Zinc transporter SLC39A7: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
Citation bot (talk | contribs)
Add: pmid, issue, volume. | Use this bot. Report bugs. | #UCB_CommandLine
Citation bot (talk | contribs)
Altered issue. Formatted dashes. | Use this bot. Report bugs. | Suggested by Boghog | #UCB_webform
(9 intermediate revisions by 4 users not shown)
Line 1: Line 1:
{{Short description|Protein found in humans}}
{{Short description|Protein found in humans}}
{{cs1 config|name-list-style=vanc|display-authors=6}}
{{Infobox_gene}}
{{Infobox_gene}}
'''Zinc transporter SLC39A7''' ('''ZIP7'''), also known as '''solute carrier family 39 member 7''', is a [[protein]] that in humans is encoded by the ''SLC39A7'' [[gene]].<ref name="pmid8812499">{{cite journal | vauthors = Ando A, Kikuti YY, Shigenari A, Kawata H, Okamoto N, Shiina T, Chen L, Ikemura T, Abe K, Kimura M, Inoko H | title = cDNA cloning of the human homologues of the mouse Ke4 and Ke6 genes at the centromeric end of the human MHC region | journal = Genomics | volume = 35 | issue = 3 | pages = 600–2 |date=Dec 1996 | pmid = 8812499 | doi = 10.1006/geno.1996.0405 }}</ref><ref name="pmid1855816">{{cite journal | vauthors = Hanson IM, Trowsdale J | title = Colinearity of novel genes in the class II regions of the MHC in mouse and human | journal = Immunogenetics | volume = 34 | issue = 1 | pages = 5–11 |date=Aug 1991 | pmid = 1855816 | doi =10.1007/BF00212306 | s2cid = 30046348 }}</ref><ref name="entrez">{{cite web | title = Entrez Gene: SLC39A7 solute carrier family 39 (zinc transporter), member 7| url = https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7922}}</ref> Its [[Drosophilia|fruit fly]] [[orthologue]] is [[Catecholamines up|''Catsup'']].
'''Zinc transporter SLC39A7''' ('''ZIP7'''), also known as '''solute carrier family 39 member 7''', is a [[transmembrane protein]] that in humans is encoded by the ''SLC39A7'' [[gene]].<ref name="pmid8812499">{{cite journal | vauthors = Ando A, Kikuti YY, Shigenari A, Kawata H, Okamoto N, Shiina T, Chen L, Ikemura T, Abe K, Kimura M, Inoko H | title = cDNA cloning of the human homologues of the mouse Ke4 and Ke6 genes at the centromeric end of the human MHC region | journal = Genomics | volume = 35 | issue = 3 | pages = 600–602 | date = August 1996 | pmid = 8812499 | doi = 10.1006/geno.1996.0405 }}</ref><ref name="pmid1855816">{{cite journal | vauthors = Hanson IM, Trowsdale J | title = Colinearity of novel genes in the class II regions of the MHC in mouse and human | journal = Immunogenetics | volume = 34 | issue = 1 | pages = 5–11 | date = Aug 1991 | pmid = 1855816 | doi = 10.1007/BF00212306 | s2cid = 30046348 }}</ref><ref name="entrez">{{cite web | title = Entrez Gene: SLC39A7 solute carrier family 39 (zinc transporter), member 7| url = https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7922}}</ref><ref name="Zhao_2019">{{cite journal | vauthors = Zhao T, Huang Q, Su Y, Sun W, Huang Q, Wei W | title = Zinc and its regulators in pancreas | journal = Inflammopharmacology | volume = 27 | issue = 3 | pages = 453–464 | date = June 2019 | pmid = 30756223 | doi = 10.1007/s10787-019-00573-w }}</ref><ref name="Kambe_2017">{{cite journal | vauthors = Kambe T, Matsunaga M, Takeda TA | title = Understanding the Contribution of Zinc Transporters in the Function of the Early Secretory Pathway | journal = International Journal of Molecular Sciences | volume = 18 | issue = 10 | pages = 2179 | date = October 2017 | pmid = 29048339 | pmc = 5666860 | doi = 10.3390/ijms18102179 | doi-access = free }}</ref><ref name="Zhang_2017">{{cite journal | vauthors = Zhang T, Liu J, Fellner M, Zhang C, Sui D, Hu J | title = Crystal structures of a ZIP zinc transporter reveal a binuclear metal center in the transport pathway | journal = Science Advances | volume = 3 | issue = 8 | pages = e1700344 | date = August 2017 | pmid = 28875161 | pmc = 5573306 | doi = 10.1126/sciadv.1700344 | bibcode = 2017SciA....3E0344Z }}</ref><ref name="Baltaci_2018">{{cite journal | vauthors = Baltaci AK, Yuce K | title = Zinc Transporter Proteins | journal = Neurochemical Research | volume = 43 | issue = 3 | pages = 517–530 | date = March 2018 | pmid = 29243032 | doi = 10.1007/s11064-017-2454-y }}</ref> It belongs to the ZIP family, which consists of 14 proteins that transport [[zinc]] into the [[cytoplasm]].<ref name="Zhao_2019" /><ref name="Kambe_2017" /><ref name="Zhang_2017" /><ref name="Baltaci_2018" /> It's primary role is to control the transport of zinc from the [[Endoplasmic reticulum|ER]] and [[Golgi apparatus]] to the cytoplasm.<ref name="Zhao_2019" /><ref name="Kambe_2017" /><ref name="Zhang_2017" /><ref name="Baltaci_2018" /> It also plays a role in [[Carbohydrate metabolism|glucose metabolism]].<ref name="Zhao_2019" /><ref name="Zhang_2017" /><ref name = "Fukunaka_2018">{{cite journal | vauthors = Fukunaka A, Fujitani Y | title = Role of Zinc Homeostasis in the Pathogenesis of Diabetes and Obesity | journal = International Journal of Molecular Sciences | volume = 19 | issue = 2 | pages = 476 | date = February 2018 | pmid = 29415457 | pmc = 5855698 | doi = 10.3390/ijms19020476 | doi-access = free }}</ref> Its structure consists of [[Alpha helix|helices]] that bind to zinc in a binuclear metal center.<ref name="Kambe_2017" /><ref name="Zhang_2017" /> Its [[Drosophilia|fruit fly]] [[orthologue]] is [[Catecholamines up|''Catsup'']].[[File:Full 3D Alpha Fold Structure of ZIP7.png|thumb|ZIP7 3D Alpha Fold structure modeled in ChimeraX. Accession number: AF-Q92504-F1<ref name="www.uniprot.org">{{Cite web |title=UniProt |url=https://www.uniprot.org/uniprotkb/Q92504/entry |access-date=2024-04-13 |website=www.uniprot.org}}</ref>]]


