TPD52 (tumor protein D52)

2010-09-01   Austin Della-Franca , Jennifer Byrne 

Childrens Cancer Research Unit, Kids Research Institute, The Childrenis Hospital at Westmead, Sydney, Australia

Identity

HGNC
LOCATION
8q21.13
LOCUSID
ALIAS
D52,N8L,PC-1,PrLZ,hD52
FUSION GENES

DNA/RNA

Description

The TPD52 gene is found on chromosome 8, at location 81109658-81246391 bp. It is composed of at least 9 exons spanning a genomic region of 138.88 kb and the open reading frame of the coding region is 555 bp.

Transcription

Experimentally, an increase in TPD52 protein or mRNA transcript levels has been demonstrated or predicted following a number of different types of stimuli, such as hormone receptor activation(using dihydrotestosterone (DePrimo et al., 2002), estradiol (Byrne et al., 1996) and the synthetic androgens R1881 (DePrimo et al., 2002; Nelson et al., 2002; Rubin et al., 2004) and methyltrienolone (Wang et al., 2004)), immune receptor activation (toll-like receptor 4 (TLR-4) using bacterial Lipopolysaccharide (LPS) and triggering receptors expressed on myeloid cells 1 (TREM-1) using anti-TREM-1 cross-linking antibody (Dower et al., 2008)), Erbb2 over-expression (Landis et al., 2005), Platelet-Derived Growth Factor (PDGF) signalling (Ma et al., 2005) and BRCA1 tumor suppressor (containing a single amino acid substitution specifically at Ser1841Asn) expression (Crugliano et al., 2007). A decrease in TPD52 transcript levels has been observed or predicted following treatment with the differentiating agent 12-O-tetradecanoylphorbol-13-acetate (TPA) (Byrne et al., 1996), myc onco-protein overexpression (Guo et al., 2000) and MyoD gene knockdown (Asakura et al., 2007).

Pseudogene

No human pseudogene for TPD52 has been identified (see Pseudogene In Family).

Proteins

Description

The TPD52 gene encodes multiple proteins, predominantly TPD52 (184 aa, isoform 3, accession no. NP_005070.1) and PrLZ/PC-1 (224 aa, isoform 1, accession no. NP_001020423.1). Both are encoded by 6 exons, with the first exon being unique to each isoform. They share a coiled-coil motif located towards the N-terminus but possess alternate N-terminal domains. Since TPD52 is more ubiquitously expressed than PrLZ (for TPD52 expression, see Byrne et al., 1995 and Chen et al., 1996; for PrLZ expression, see Wang et al., 2004), TPD52 will be the primary focus of this summary.
TPD52 is a 184-amino acid polypeptide with a predicted molecular mass of 19.8 kDa. The protein is indicated to be largely hydrophilic, has a calculated isoelectric point of 4.75, and contains an identified phosphorylation site at Serine 136 (Chew et al., 2008; Thomas et al., 2010).
Using glutaraldehyde cross-linking, Chen et al. (1997) showed that TPD52 is able to bind itself (also shown by Byrne et al., 1998; Sathasivam et al., 2001, and Thomas et al., 2001). Byrne et al. (1998) also showed that TPD52 binds the related TPD52L1 and TPD52L2 proteins. These authors proposed that TPD52 may exert and/or regulate its activities through interaction with itself and its related proteins and that the coiled-coil motif predicted within the TPD52 protein is necessary for interactions (Byrne et al., 1998). A subsequent analysis indicated that C-terminal regions may also facilitate and/or stabilise these interactions (Sathasivam et al., 2001).
The first heterologous binding partner identified for TPD52 was the proteolipid MAL2 (Wilson et al., 2001), which is similar to MAL, an integral component of the apical transport machinery in polarised cell types (Martin-Belmonte et al., 1998; Puertollano et al., 1999; Cheong et al., 1999). MAL2 was shown to be a binding partner for TPD52 using the yeast two-hybrid system (Wilson et al., 2001) and more recently to co-immunoprecipitate with TPD52 using Myc-MAL2 overexpressing MCF-10A breast cancer cells (Fanayan et al., 2009).
TPD52 also binds Annexin VI, which is involved in membrane trafficking, and this binding is Ca2+- dependent (Thomas et al., 2002; Tiacci et al., 2005). TPD52 (along with Tip47, Rab5, Rab6 and Rab9) co-immunoprecipitated with the bacterial product ExoS, as shown by a study which examined the transport of ExoS from plasma membrane to perinuclear regions (Zhang et al., 2007a).

