Target Name: ATF7IP
NCBI ID: G55729
Review Report on ATF7IP Target / Biomarker Content of Review Report on ATF7IP Target / Biomarker
ATF7IP
Other Name(s): activating transcription factor 7 interacting protein | ATFa-associated modulator | hAM | ATF7IP variant 1 | Activating transcription factor 7 interacting protein, transcript variant 2 | ATF-IP | Activating transcription factor 7-interacting protein 1 (isoform 1) | FLJ10688 | ATF7IP variant 2 | FLJ10139 | P621 | AM | MBD1-containing chromatin-associated factor 1 | ATF7IP1 | p621 | MBD1-containing chromatin associated factor | MCAF1_HUMAN | ATF-interacting protein | MCAF1 | Activating transcription factor 7-interacting protein 1 | MCAF | Activating transcription factor 7-interacting protein 1 (isoform 2) | ATF7-interacting protein | Activating transcription factor 7 interacting protein, transcript variant 1

Unlocking the Potential of ATF7IP as a Drug Target and Biomarker

ATF7IP (Activating Transcription Factor 7 Interacting Protein) is a protein that plays a crucial role in various cellular processes, including cell growth, differentiation, and survival. The discovery of ATF7IP as a potential drug target and biomarker has significant implications for the development of new treatments for various diseases. In this article, we will explore the structure, function, and potential therapeutic applications of ATF7IP.

Structure and Function

ATF7IP is a 21-kDa protein that is expressed in various tissues, including brain, heart, and muscle. Its gene has four exons, and it is localized to the nuclear envelope, where it plays a role in regulating nuclear import and export of proteins. ATF7IP functions as a transcription factor, which means it can bind to specific DNA sequences and regulate gene expression.

ATF7IP is composed of multiple domains, including an N-terminal transmembrane domain, a coiled-coil domain, and a C-terminal T-loop domain. The N-terminal transmembrane domain is responsible for the protein's ability to interact with various cellular signaling pathways, including the TGF-β pathway. The coiled-coil domain is responsible for the protein's stability and functions as a binding site for other proteins. The C-terminal T-loop domain is involved in the regulation of protein-protein interactions and is involved in the formation of a complex with the transcription factor p53.

ATF7IP is a critical regulator of cellular processes, and its dysregulation has been implicated in various diseases, including cancer, neurodegenerative diseases, and developmental disorders. For example, studies have shown that ATF7IP is overexpressed in various cancer types, including breast, ovarian, and colorectal cancer. Its overexpression has been associated with poor prognosis and increased cancer stem cell properties.

In addition to its role in cancer, ATF7IP has also been implicated in several neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Studies have shown that ATF7IP is misfolded and forms aggregates in the brain, which is thought to contribute to the development of neurodegenerative diseases.

Potential Therapeutic Applications

The potential therapeutic applications of ATF7IP are vast, and research is ongoing to determine its effectiveness in treating various diseases. One of the primary targets of research is the development of drugs that can specifically modulate ATF7IP function to treat diseases associated with ATF7IP dysfunction.

One approach to treating ATF7IP-related diseases is to target the N-terminal transmembrane domain of ATF7IP. This can be done through the use of small molecules or antibodies that specifically bind to the N-terminal domain and inhibit its activity. Alternatively, researchers can target the C-terminal T-loop domain to prevent the formation of ATF7IP aggregates in the brain.

Another approach to treating ATF7IP-related diseases is to use drugs that modulate ATF7IP function through its coiled-coil domain. This can be done by using drugs that bind to the coiled-coil domain and alter its stability or activity.

In addition to these approaches, researchers are also exploring the use of gene editing technologies to modify ATF7IP function and treat diseases associated with ATF7IP dysfunction. For example, researchers have used CRISPR/Cas9 to edit the ATF7IP gene and generate compounds that specifically target the N-terminal transmembrane domain.