== Function ==
== Function ==


[[Zinc]] is an essential [[cofactor (biochemistry)|cofactor]] for more than 50 classes of [[enzyme]]s. It is involved in protein, nucleic acid, carbohydrate, and lipid metabolism, as well as in the control of [[gene transcription]], growth, development, and differentiation. Zinc cannot passively diffuse across cell membranes and requires specific transporters, such as SLC39A7, to enter the [[cytosol]] from both the extracellular environment and from intracellular storage compartments.<ref name="entrez" />
[[Zinc]] is an essential [[cofactor (biochemistry)|cofactor]] for more than 50 classes of [[enzyme]]s. It is involved in protein, nucleic acid, carbohydrate, and lipid metabolism, as well as in the control of [[gene transcription]], growth, development, and differentiation. Zinc cannot passively diffuse across cell membranes and requires specific transporters, such as SLC39A7, to enter the [[cytosol]] from both the extracellular environment and from intracellular storage compartments.<ref name="entrez" /> The presence of zinc regulates the expression of ZIP transporters.<ref name="Zhao_2019" />


ZIP7 is a [[membrane transport protein]] of the [[endoplasmic reticulum]].<ref>{{cite journal | vauthors = Taylor KM, Morgan HE, Johnson A, Nicholson RI | title = Structure-function analysis of HKE4, a member of the new LIV-1 subfamily of zinc transporters | journal = The Biochemical Journal | volume = 377 | issue = Pt 1 | pages = 131–139 | date = January 2004 | pmid = 14525538 | pmc = 1223853 | doi = 10.1042/BJ20031183 }}</ref> Phosphorylation of ZIP7 by [[casein kinase 2]] stimulates the release of [[zinc]] ions from the endoplasmic reticulum<ref>{{cite journal | vauthors = Taylor KM, Kille P, Hogstrand C | title = Protein kinase CK2 opens the gate for zinc signaling | journal = Cell Cycle | volume = 11 | issue = 10 | pages = 1863–1864 | date = May 2012 | pmid = 22580452 | pmc = 3359116 | doi = 10.4161/cc.20414 }}</ref> This provides a [[signal transduction]] pathway by which activation of cell surface receptors such as the [[epidermal growth factor receptor]] can regulate the activity of downstream [[phosphatase]]s and [[kinase]]s. ZIP7 is responsible for maintaining zinc [[homeostasis]] in the ER.<ref name="Kambe_2017" /> Due to it's key role in several signaling pathways, the loss of ZIP7 results in an accumulation in the endoplasmic reticulum and cause [[ER stress]].<ref name="Zhao_2019" /><ref name="Kambe_2017" /><ref name="Baltaci_2018" />
ZIP7 is a [[membrane transport protein]] of the [[endoplasmic reticulum]].<ref>{{Cite journal

| vauthors = Taylor KM, Morgan HE, Johnson A, Nicholson RI
ZIP7 is involved in controlling [[Carbohydrate metabolism|glucose metabolism]] in the [[Skeletal muscle|skeletal muscle cells]] by affecting the [[Insulin signal transduction pathway|insulin signaling]] pathway.<ref name="Zhao_2019" /><ref name="Zhang_2017" /><ref name = "Fukunaka_2018" /> Reduced expression in glucose metabolism genes and proteins such as [[GLUT4|Glut4]], [[Insulin receptor substrate 1|IRS1]], [[IRS2]], and Akt phosphorylation occur when ZIP7 mRNA is downregulated.<ref name="Zhao_2019" /><ref name="Fukunaka_2018" /> When zinc released from ZIP7 binds to PTP1B, the insulin signaling pathway is activated.<ref name="Fukunaka_2018" />
| title = Structure‒function analysis of HKE4, a member of the new LIV-1 subfamily of zinc transporters

| doi = 10.1042/BJ20031183
== Structure ==
| journal = Biochemical Journal
[[File:Zoomed_in_3D_Alpha_Fold_Structure_of_ZIP7.png|thumb|Zoomed in ZIP7 3D Alpha Fold structure modeled in ChimeraX. No zinc atom is present. The dark blue helix is TM4 and the orange helix is TM5. The red residues are amino acids involved in binding to two zinc atoms.<ref name="Zhang_2017" /> Accession number: AF-Q92504-F1<ref name="www.uniprot.org" />]]
| volume = 377
There are no experimentally solved structures of ZIP7 in its entirety.<ref name="www.uniprot.org" /> ZIP7 has an predicted [[AlphaFold]] structure.<ref name="www.uniprot.org" /> ZIP7, like other ZIP proteins, has eight transmembrane (TM) helices with a binuclear metal center.<ref name="Kambe_2017" /><ref name="Zhang_2017" /> Two zinc ions bind to residues on TM4 (His329, Asn330, and Asp333) and TM5 (His358, Glu395, and His362).<ref name="Kambe_2017" /><ref name="Zhang_2017" /><ref name="www.uniprot.org" /> ZIP proteins are known to make homo- or heterodimeric complexes.<ref name="Kambe_2017" /> The specific mode of transport zinc takes through ZIP transporters has not yet been determined.<ref name="Kambe_2017" />
| issue = Pt 1
| pages = 131–139
| year = 2004
| pmid = 14525538
| pmc =1223853
}}</ref> Phosphorylation of ZIP7 by [[casein kinase 2]] stimulates the release of [[zinc]] ions from the endoplasmic reticulum<ref>{{Cite journal
| vauthors = Taylor KM, Kille P, Hogstrand C
| doi = 10.4161/cc.20414
| title = Protein kinase CK2 opens the gate for zinc signaling
| journal = Cell Cycle
| volume = 11
| issue = 10
| pages = 1863–1864
| year = 2012
| pmid = 22580452
| pmc =3359116
}}</ref> This provides a [[signal transduction]] pathway by which activation of cell surface receptors such as the [[epidermal growth factor receptor]] can regulate the activity of downstream [[phosphatase]]s and [[kinase]]s.