Expression

Early studies using Northern blot analysis undertaken by Byrne et al. (1995) showed strong expression of TPD52 transcripts in colon and kidney tissues. Chen et al. (1996) used the same technique to identify comparatively high TPD52 transcript levels in kidney, prostate, small intestine and kidney, as well as moderate levels in brain, liver, placenta, pancreas epithelia, and low levels in heart, lung, ovary, peripheral blood leukocyte, skeletal muscle, spleen, testis, and thymus tissues. Chen et al. (1997) followed up with immunohistochemistry and multiple tissue western blot analysis indicating strong detection of TPD52 protein in colon and kidney, moderate levels in brain and liver tissue, and low levels in heart, lung and skeletal muscle. Groblewski et al. (1999) used western blotting and immunofluorescence studies to identify high levels of TPD52 protein in colon epithelia and small intestine epithelia, as well as moderate expression in the lacrimal gland, pancreas, parotid gland, stomach epithelia and submandibular gland. More recently, Wang et al. (2004) identified high levels of the PrLZ transcript in prostate using a multiple tissue expression array, whilst Tiacci et al. (2005) found high levels of TPD52 protein via immunofluorescence in B cells (including plasma cells) and colon tissue, and moderate levels in stomach and pancreas epithelia.

Localisation

TPD52 is partitioned between soluble and insoluble cellular protein fractions (Groblewski et al., 1999; Balleine et al., 2000), suggesting that TPD52 may have multiple subcellular locations. TPD52 is abundant around secretory granules in the apical cytoplasm of epithelial cells of exocrine glands, gastrointestinal tissues and in cultured mucosal T84 cells of the rat (Groblewski et al., 1999; Kaspar et al., 2003a). Kaspar et al. (2003a) also showed that TPD52 exists in perinuclear regions of T84 cells, whilst Thomas et al. (2004) showed that TPD52 exists within or around both endocytic and exocytic compartments within rat pancreatic acinar cells. Using breast cancer samples, immunohistochemical analysis by Balleine et al. (2000) revealed TPD52 staining within the cytoplasm and less frequently in the perinucleus, whilst benign epithelial elements displayed little or no staining. Myeloma cells also strongly expressed TPD52 in the cytoplasm (Tiacci et al., 2005).

