted Conserved microRNA Targets dataset. | |
TargetScan Predicted Nonconserved microRNA Targets | microRNAs regulating expression of PTPRD 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 PTPRD 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 PTPRD protein from the TISSUES Curated Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Curated Tissue Protein Expression Evidence Scores 2025 | tissues with high expression of PTPRD protein from the TISSUES Curated Tissue Protein Expression Evidence Scores 2025 dataset. | |
TISSUES Experimental Tissue Protein Expression Evidence Scores | tissues with high expression of PTPRD 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 PTPRD 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 PTPRD 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 PTPRD protein in abstracts of biomedical publications from the TISSUES Text-mining Tissue Protein Expression Evidence Scores 2025 dataset. | |
WikiPathways Pathways 2024 | pathways involving PTPRD protein from the WikiPathways Pathways 2024 dataset. | |