== Role in Cancer ==
== Role in Cancer ==


ZIP7, a member of the solute carrier family 39 (SLC39) of zinc transporters, emerges as a pivotal factor in cancer progression across multiple malignancies. In breast cancer, ZIP7 expression is markedly elevated in primary tumors, particularly in basal and Her2 subtypes, and correlates with advanced disease stage, metastasis, recurrence, and poorer prognosis. Notably, its hyperactivation is implicated in endocrine resistance, suggesting a crucial role in Endocrine therapy resistance mechanisms.<ref>{{cite journal |last1=Liu |first1=Limei |last2=Yang |first2=Jiaomin |last3=Wang |first3=Chao |title=Analysis of the prognostic significance of solute carrier (SLC) family 39 genes in breast cancer |journal=Bioscience Reports |date=28 August 2020 |volume=40 |issue=8 |doi=10.1042/BSR20200764|pmid=32744318 |pmc=7426635 }}</ref><ref>{{cite journal |last1=Samuel |first1=Jones |last2=Georgia |first2=Farr |last3=Thirayost |first3=Nimmanon |last4=Silvia |first4=Ziliotto |last5=Julia M.W. |first5=Gee |last6=Kathryn M. |first6=Taylor |title=The importance of targeting signalling mechanisms of the SLC39A family of zinc transporters to inhibit endocrine resistant breast cancer |journal=Exploration of Targeted Anti-tumor Therapy |date=26 April 2022 |volume=3 |issue=2 |pages=224–239 |doi=10.37349/etat.2022.00080|pmid=35591900 |pmc=7612740 }}</ref>
ZIP7, a member of the solute carrier family 39 (SLC39) of zinc transporters, emerges as a pivotal factor in cancer progression across multiple malignancies. In breast cancer, ZIP7 expression is markedly elevated in primary tumors, particularly in basal and Her2 subtypes, and correlates with advanced disease stage, metastasis, recurrence, and poorer prognosis. Notably, its hyperactivation is implicated in endocrine resistance, suggesting a crucial role in Endocrine therapy resistance mechanisms.<ref>{{cite journal | vauthors = Liu L, Yang J, Wang C | title = Analysis of the prognostic significance of solute carrier (SLC) family 39 genes in breast cancer | journal = Bioscience Reports | volume = 40 | issue = 8 | date = August 2020 | pmid = 32744318 | pmc = 7426635 | doi = 10.1042/BSR20200764 }}</ref><ref>{{cite journal | vauthors = Jones S, Farr G, Nimmanon T, Ziliotto S, Gee JM, Taylor KM | title = The importance of targeting signalling mechanisms of the SLC39A family of zinc transporters to inhibit endocrine resistant breast cancer | journal = Exploration of Targeted Anti-Tumor Therapy | volume = 3 | issue = 2 | pages = 224–239 | date = April 2022 | pmid = 35591900 | pmc = 7612740 | doi = 10.37349/etat.2022.00080 }}</ref>


In colorectal cancer, ZIP7 upregulation is observed in tumor tissues compared to normal counterparts. Inhibition of ZIP7 leads to suppressed cell proliferation, colony formation, and enhanced apoptosis, while its heightened presence correlates with adverse patient outcomes, highlighting its significance as a potential prognostic marker.<ref>{{cite journal |last1=Sheng |first1=Nengquan |last2=Yan |first2=Li |last3=You |first3=Weiqiang |last4=Tan |first4=Gewen |last5=Gong |first5=Jianfeng |last6=Chen |first6=Hongqi |last7=Yang |first7=Yi |last8=Hu |first8=Landian |last9=Wang |first9=Zhigang |title=Knockdown of SLC39A7 inhibits cell growth and induces apoptosis in human colorectal cancer cells |journal=Acta Biochimica et Biophysica Sinica |date=1 October 2017 |volume=49 |issue=10 |pages=926–934 |doi=10.1093/abbs/gmx094}}</ref><ref>{{cite journal |last1=Luo |first1=Yang |last2=Shen |first2=Yicheng |last3=Ju |first3=Zhong |last4=Zhang |first4=Zhi |title=ZIP7 (SLC39A7) expression in colorectal cancer and its correlation with clinical prognosis |journal=Translational Cancer Research |date=October 2020 |volume=9 |issue=10 |pages=6471–6478 |doi=10.21037/tcr-20-2640|doi-access=free |pmid=35117255 |pmc=8798949 }}</ref> Similarly, ZIP7 exhibits elevated expression in cervical cancer tissues, where its knockdown results in inhibited proliferation, migration, and invasion of cancer cells. Furthermore, modulation of epithelial-mesenchymal transition markers underscores ZIP7's involvement in metastatic processes, suggesting its potential as a therapeutic target to impede disease progression.<ref>{{cite journal |last1=Wei |first1=Yongqing |last2=Dong |first2=Jie |last3=Li |first3=Fuli |last4=Wei |first4=Zhuqing |last5=Tian |first5=Yuling |title=Knockdown of SLC39A7 suppresses cell proliferation, migration and invasion in cervical cancer |journal=EXCLI Journal; 16:Doc1165; ISSN 1611-2156 |date=2017 |doi=10.17179/excli2017-690}}</ref> In hepatocellular carcinoma, specific inhibition of ZIP7 attenuates PI3K/AKT signaling, leading to suppressed cell growth, colony formation, migration, invasion, and enhanced apoptosis both in vitro and in vivo. This underscores ZIP7's critical role in hepatocellular carcinoma tumorigenesis and its potential as a therapeutic target in this malignancy.<ref>{{cite journal |last1=Tong |first1=Qing |last2=Yan |first2=Dong |last3=Cao |first3=Yan |last4=Dong |first4=Xiaogang |last5=Abula |first5=Yimamumaimaitijiang |last6=Yang |first6=Huan |last7=Kong |first7=Panpan |last8=Yi |first8=Mingyu |title=NVS-ZP7-4 inhibits hepatocellular carcinoma tumorigenesis and promotes apoptosis via PI3K/AKT signaling |journal=Scientific Reports |date=21 July 2023 |volume=13 |issue=1 |page=11795 |doi=10.1038/s41598-023-38596-7|pmid=37479837 |pmc=10362011 |bibcode=2023NatSR..1311795T }}</ref>
In colorectal cancer, ZIP7 upregulation is observed in tumor tissues compared to normal counterparts. Inhibition of ZIP7 leads to suppressed cell proliferation, colony formation, and enhanced apoptosis, while its heightened presence correlates with adverse patient outcomes, highlighting its significance as a potential prognostic marker.<ref>{{cite journal | vauthors = Sheng N, Yan L, You W, Tan G, Gong J, Chen H, Yang Y, Hu L, Wang Z | title = Knockdown of SLC39A7 inhibits cell growth and induces apoptosis in human colorectal cancer cells | journal = Acta Biochimica et Biophysica Sinica | volume = 49 | issue = 10 | pages = 926–934 | date = October 2017 | pmid = 28981607 | doi = 10.1093/abbs/gmx094 }}</ref><ref>{{cite journal | vauthors = Luo Y, Shen Y, Ju Z, Zhang Z | title = ZIP7 (SLC39A7) expression in colorectal cancer and its correlation with clinical prognosis | journal = Translational Cancer Research | volume = 9 | issue = 10 | pages = 6471–6478 | date = October 2020 | pmid = 35117255 | pmc = 8798949 | doi = 10.21037/tcr-20-2640 | doi-access = free }}</ref> Similarly, ZIP7 exhibits elevated expression in cervical cancer tissues, where its knockdown results in inhibited proliferation, migration, and invasion of cancer cells. Furthermore, modulation of epithelial-mesenchymal transition markers underscores ZIP7's involvement in metastatic processes, suggesting its potential as a therapeutic target to impede disease progression.<ref>{{cite journal | vauthors = Wei Y, Dong J, Li F, Wei Z, Tian Y | title = Knockdown of SLC39A7 suppresses cell proliferation, migration and invasion in cervical cancer | journal = EXCLI Journal | volume = 16 | pages = 1165–1176 | date = 2017 | pmid = 29285013 | doi = 10.17179/excli2017-690 }}</ref> In hepatocellular carcinoma, specific inhibition of ZIP7 attenuates PI3K/AKT signaling, leading to suppressed cell growth, colony formation, migration, invasion, and enhanced apoptosis both in vitro and in vivo. This underscores ZIP7's critical role in hepatocellular carcinoma tumorigenesis and its potential as a therapeutic target in this malignancy.<ref>{{cite journal | vauthors = Tong Q, Yan D, Cao Y, Dong X, Abula Y, Yang H, Kong P, Yi M | title = NVS-ZP7-4 inhibits hepatocellular carcinoma tumorigenesis and promotes apoptosis via PI3K/AKT signaling | journal = Scientific Reports | volume = 13 | issue = 1 | pages = 11795 | date = July 2023 | pmid = 37479837 | pmc = 10362011 | doi = 10.1038/s41598-023-38596-7 | bibcode = 2023NatSR..1311795T }}</ref>