Function

At a subcellular level, TPD52 appears to be involved in plasma membrane-based exocytic (Thomas et al., 2001; Kaspar et al., 2003a; Kaspar et al., 2003b; Chew et al., 2008) and endocytic functions (Thomas et al., 2004), and trafficking within the exo- and endocytic pathways (Thomas et al., 2002; Thomas et al., 2004). At a cellular level, TPD52 has been shown to promote proliferation (Lewis et al., 2007; Zhang et al., 2007b; Shehata et al., 2008a; Li et al., 2009) and tissue invasion (Lewis et al., 2007), as well as B-cell maturation (Tiacci et al., 2005). However, a clear link between TPD52s subcellular and cellular influences has yet to be established.
TPD52 was initially predicted to function as a calcium-sensitive signalling molecule, since rabbit TPD52 is phosphorylated in gastric parietal cells upon cholinergic stimulation (Parente et al., 1996). At least two phosphorylation events for human and rat TPD52 have been demonstrated (Groblewski et al., 1996; Kaspar et al., 2003a; 2003b) and more are predicted (Olsen et al., 2006; Molina et al., 2007). Using mass spectrometry and site-directed mutagenesis, Chew et al. (2008) showed that the calcium/calmodulin-dependent phosphorylation of TPD52 on serine residue 136 may be mediated by CAMK2delta6. Significantly, the time-course of such phosphorylation was found to correlate with exocytosis, suggesting that TPD52 may promote exocytosis following phosphorylation. Thomas et al. (2010) recently showed that this phosphorylation of TPD52 caused an increase of LAMP-1 exocytosis from CHO-K1 cells. This had earlier been suggested in studies involving stimulation of pancreatic (using cholecystokinin-octapeptide) and colonic (using carbachol) secretion in rat acinar and mucosal T-84 cells, respectively (Kaspar et al., 2003a; 2003b). On par with this, amylase was shown to be released from rat pancreatic acinar cells in a calcium-dependent manner, following the introduction of recombinant TPD52 (Thomas et al., 2001). Furthermore, TGF-beta1 was found to be secreted from stably transfected mouse Tpd52-expressing 3T3 fibroblasts (Lewis et al., 2007). However, it has not been determined if TPD52 is directly involved with the above-mentioned release of TGF-beta1 or if this is perhaps a downstream consequence of TPD52 expression.
TPD52 binds Annexin VI (Thomas et al., 2002; Tiacci et al., 2005) and MAL2 (Wilson et al., 2001) which have been implicated in clathrin-mediated endocytosis (Kamal et al., 1998; Thomas et al., 2002) and indirect apical transcytosis (de Marco et al., 2002), respectively. Using CCK-8 treatment in pancreatic acinar cells, Thomas et al. (2004) showed TPD52 to localise to early endosomes and the limiting membrane of zymogen granules, suggesting that TPD52 may play a role in protein and phospholipid distribution within the secretory pathway. They also recently examined LAMP-1 trafficking and AP-3/TPD52 co-localisation in CHO-K1 cells which suggested TPD52 may participate in lysosome-like vesicle formation (post-golgi) as well as endocytic retrieval (Thomas et al., 2010). TPD52s potential endocytic involvement was also demonstrated through its co-immunoprecipitation with the bacterial product ExoS (along with Tip47, Rab5, Rab6 and Rab9), perhaps enabling the transport of the bacterial product ExoS from endosome to golgi and perinuclear regions (Zhang et al., 2007a). TPD52 also binds family member TPD52L1, which itself has been implicated in the regulation of SNARE complexes on early endosomes (Proux-Gillardeaux et al., 2003).
Exogenous expression of TPD52 isoforms in various cell lines has produced increases in anchorage-independent colony formation (Lewis et al., 2007; Zhang et al., 2007b; Shehata et al., 2008a), proliferation (Lewis et al., 2007; Zhang et al., 2007b; Shehata et al., 2008a; Li et al., 2009), metastatic ability post-inoculation (Lewis et al., 2007), tumor volume post-inoculation (Zhang et al., 2007b; Li et al., 2009) and migration/invasion rate (Li et al., 2009). Also, a decrease in TPD52 level following siRNA treatment caused an increase in apoptosis in the SK-BR-3 breast cancer cell line (Shehata et al., 2008a). Zhang et al. (2007b) showed that stable transfection of TPD52 in LNCaP and its derivative androgen-independent C4-2 prostate cancer cell lines results in the phosphorylation of signalling intermediates that are also implicated in producing the above-mentioned phenotypes, namely AKT, Raf and GSK-3beta. The increased phosphorylation of AKT has since been confirmed by Ummanni et al. (2008), who also examined the LNCaP cell line.

Homology

Subsequent to the characterisation of TPD52 in 1995 (Byrne et al., 1995), there have been three related genes described. These are TPD52L1 (D53) (Byrne et al., 1996), TPD52L2 (D54) (Nourse et al., 1998) and NYD-SP25/TPD52L3 (D55) (Cao et al., 2006). TPD52-like genes share about 50% nucleotide sequence homology, and are not similar to proteins of known function in any species (Nourse et al., 1998). TPD52 and TPD52-like genes appear to have evolved from ancestral TPD52-like sequences such as those found in Drosophila melanogaster and Caenorhabditis elegans through gene duplication events (Boutros et al., 2004).