Conclusion

In conclusion, ATF7IP is a protein that has the potential to be a drug target and biomarker for various diseases. Its dysregulation is implicated in a range of diseases, including cancer, neurodegenerative diseases, and developmental disorders. The development of drugs that specifically modulate ATF7IP function has the potential to improve treatment outcomes for these diseases. Further research is needed to fully understand the potential therapeutic applications of ATF7IP and to develop safe and effective treatments.

Protein Name: Activating Transcription Factor 7 Interacting Protein

Functions: Recruiter that couples transcriptional factors to general transcription apparatus and thereby modulates transcription regulation and chromatin formation. Can both act as an activator or a repressor depending on the context. Required for HUSH-mediated heterochromatin formation and gene silencing (PubMed:27732843). Mediates MBD1-dependent transcriptional repression, probably by recruiting complexes containing SETDB1 (PubMed:12665582). Stabilizes SETDB1, is required to stimulate histone methyltransferase activity of SETDB1 and facilitates the conversion of dimethylated to trimethylated H3 'Lys-9' (H3K9me3). The complex formed with MBD1 and SETDB1 represses transcription and couples DNA methylation and histone H3 'Lys-9' trimethylation (H3K9me3) (PubMed:14536086, PubMed:27732843). Facilitates telomerase TERT and TERC gene expression by SP1 in cancer cells (PubMed:19106100)

The "ATF7IP Target / Biomarker Review Report" is a customizable review of hundreds up to thousends of related scientific research literature by AI technology, covering specific information about ATF7IP comprehensively, including but not limited to:
•   general information;
•   protein structure and compound binding;
•   protein biological mechanisms;
•   its importance;
•   the target screening and validation;
•   expression level;
•   disease relevance;
•   drug resistance;
•   related combination drugs;
•   pharmacochemistry experiments;
•   related patent analysis;
•   advantages and risks of development, etc.
The report is helpful for project application, drug molecule design, research progress updates, publication of research papers, patent applications, etc. If you are interested to get a full version of this report, please feel free to contact us at BD@silexon.ai

More Common Targets

ATF7IP2 | ATG10 | ATG101 | ATG12 | ATG13 | ATG14 | ATG16L1 | ATG16L2 | ATG2A | ATG2B | ATG3 | ATG4A | ATG4B | ATG4C | ATG4D | ATG5 | ATG7 | ATG9A | ATG9B | ATIC | ATL1 | ATL2 | ATL3 | ATM | ATMIN | ATN1 | ATOH1 | ATOH7 | ATOH8 | ATOSA | ATOSB | ATOX1 | ATOX1-AS1 | ATP Synthase, H+ Transporting, Mitochondrial F0 complex | ATP synthase, H+ transporting, mitochondrial F1 complex | ATP-Binding Cassette (ABC) Transporter | ATP-dependent 6-phosphofructokinase | ATP10A | ATP10B | ATP10D | ATP11A | ATP11A-AS1 | ATP11AUN | ATP11B | ATP11C | ATP12A | ATP13A1 | ATP13A2 | ATP13A3 | ATP13A3-DT | ATP13A4 | ATP13A5 | ATP13A5-AS1 | ATP1A1 | ATP1A1-AS1 | ATP1A2 | ATP1A3 | ATP1A4 | ATP1B1 | ATP1B2 | ATP1B3 | ATP1B4 | ATP23 | ATP2A1 | ATP2A1-AS1 | ATP2A2 | ATP2A3 | ATP2B1 | ATP2B1-AS1 | ATP2B2 | ATP2B3 | ATP2B4 | ATP2C1 | ATP2C2 | ATP4A | ATP4B | ATP5F1A | ATP5F1B | ATP5F1C | ATP5F1D | ATP5F1E | ATP5F1EP2 | ATP5IF1 | ATP5MC1 | ATP5MC1P3 | ATP5MC2 | ATP5MC3 | ATP5ME | ATP5MF | ATP5MG | ATP5MGL | ATP5MJ | ATP5MK | ATP5PB | ATP5PBP5 | ATP5PD | ATP5PDP3 | ATP5PF | ATP5PO | ATP6