Moreover, microRNAs play a regulatory role in ZIP7 expression across different cancer types. For instance, in prostate cancer, miR-15a-3p targets ZIP7, leading to the suppression of the Wnt/β-catenin signaling pathway and inhibition of proliferation, invasion, and epithelial-mesenchymal transition. Similarly, in gastric cancer, miR-139-5p negatively regulates ZIP7, inhibiting ZIP7-mediated activation of the Akt/mTOR pathway, thereby suppressing cell proliferation and migration while promoting apoptosis.<ref>{{cite journal |last1=Cui |first1=Yu |last2=Yang |first2=Yi |last3=Ren |first3=Liang |last4=Yang |first4=Jun |last5=Wang |first5=Bin |last6=Xing |first6=Tianjun |last7=Chen |first7=Huiqing |last8=Chen |first8=Mingxiao |title=miR-15a-3p Suppresses Prostate Cancer Cell Proliferation and Invasion by Targeting SLC39A7 Via Downregulating Wnt/β-Catenin Signaling Pathway |journal=Cancer Biotherapy and Radiopharmaceuticals |date=1 September 2019 |volume=34 |issue=7 |pages=472–479 |doi=10.1089/cbr.2018.2722|pmid=31135177 }}</ref><ref>{{cite journal |last1=Zhang |first1=Yanting |last2=Bai |first2=Jie |last3=Si |first3=Wangli |last4=Yuan |first4=Shanshan |last5=Li |first5=Yijun |last6=Chen |first6=Xiaolu |title=SLC39A7, regulated by miR-139-5p, induces cell proliferation, migration and inhibits apoptosis in gastric cancer via Akt/mTOR signaling pathway |journal=Bioscience Reports |date=28 February 2020 |volume=40 |issue=2 |doi=10.1042/BSR20200041|pmid=32109290 |pmc=7048674 }}</ref>
Moreover, microRNAs play a regulatory role in ZIP7 expression across different cancer types. For instance, in prostate cancer, miR-15a-3p targets ZIP7, leading to the suppression of the Wnt/β-catenin signaling pathway and inhibition of proliferation, invasion, and epithelial-mesenchymal transition. Similarly, in gastric cancer, miR-139-5p negatively regulates ZIP7, inhibiting ZIP7-mediated activation of the Akt/mTOR pathway, thereby suppressing cell proliferation and migration while promoting apoptosis.<ref>{{cite journal | vauthors = Cui Y, Yang Y, Ren L, Yang J, Wang B, Xing T, Chen H, Chen M | title = miR-15a-3p Suppresses Prostate Cancer Cell Proliferation and Invasion by Targeting SLC39A7 Via Downregulating Wnt/β-Catenin Signaling Pathway | journal = Cancer Biotherapy & Radiopharmaceuticals | volume = 34 | issue = 7 | pages = 472–479 | date = September 2019 | pmid = 31135177 | doi = 10.1089/cbr.2018.2722 }}</ref><ref>{{cite journal | vauthors = Zhang Y, Bai J, Si W, Yuan S, Li Y, Chen X | title = SLC39A7, regulated by miR-139-5p, induces cell proliferation, migration and inhibits apoptosis in gastric cancer via Akt/mTOR signaling pathway | journal = Bioscience Reports | volume = 40 | issue = 2 | date = February 2020 | pmid = 32109290 | pmc = 7048674 | doi = 10.1042/BSR20200041 }}</ref>