Implicated in

Entity name
Various cancers
Note
Association to diseases (breast neoplasms; carcinoma, basal cell; neoplasms; ovarian neoplasms; prostatic neoplasms) and proposed to participate in processes (anatomical structure morphogenesis, B cell differentiation, secretion).
Disease
In situ hybridisation and Northern blot studies first revealed TPD52 overexpression in breast cancer (Byrne et al., 1995). Chen et al. (1996, 1997) also found TPD52 mRNA/protein to be expressed in breast cancer derived cell lines, as well as those derived from lung cancer, colon cancer, pancreatic cancer, prostate cancer, kidney cancer, leukemia and Burkitts lymphoma. Other more recent investigations have confirmed TPD52 or PrLZ RNA/protein overexpression in prostate cancer (Rubin et al., 2004; Wang et al., 2004).
TPD52 overexpression has also been predicted via expression microarray studies for prostate (Rhodes et al., 2002; Ahram et al., 2002; Best et al., 2003), ovarian (Shridhar et al., 2001), endometrial (Risinger et al., 2003; Cai et al., 2007), hepatocellular (Chen et al., 2002), lung (Garber et al., 2001; Bhattacharjee et al., 2001), melanoma (Bittner et al., 2000; Zhou et al., 2004; Hoek, 2007; Roesch et al., 2007) and testicular germ cell tumours (Sperger et al., 2003; Skotheim et al., 2005; Korkola et al., 2006; Skotheim et al., 2006; McIntyre et al., 2007; Korkola et al., 2008). TPD52 protein overexpression has been confirmed in melanoma (Roesch et al., 2007) and testicular germ cell tumours (Alagaratnam et al., 2009).
Prognosis
Expression microarray studies have revealed TPD52 gene overexpression to be associated with adverse outcome for patients with breast cancer (Adler et al., 2006; Liu et al., 2007), prostate cancer (Bismar et al., 2006) and mantle cell lymphoma (Ma et al., 2007). Higher levels of TPD52 expression amongst breast cancer patients has also been shown to be associated with reduced overall patient survival (Shehata et al., 2008a).
Cytogenetics
Using TPD52 expression and relative TPD52 copy number studies in breast specimens, Balleine et al. (2000) first proposed TPD52 as a target gene driving increased copy number of 8q21. Similar studies have since identified an association between TPD52 copy number gain and TPD52 expression in prostate cancer (Rubin et al., 2004; Wang et al., 2004) and ovarian cancer (Byrne et al., 2005). TPD52 has also been found to be amplified and overexpressed in multiple myeloma (Largo et al., 2006), pancreatic cancer xenografts (Loukopolous et al., 2007) and testicular germ cell tumours (Skotheim et al., 2006; Mcintyre et al., 2007; Korkola et al., 2008). However, TPD52 overexpression may also occur in the absence of gene amplification or gain (Balleine et al., 2000; Adelaide et al., 2007).
Oncogenesis
An increase in TPD52 expression has been found to be a comparatively early event in breast (Porter et al., 2003; Yu et al., 2004; Shehata et al., 2008a), prostate (Rubin et al., 2004; Wang et al., 2004) and ovarian cancer (Byrne et al., 2005). Other analyses also suggest that increased TPD52 expression may be a marker of tumour predisposition (Sims et al., 2007) or very early preneoplastic changes (Byrne et al., 2005). High TPD52 expression may also contribute to the development of primary carcinomas (Rubin et al., 2004; Wang et al., 2004; Bismar et al., 2006; Wang et al., 2007). The prevalence of high TPD52 expression in early and late stages of cancer suggest that it may contribute to tumour initiation and progression, possibly through independent mechanisms. It would be expected that TPD52 perturbs fundamental cell properties, as TPD52 overexpression exists in tumours of different cellular origins (Shehata et al., 2008b - review). Linking the ever-increasing experimental data obtained using TPD52-transfected cell lines with clinical studies should assist in forming more reliable predictions as to how TPD52 contributes to carcinogenesis and/or metastasis.

Bibliography

Pubmed IDLast YearTitleAuthors
180897852007Integrated profiling of basal and luminal breast cancers.Adélaïde J et al
165184022006Genetic regulators of large-scale transcriptional signatures in cancer.Adler AS et al
118079532002Proteomic analysis of human prostate cancer.Ahram M et al
190413652009TPD52, a candidate gene from genomic studies, is overexpressed in testicular germ cell tumours.Alagaratnam S et al
179400482007Increased survival of muscle stem cells lacking the MyoD gene after transplantation into regenerating skeletal muscle.Asakura A et al
109183932000The hD52 (TPD52) gene is a candidate target gene for events resulting in increased 8q21 copy number in human breast carcinoma.Balleine RL et al
127666102003Molecular differentiation of high- and moderate-grade human prostate cancer by cDNA microarray analysis.Best CJ et al
117075672001Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses.Bhattacharjee A et al
165334272006Defining aggressive prostate cancer using a 12-gene model.Bismar TA et al
109523172000Molecular classification of cutaneous malignant melanoma by gene expression profiling.Bittner M et al
155555432004The tumor protein D52 family: many pieces, many puzzles.Boutros R et al
159864282005Tumor protein D52 (TPD52) is overexpressed and a gene amplification target in ovarian cancer.Byrne JA et al
88124871996Definition of the tumor protein D52 (TPD52) gene family through cloning of D52 homologues in human (hD53) and mouse (mD52).Byrne JA et al
94847781998Identification of homo- and heteromeric interactions between members of the breast carcinoma-associated D52 protein family using the yeast two-hybrid system.Byrne JA et al
77964181995A screening method to identify genes commonly overexpressed in carcinomas and the identification of a novel complementary DNA sequence.Byrne JA et al
181791352007Comparison of the molecular classification with FIGO stage and histological grade on endometrial cancer.Cai B et al
166316102006A testis-specific and testis developmentally regulated tumor protein D52 (TPD52)-like protein TPD52L3/hD55 interacts with TPD52 family proteins.Cao Q et al
86328961996Isolation and characterization of a novel gene expressed in multiple cancers.Chen SL et al
120580602002Gene expression patterns in human liver cancers.Chen X et al
103395721999VIP17/MAL, a lipid raft-associated protein, is involved in apical transport in MDCK cells.Cheong KH et al
188324492008Calcium/calmodulin-dependent phosphorylation of tumor protein D52 on serine residue 136 may be mediated by CAMK2delta6.Chew CS et al
170054332007Specific changes in the proteomic pattern produced by the BRCA1-Ser1841Asn missense mutation.Crugliano T et al
121848062002Transcriptional programs activated by exposure of human prostate cancer cells to androgen.DePrimo SE et al
182925792008Innate immune responses to TREM-1 activation: overlap, divergence, and positive and negative cross-talk with bacterial lipopolysaccharide.Dower K et al
191759402009Mucin 1 (MUC1) is a novel partner for MAL2 in breast carcinoma cells.Fanayan S et al
117075902001Diversity of gene expression in adenocarcinoma of the lung.Garber ME et al
98869991999Immunolocalization of CRHSP28 in exocrine digestive glands and gastrointestinal tissues of the rat.Groblewski GE et al
110855042000Identification of c-myc responsive genes using rat cDNA microarray.Guo QM et al
179354902007DNA microarray analyses of melanoma gene expression: a decade in the mines.Hoek KS et al
97226071998Annexin VI-mediated loss of spectrin during coated pit budding is coupled to delivery of LDL to lysosomes.Kamal A et al
145197862003Dietary and hormonal stimulation of rat exocrine pancreatic function regulates CRHSP-28 phosphorylation in vivo.Kaspar KM et al
179439722008In vivo differentiation and genomic evolution in adult male germ cell tumors.Korkola JE et al
158978832005Gene expression profiling of cancer progression reveals intrinsic regulation of transforming growth factor-beta signaling in ErbB2/Neu-induced tumors from transgenic mice.Landis MD et al
164613022006Identification of overexpressed genes in frequently gained/amplified chromosome regions in multiple myeloma.Largo C et al
173142712007Induction of tumorigenesis and metastasis by the murine orthologue of tumor protein D52.Lewis JD et al
188003462009PrLZ expression is associated with the progression of prostate cancer LNCaP cells.Li L et al
172299492007The prognostic role of a gene signature from tumorigenic breast-cancer cells.Liu R et al
172338152007Genome-wide array-based comparative genomic hybridization analysis of pancreatic adenocarcinoma: identification of genetic indicators that predict patient outcome.Loukopoulos P et al