In a study presented at the 2024 American Association for Cancer Research (AACR) Annual Meeting, researchers introduced rabbit polyclonal antibodies specifically targeting ZIP7 to both human triple-negative breast cancer cells (TNBC) and normal breast epithelial cells (NBE) obtained from the same patient. Utilizing flow cytometry analysis, they observed substantial binding of the ZIP7 antibodies to TNBC cells, while minimal binding was noted in NBE cells from the same individual. Moreover, cytotoxicity assays revealed that the ZIP7-targeted antibodies, in combination with a secondary anti-rabbit Antibody-Drug Conjugate (ADC), selectively induced cell death in TNBC cells over NBE cells. Importantly, this preferential killing effect was attributed to the aberrant surface expression of ZIP7 on TNBC cells, coupled with its involvement in cell proliferation-related signaling pathways specific to TNBC.<ref>{{cite journal |last1=Manavalan |first1=John Sanil |last2=Mor |first2=Danielle |last3=Davis |first3=Joseph |last4=Saini |first4=Sharanjot |last5=Pal |first5=Ipsita |last6=Feith |first6=David |last7=Hikichi |first7=Yuichi |last8=Davis |first8=Elliot |title=Abstract 4109: Discovery of a novel cancer-specific antigen for therapeutic targeting using the Oncotope platform |journal=Cancer Research |date=22 March 2024 |volume=84 |issue=6_Supplement |pages=4109 |doi=10.1158/1538-7445.AM2024-4109}}</ref>
In a study presented at the 2024 American Association for Cancer Research (AACR) Annual Meeting, researchers introduced rabbit polyclonal antibodies specifically targeting ZIP7 to both human triple-negative breast cancer cells (TNBC) and normal breast epithelial cells (NBE) obtained from the same patient. Utilizing flow cytometry analysis, they observed substantial binding of the ZIP7 antibodies to TNBC cells, while minimal binding was noted in NBE cells from the same individual. Moreover, cytotoxicity assays revealed that the ZIP7-targeted antibodies, in combination with a secondary anti-rabbit Antibody-Drug Conjugate (ADC), selectively induced cell death in TNBC cells over NBE cells. Importantly, this preferential killing effect was attributed to the aberrant surface expression of ZIP7 on TNBC cells, coupled with its involvement in cell proliferation-related signaling pathways specific to TNBC.<ref>{{cite journal | vauthors = Manavalan JS, Mor D, Davis J, Saini S, Pal I, Feith D, Hikichi Y, Davis E |title=Abstract 4109: Discovery of a novel cancer-specific antigen for therapeutic targeting using the Oncotope platform |journal=Cancer Research |date=22 March 2024 |volume=84 |issue=6_Supplement |pages=4109 |doi=10.1158/1538-7445.AM2024-4109}}</ref>


In summary, ZIP7 emerges as a critical regulator of cancer progression, influencing key cellular processes such as proliferation, invasion, migration, and apoptosis across various malignancies. Targeting ZIP7 or its regulatory mechanisms holds therapeutic promise in cancer treatment strategies, highlighting its potential as a prognostic marker and therapeutic target in oncology research.
In summary, ZIP7 emerges as a critical regulator of cancer progression, influencing key cellular processes such as proliferation, invasion, migration, and apoptosis across various malignancies. Targeting ZIP7 or its regulatory mechanisms holds therapeutic promise in cancer treatment strategies, highlighting its potential as a prognostic marker and therapeutic target in oncology research.


==See also==
== See also ==
* [[Solute carrier family]]
* [[Solute carrier family]]