159949242005Autocrine platelet-derived growth factor-dependent gene expression in glioblastoma cells is mediated largely by activation of the transcription factor sterol regulatory element binding protein and is associated with altered genotype and patient survival in human brain tumors.Ma D et al
171827002007Clustering threshold gradient descent regularization: with applications to microarray studies.Ma S et al
95289961998Expression of the MAL gene in the thyroid: the MAL proteolipid, a component of glycolipid-enriched membranes, is apically distributed in thyroid follicles.Martín-Belmonte F et al
180594022007Genomic copy number and expression patterns in testicular germ cell tumours.McIntyre A et al
172873402007Global proteomic profiling of phosphopeptides using electron transfer dissociation tandem mass spectrometry.Molina H et al
121852492002The program of androgen-responsive genes in neoplastic prostate epithelium.Nelson PS et al
98380881998Cloning of a third member of the D52 gene family indicates alternative coding sequence usage in D52-like transcripts.Nourse CR et al
170819832006Global, in vivo, and site-specific phosphorylation dynamics in signaling networks.Olsen JV et al
87027301996Purification, cloning, and expression of a novel, endogenous, calcium-sensitive, 28-kDa phosphoprotein.Parente JA et al
126519092003Molecular markers in ductal carcinoma in situ of the breast.Porter D et al
123760032003D53 is a novel endosomal SNARE-binding protein that enhances interaction of syntaxin 1 with the synaptobrevin 2 complex in vitro.Proux-Gillardeaux V et al
101893741999The MAL proteolipid is necessary for normal apical transport and accurate sorting of the influenza virus hemagglutinin in Madin-Darby canine kidney cells.Puertollano R et al
121540502002Meta-analysis of microarrays: interstudy validation of gene expression profiles reveals pathway dysregulation in prostate cancer.Rhodes DR et al
125177682003Microarray analysis reveals distinct gene expression profiles among different histologic types of endometrial cancer.Risinger JI et al
180069412007Ataxia telangiectasia-mutated gene is a possible biomarker for discrimination of infiltrative deep penetrating nevi and metastatic vertical growth phase melanoma.Roesch A et al
151729882004Overexpression, amplification, and androgen regulation of TPD52 in prostate cancer.Rubin MA et al
115947512001The role of the coiled-coil motif in interactions mediated by TPD52.Sathasivam P et al
186980232008Nonredundant functions for tumor protein D52-like proteins support specific targeting of TPD52.Shehata M et al
191055692008Tumor protein D52 overexpression and gene amplification in cancers from a mosaic of microarrays.Shehata M et al
114792312001Genetic analysis of early- versus late-stage ovarian tumors.Shridhar V et al
174876802007TPD52 and NFKB1 gene expression levels correlate with G2 chromosomal radiosensitivity in lymphocytes of women with and at risk of hereditary breast cancer.Sims AH et al
171671842006Novel genomic aberrations in testicular germ cell tumors by array-CGH, and associated gene expression changes.Skotheim RI et al
145950152003Gene expression patterns in human embryonic stem cells and human pluripotent germ cell tumors.Sperger JM et al
121051902002Identification of annexin VI as a Ca2+-sensitive CRHSP-28-binding protein in pancreatic acinar cells.Thomas DD et al
200325132010Tumor protein D52 expression and Ca2+-dependent phosphorylation modulates lysosomal membrane protein trafficking to the plasma membrane.Thomas DD et al
113849732001CRHSP-28 regulates Ca(2+)-stimulated secretion in permeabilized acinar cells.Thomas DD et al
149776332004Secretagogue-induced translocation of CRHSP-28 within an early apical endosomal compartment in acinar cells.Thomas DD et al
155764732005Tumor protein D52 (TPD52): a novel B-cell/plasma-cell molecule with unique expression pattern and Ca(2+)-dependent association with annexin VI.Tiacci E et al
189597552008Altered expression of tumor protein D52 regulates apoptosis and migration of prostate cancer cells.Ummanni R et al
179474662007PrLZ is expressed in normal prostate development and in human prostate cancer progression.Wang R et al
115493202001Identification of MAL2, a novel member of the mal proteolipid family, though interactions with TPD52-like proteins in the yeast two-hybrid system.Wilson SH et al
153281902004Conservation of breast cancer molecular subtypes and transcriptional patterns of tumor progression across distinct ethnic populations.Yu K et al
178757332007PC-1/PrLZ contributes to malignant progression in prostate cancer.Zhang H et al
173119212007Intracellular localization of type III-delivered Pseudomonas ExoS with endosome vesicles.Zhang Y et al
123702462002MAL2, a novel raft protein of the MAL family, is an essential component of the machinery for transcytosis in hepatoma HepG2 cells.de Marco MC et al

Other Information

Locus ID:

NCBI: 7163
MIM: 604068
HGNC: 12005
Ensembl: ENSG00000076554

Variants:

dbSNP: 7163
ClinVar: 7163
TCGA: ENSG00000076554
COSMIC: TPD52

RNA/Proteins

Gene IDTranscript IDUniprot
ENSG00000076554ENST00000379096P55327
ENSG00000076554ENST00000379097P55327
ENSG00000076554ENST00000448733P55327
ENSG00000076554ENST00000517427P55327
ENSG00000076554ENST00000517462P55327
ENSG00000076554ENST00000518517E5RGI1
ENSG00000076554ENST00000518937P55327
ENSG00000076554ENST00000519303E5RK35
ENSG00000076554ENST00000520527P55327
ENSG00000076554ENST00000520795H0YC44
ENSG00000076554ENST00000521241E5RFR7
ENSG00000076554ENST00000521354E5RJ27
ENSG00000076554ENST00000523753E5RGI1

Expression (GTEx)

0
10
20
30
40
50
60

Pathways

PathwaySourceExternal ID
Vesicle-mediated transportREACTOMER-HSA-5653656
Membrane TraffickingREACTOMER-HSA-199991
trans-Golgi Network Vesicle BuddingREACTOMER-HSA-199992
Clathrin derived vesicle buddingREACTOMER-HSA-421837
Golgi Associated Vesicle BiogenesisREACTOMER-HSA-432722

Protein levels (Protein atlas)

Not detected
Low
Medium
High

References

Pubmed IDYearTitleCitations
209238222010Radiation pharmacogenomics: a genome-wide association approach to identify radiation response biomarkers using human lymphoblastoid cell lines.79
203796142010Personalized smoking cessation: interactions between nicotine dose, dependence and quit-success genotype score.62
206410332011Polymorphisms in predicted miRNA binding sites and osteoporosis.33
247689952014Tumour-suppressive microRNA-224 inhibits cancer cell migration and invasion via targeting oncogenic TPD52 in prostate cancer.29
198345352009Sequential use of transcriptional profiling, expression quantitative trait mapping, and gene association implicates MMP20 in human kidney aging.27
159864282005Tumor protein D52 (TPD52) is overexpressed and a gene amplification target in ovarian cancer.25
187106592008Expression of chloride intracellular channel protein 1 (CLIC1) and tumor protein D52 (TPD52) as potential biomarkers for colorectal cancer.24
189597552008Altered expression of tumor protein D52 regulates apoptosis and migration of prostate cancer cells.24
247989742014Tumor protein D52 (TPD52) and cancer-oncogene understudy or understudied oncogene?22
255117012015microRNA-218 inhibits prostate cancer cell growth and promotes apoptosis by repressing TPD52 expression.20

Citation

Austin Della-Franca ; Jennifer Byrne

TPD52 (tumor protein D52)

Atlas Genet Cytogenet Oncol Haematol. 2010-09-01

Online version: http://atlasgeneticsoncology.org/gene/42676/tpd52