==References==
== References ==
{{reflist}}
{{reflist}}


==Further reading==
== Further reading ==
{{refbegin | 2}}
{{refbegin | 2}}
*{{cite journal | vauthors=Maruyama K, Sugano S |title=Oligo-capping: a simple method to replace the cap structure of eukaryotic mRNAs with oligoribonucleotides |journal=Gene |volume=138 |issue= 1–2 |pages= 171–4 |year= 1994 |pmid= 8125298 |doi=10.1016/0378-1119(94)90802-8 }}
* {{cite journal | vauthors = Maruyama K, Sugano S | title = Oligo-capping: a simple method to replace the cap structure of eukaryotic mRNAs with oligoribonucleotides | journal = Gene | volume = 138 | issue = 1–2 | pages = 171–174 | date = January 1994 | pmid = 8125298 | doi = 10.1016/0378-1119(94)90802-8 }}
*{{cite journal |vauthors=Suzuki Y, Yoshitomo-Nakagawa K, Maruyama K, etal |title=Construction and characterization of a full length-enriched and a 5'-end-enriched cDNA library |journal=Gene |volume=200 |issue= 1–2 |pages= 149–56 |year= 1997 |pmid= 9373149 |doi=10.1016/S0378-1119(97)00411-3 }}
* {{cite journal | vauthors = Suzuki Y, Yoshitomo-Nakagawa K, Maruyama K, Suyama A, Sugano S | title = Construction and characterization of a full length-enriched and a 5'-end-enriched cDNA library | journal = Gene | volume = 200 | issue = 1–2 | pages = 149–156 | date = October 1997 | pmid = 9373149 | doi = 10.1016/S0378-1119(97)00411-3 }}
*{{cite journal |vauthors=Strausberg RL, Feingold EA, Grouse LH, etal |title=Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences |journal=Proc. Natl. Acad. Sci. U.S.A. |volume=99 |issue= 26 |pages= 16899–903 |year= 2003 |pmid= 12477932 |doi= 10.1073/pnas.242603899 | pmc=139241 |bibcode=2002PNAS...9916899M |doi-access=free }}
* {{cite journal | vauthors = Taylor KM, Morgan HE, Johnson A, Hadley LJ, Nicholson RI | title = Structure-function analysis of LIV-1, the breast cancer-associated protein that belongs to a new subfamily of zinc transporters | journal = The Biochemical Journal | volume = 375 | issue = Pt 1 | pages = 51–59 | date = October 2003 | pmid = 12839489 | pmc = 1223660 | doi = 10.1042/BJ20030478 }}
*{{cite journal |vauthors=Taylor KM, Morgan HE, Johnson A, etal |title=Structure-function analysis of LIV-1, the breast cancer-associated protein that belongs to a new subfamily of zinc transporters |journal=Biochem. J. |volume=375 |issue= Pt 1 |pages= 51–9 |year= 2003 |pmid= 12839489 |doi= 10.1042/BJ20030478 | pmc=1223660 }}
* {{cite journal | vauthors = Huang L, Kirschke CP, Zhang Y, Yu YY | title = The ZIP7 gene (Slc39a7) encodes a zinc transporter involved in zinc homeostasis of the Golgi apparatus | journal = The Journal of Biological Chemistry | volume = 280 | issue = 15 | pages = 15456–15463 | date = April 2005 | pmid = 15705588 | doi = 10.1074/jbc.M412188200 | doi-access = free }}
*{{cite journal | vauthors=Taylor KM, Morgan HE, Johnson A, Nicholson RI |title=Structure-function analysis of HKE4, a member of the new LIV-1 subfamily of zinc transporters |journal=Biochem. J. |volume=377 |issue= Pt 1 |pages= 131–9 |year= 2004 |pmid= 14525538 |doi= 10.1042/BJ20031183 | pmc=1223853 }}
* {{cite journal | vauthors = Kim JE, Tannenbaum SR, White FM | title = Global phosphoproteome of HT-29 human colon adenocarcinoma cells | journal = Journal of Proteome Research | volume = 4 | issue = 4 | pages = 1339–1346 | year = 2005 | pmid = 16083285 | doi = 10.1021/pr050048h }}
* {{cite journal | vauthors = Kimura K, Wakamatsu A, Suzuki Y, Ota T, Nishikawa T, Yamashita R, Yamamoto J, Sekine M, Tsuritani K, Wakaguri H, Ishii S, Sugiyama T, Saito K, Isono Y, Irie R, Kushida N, Yoneyama T, Otsuka R, Kanda K, Yokoi T, Kondo H, Wagatsuma M, Murakawa K, Ishida S, Ishibashi T, Takahashi-Fujii A, Tanase T, Nagai K, Kikuchi H, Nakai K, Isogai T, Sugano S | title = Diversification of transcriptional modulation: large-scale identification and characterization of putative alternative promoters of human genes | journal = Genome Research | volume = 16 | issue = 1 | pages = 55–65 | date = January 2006 | pmid = 16344560 | pmc = 1356129 | doi = 10.1101/gr.4039406 }}
*{{cite journal |vauthors=Mungall AJ, Palmer SA, Sims SK, etal |title=The DNA sequence and analysis of human chromosome 6 |journal=Nature |volume=425 |issue= 6960 |pages= 805–11 |year= 2003 |pmid= 14574404 |doi= 10.1038/nature02055 |bibcode=2003Natur.425..805M |doi-access=free }}
* {{cite journal | vauthors = Ewing RM, Chu P, Elisma F, Li H, Taylor P, Climie S, McBroom-Cerajewski L, Robinson MD, O'Connor L, Li M, Taylor R, Dharsee M, Ho Y, Heilbut A, Moore L, Zhang S, Ornatsky O, Bukhman YV, Ethier M, Sheng Y, Vasilescu J, Abu-Farha M, Lambert JP, Duewel HS, Stewart II, Kuehl B, Hogue K, Colwill K, Gladwish K, Muskat B, Kinach R, Adams SL, Moran MF, Morin GB, Topaloglou T, Figeys D | title = Large-scale mapping of human protein-protein interactions by mass spectrometry | journal = Molecular Systems Biology | volume = 3 | issue = 1 | pages = 89 | year = 2007 | pmid = 17353931 | pmc = 1847948 | doi = 10.1038/msb4100134 }}
*{{cite journal |vauthors=Gerhard DS, Wagner L, Feingold EA, etal |title=The Status, Quality, and Expansion of the NIH Full-Length cDNA Project: The Mammalian Gene Collection (MGC) |journal=Genome Res. |volume=14 |issue= 10B |pages= 2121–7 |year= 2004 |pmid= 15489334 |doi= 10.1101/gr.2596504 | pmc=528928 }}
*{{cite journal | vauthors=Huang L, Kirschke CP, Zhang Y, Yu YY |title=The ZIP7 gene (Slc39a7) encodes a zinc transporter involved in zinc homeostasis of the Golgi apparatus |journal=J. Biol. Chem. |volume=280 |issue= 15 |pages= 15456–63 |year= 2005 |pmid= 15705588 |doi= 10.1074/jbc.M412188200 |doi-access= free }}
*{{cite journal | vauthors=Kim JE, Tannenbaum SR, White FM |title=Global phosphoproteome of HT-29 human colon adenocarcinoma cells |journal=J. Proteome Res. |volume=4 |issue= 4 |pages= 1339–46 |year= 2005 |pmid= 16083285 |doi= 10.1021/pr050048h }}
*{{cite journal |vauthors=Kimura K, Wakamatsu A, Suzuki Y, etal |title=Diversification of transcriptional modulation: Large-scale identification and characterization of putative alternative promoters of human genes |journal=Genome Res. |volume=16 |issue= 1 |pages= 55–65 |year= 2006 |pmid= 16344560 |doi= 10.1101/gr.4039406 | pmc=1356129 }}
*{{cite journal |vauthors=Ewing RM, Chu P, Elisma F, etal |title=Large-scale mapping of human protein–protein interactions by mass spectrometry |journal=Mol. Syst. Biol. |volume=3 |issue= 1|pages= 89 |year= 2007 |pmid= 17353931 |doi= 10.1038/msb4100134 | pmc=1847948 }}
{{refend}}
{{refend}}


Line 68: Line 49:


[[Category:Solute carrier family]]
[[Category:Solute carrier family]]



{{membrane-protein-stub}}
{{membrane-protein-stub}}

Revision as of 07:13, 4 May 2024

SLC39A7
Identifiers
AliasesSLC39A7, D6S115E, D6S2244E, H2-KE4, HKE4, KE4, RING5, ZIP7, solute carrier family 39 member 7, AGM9
External IDsOMIM: 601416 MGI: 95909 HomoloGene: 5072 GeneCards: SLC39A7
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_006979
NM_001077516
NM_001288777

NM_001077709
NM_008202

RefSeq (protein)

NP_001070984
NP_001275706
NP_008910
NP_001070984.1
NP_008910.2

NP_001071177
NP_032228

Location (UCSC)Chr 6: 33.2 – 33.2 MbChr 17: 34.25 – 34.25 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

Zinc transporter SLC39A7 (ZIP7), also known as solute carrier family 39 member 7, is a transmembrane protein that in humans is encoded by the SLC39A7 gene.[5][6][7][8][9][10][11] It belongs to the ZIP family, which consists of 14 proteins that transport zinc into the cytoplasm.[8][9][10][11] It's primary role is to control the transport of zinc from the ER and Golgi apparatus to the cytoplasm.[8][9][10][11] It also plays a role in glucose metabolism.[8][10][12] Its structure consists of helices that bind to zinc in a binuclear metal center.[9][10] Its fruit fly orthologue is Catsup.

ZIP7 3D Alpha Fold structure modeled in ChimeraX. Accession number: AF-Q92504-F1[13]

Function

Zinc is an essential cofactor for more than 50 classes of enzymes. It is involved in protein, nucleic acid, carbohydrate, and lipid metabolism, as well as in the control of gene transcription, growth, development, and differentiation. Zinc cannot passively diffuse across cell membranes and requires specific transporters, such as SLC39A7, to enter the cytosol from both the extracellular environment and from intracellular storage compartments.[7] The presence of zinc regulates the expression of ZIP transporters.[8]

ZIP7 is a membrane transport protein of the endoplasmic reticulum.[14] Phosphorylation of ZIP7 by casein kinase 2 stimulates the release of zinc ions from the endoplasmic reticulum[15] This provides a signal transduction pathway by which activation of cell surface receptors such as the epidermal growth factor receptor can regulate the activity of downstream phosphatases and kinases. ZIP7 is responsible for maintaining zinc homeostasis in the ER.[9] Due to it's key role in several signaling pathways, the loss of ZIP7 results in an accumulation in the endoplasmic reticulum and cause ER stress.[8][9][11]

ZIP7 is involved in controlling glucose metabolism in the skeletal muscle cells by affecting the insulin signaling pathway.[8][10][12] Reduced expression in glucose metabolism genes and proteins such as Glut4, IRS1, IRS2, and Akt phosphorylation occur when ZIP7 mRNA is downregulated.[8][12] When zinc released from ZIP7 binds to PTP1B, the insulin signaling pathway is activated.[12]

Structure

Zoomed in ZIP7 3D Alpha Fold structure modeled in ChimeraX. No zinc atom is present. The dark blue helix is TM4 and the orange helix is TM5. The red residues are amino acids involved in binding to two zinc atoms.[10] Accession number: AF-Q92504-F1[13]

There are no experimentally solved structures of ZIP7 in its entirety.[13] ZIP7 has an predicted AlphaFold structure.[13] ZIP7, like other ZIP proteins, has eight transmembrane (TM) helices with a binuclear metal center.[9][10] Two zinc ions bind to residues on TM4 (His329, Asn330, and Asp333) and TM5 (His358, Glu395, and His362).[9][10][13] ZIP proteins are known to make homo- or heterodimeric complexes.[9] The specific mode of transport zinc takes through ZIP transporters has not yet been determined.[9]

Role in Cancer

ZIP7, a member of the solute carrier family 39 (SLC39) of zinc transporters, emerges as a pivotal factor in cancer progression across multiple malignancies. In breast cancer, ZIP7 expression is markedly elevated in primary tumors, particularly in basal and Her2 subtypes, and correlates with advanced disease stage, metastasis, recurrence, and poorer prognosis. Notably, its hyperactivation is implicated in endocrine resistance, suggesting a crucial role in Endocrine therapy resistance mechanisms.[16][17]

In colorectal cancer, ZIP7 upregulation is observed in tumor tissues compared to normal counterparts. Inhibition of ZIP7 leads to suppressed cell proliferation, colony formation, and enhanced apoptosis, while its heightened presence correlates with adverse patient outcomes, highlighting its significance as a potential prognostic marker.[18][19] Similarly, ZIP7 exhibits elevated expression in cervical cancer tissues, where its knockdown results in inhibited proliferation, migration, and invasion of cancer cells. Furthermore, modulation of epithelial-mesenchymal transition markers underscores ZIP7's involvement in metastatic processes, suggesting its potential as a therapeutic target to impede disease progression.[20] In hepatocellular carcinoma, specific inhibition of ZIP7 attenuates PI3K/AKT signaling, leading to suppressed cell growth, colony formation, migration, invasion, and enhanced apoptosis both in vitro and in vivo. This underscores ZIP7's critical role in hepatocellular carcinoma tumorigenesis and its potential as a therapeutic target in this malignancy.[21]

Moreover, microRNAs play a regulatory role in ZIP7 expression across different cancer types. For instance, in prostate cancer, miR-15a-3p targets ZIP7, leading to the suppression of the Wnt/β-catenin signaling pathway and inhibition of proliferation, invasion, and epithelial-mesenchymal transition. Similarly, in gastric cancer, miR-139-5p negatively regulates ZIP7, inhibiting ZIP7-mediated activation of the Akt/mTOR pathway, thereby suppressing cell proliferation and migration while promoting apoptosis.[22][23]

In a study presented at the 2024 American Association for Cancer Research (AACR) Annual Meeting, researchers introduced rabbit polyclonal antibodies specifically targeting ZIP7 to both human triple-negative breast cancer cells (TNBC) and normal breast epithelial cells (NBE) obtained from the same patient. Utilizing flow cytometry analysis, they observed substantial binding of the ZIP7 antibodies to TNBC cells, while minimal binding was noted in NBE cells from the same individual. Moreover, cytotoxicity assays revealed that the ZIP7-targeted antibodies, in combination with a secondary anti-rabbit Antibody-Drug Conjugate (ADC), selectively induced cell death in TNBC cells over NBE cells. Importantly, this preferential killing effect was attributed to the aberrant surface expression of ZIP7 on TNBC cells, coupled with its involvement in cell proliferation-related signaling pathways specific to TNBC.[24]

In summary, ZIP7 emerges as a critical regulator of cancer progression, influencing key cellular processes such as proliferation, invasion, migration, and apoptosis across various malignancies. Targeting ZIP7 or its regulatory mechanisms holds therapeutic promise in cancer treatment strategies, highlighting its potential as a prognostic marker and therapeutic target in oncology research.

See also

References

  1. ^ a b c ENSG00000229802, ENSG00000226614, ENSG00000112473, ENSG00000206288, ENSG00000224399 GRCh38: Ensembl release 89: ENSG00000227402, ENSG00000229802, ENSG00000226614, ENSG00000112473, ENSG00000206288, ENSG00000224399Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000024327Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ Ando A, Kikuti YY, Shigenari A, Kawata H, Okamoto N, Shiina T, et al. (August 1996). "cDNA cloning of the human homologues of the mouse Ke4 and Ke6 genes at the centromeric end of the human MHC region". Genomics. 35 (3): 600–602. doi:10.1006/geno.1996.0405. PMID 8812499.
  6. ^ Hanson IM, Trowsdale J (Aug 1991). "Colinearity of novel genes in the class II regions of the MHC in mouse and human". Immunogenetics. 34 (1): 5–11. doi:10.1007/BF00212306. PMID 1855816. S2CID 30046348.
  7. ^ a b "Entrez Gene: SLC39A7 solute carrier family 39 (zinc transporter), member 7".
  8. ^ a b c d e f g h Zhao T, Huang Q, Su Y, Sun W, Huang Q, Wei W (June 2019). "Zinc and its regulators in pancreas". Inflammopharmacology. 27 (3): 453–464. doi:10.1007/s10787-019-00573-w. PMID 30756223.
  9. ^ a b c d e f g h i j Kambe T, Matsunaga M, Takeda TA (October 2017). "Understanding the Contribution of Zinc Transporters in the Function of the Early Secretory Pathway". International Journal of Molecular Sciences. 18 (10): 2179. doi:10.3390/ijms18102179. PMC 5666860. PMID 29048339.
  10. ^ a b c d e f g h i Zhang T, Liu J, Fellner M, Zhang C, Sui D, Hu J (August 2017). "Crystal structures of a ZIP zinc transporter reveal a binuclear metal center in the transport pathway". Science Advances. 3 (8): e1700344. Bibcode:2017SciA....3E0344Z. doi:10.1126/sciadv.1700344. PMC 5573306. PMID 28875161.
  11. ^ a b c d Baltaci AK, Yuce K (March 2018). "Zinc Transporter Proteins". Neurochemical Research. 43 (3): 517–530. doi:10.1007/s11064-017-2454-y. PMID 29243032.
  12. ^ a b c d Fukunaka A, Fujitani Y (February 2018). "Role of Zinc Homeostasis in the Pathogenesis of Diabetes and Obesity". International Journal of Molecular Sciences. 19 (2): 476. doi:10.3390/ijms19020476. PMC 5855698. PMID 29415457.
  13. ^ a b c d e "UniProt". www.uniprot.org. Retrieved 2024-04-13.
  14. ^ Taylor KM, Morgan HE, Johnson A, Nicholson RI (January 2004). "Structure-function analysis of HKE4, a member of the new LIV-1 subfamily of zinc transporters". The Biochemical Journal. 377 (Pt 1): 131–139. doi:10.1042/BJ20031183. PMC 1223853. PMID 14525538.
  15. ^ Taylor KM, Kille P, Hogstrand C (May 2012). "Protein kinase CK2 opens the gate for zinc signaling". Cell Cycle. 11 (10): 1863–1864. doi:10.4161/cc.20414. PMC 3359116. PMID 22580452.
  16. ^ Liu L, Yang J, Wang C (August 2020). "Analysis of the prognostic significance of solute carrier (SLC) family 39 genes in breast cancer". Bioscience Reports. 40 (8). doi:10.1042/BSR20200764. PMC 7426635. PMID 32744318.
  17. ^ Jones S, Farr G, Nimmanon T, Ziliotto S, Gee JM, Taylor KM (April 2022). "The importance of targeting signalling mechanisms of the SLC39A family of zinc transporters to inhibit endocrine resistant breast cancer". Exploration of Targeted Anti-Tumor Therapy. 3 (2): 224–239. doi:10.37349/etat.2022.00080. PMC 7612740. PMID 35591900.
  18. ^ Sheng N, Yan L, You W, Tan G, Gong J, Chen H, et al. (October 2017). "Knockdown of SLC39A7 inhibits cell growth and induces apoptosis in human colorectal cancer cells". Acta Biochimica et Biophysica Sinica. 49 (10): 926–934. doi:10.1093/abbs/gmx094. PMID 28981607.
  19. ^ Luo Y, Shen Y, Ju Z, Zhang Z (October 2020). "ZIP7 (SLC39A7) expression in colorectal cancer and its correlation with clinical prognosis". Translational Cancer Research. 9 (10): 6471–6478. doi:10.21037/tcr-20-2640. PMC 8798949. PMID 35117255.
  20. ^ Wei Y, Dong J, Li F, Wei Z, Tian Y (2017). "Knockdown of SLC39A7 suppresses cell proliferation, migration and invasion in cervical cancer". EXCLI Journal. 16: 1165–1176. doi:10.17179/excli2017-690. PMID 29285013.
  21. ^ Tong Q, Yan D, Cao Y, Dong X, Abula Y, Yang H, et al. (July 2023). "NVS-ZP7-4 inhibits hepatocellular carcinoma tumorigenesis and promotes apoptosis via PI3K/AKT signaling". Scientific Reports. 13 (1): 11795. Bibcode:2023NatSR..1311795T. doi:10.1038/s41598-023-38596-7. PMC 10362011. PMID 37479837.
  22. ^ Cui Y, Yang Y, Ren L, Yang J, Wang B, Xing T, et al. (September 2019). "miR-15a-3p Suppresses Prostate Cancer Cell Proliferation and Invasion by Targeting SLC39A7 Via Downregulating Wnt/β-Catenin Signaling Pathway". Cancer Biotherapy & Radiopharmaceuticals. 34 (7): 472–479. doi:10.1089/cbr.2018.2722. PMID 31135177.
  23. ^ Zhang Y, Bai J, Si W, Yuan S, Li Y, Chen X (February 2020). "SLC39A7, regulated by miR-139-5p, induces cell proliferation, migration and inhibits apoptosis in gastric cancer via Akt/mTOR signaling pathway". Bioscience Reports. 40 (2). doi:10.1042/BSR20200041. PMC 7048674. PMID 32109290.
  24. ^ Manavalan JS, Mor D, Davis J, Saini S, Pal I, Feith D, et al. (22 March 2024). "Abstract 4109: Discovery of a novel cancer-specific antigen for therapeutic targeting using the Oncotope platform". Cancer Research. 84 (6_Supplement): 4109. doi:10.1158/1538-7445.AM2024-4109.

Further reading

This article incorporates text from the United States National Library of Medicine, which is in the public domain.