Transfected Stable Cell Lines
Reliable | High-Performance | Wide Rage
Precision reporter, kinase, immune receptor, biosimilar, Cas9, and knockout stable cell lines for diverse applications.
| Cat.No. | Product Name | Price |
|---|
| Cat.No. | Product Name | Price |
|---|
| Cat.No. | Product Name | Price |
|---|
| Cat.No. | Product Name | Price |
|---|
The TNK1 (Tyrosine Kinase Non-Receptor 1) gene is located on human chromosome 17p13.1 and belongs to the ACK (Activated Cdc42-associated kinase) family of non-receptor tyrosine kinases. TNK1 encodes a multi-domain kinase comprising an N-terminal catalytic tyrosine kinase domain (TyrKc), a central SH3 (Src Homology 3) domain, and a C-terminal ubiquitin-associated domain (UBA). This structural composition suggests a complex role in intracellular signaling networks. Transcript analysis reveals that TNK1 undergoes alternative splicing to produce multiple isoforms, with the major variant consisting of 1001 amino acids and an approximate molecular weight of 110 kDa. TNK1 exhibits pronounced developmental stage-specific expression, with high levels in fetal tissues and more restricted expression in adults, mainly in the central nervous system, hematopoietic stem cells, and reproductive tissues, suggesting a fundamental role during embryogenesis and tissue-specific functions in adulthood.
The regulatory mechanisms controlling TNK1 expression are not fully understood, but its promoter contains multiple transcription factor binding sites, including SP1, AP-1, and STAT3, which respond to extracellular signals such as growth factors and cytokines. Epigenetic regulation also plays a role, with DNA methylation studies showing hypomethylation of the TNK1 promoter in several cancers, correlating with aberrant activation. Post-translationally, TNK1 is tightly controlled by 14-3-3 proteins and the ubiquitin-proteasome system. Phosphorylation of TNK1 at Ser502 by MAPK promotes 14-3-3 binding, maintaining the kinase in an inactive state. Release from 14-3-3 allows the UBA domain to interact with ubiquitinated proteins, enhancing kinase activity and downstream signaling. This dual regulatory mechanism enables TNK1 to respond dynamically to intracellular signals and finely tune its functional activity.
Evolutionarily, TNK1 is highly conserved from Drosophila to mammals, reflecting its fundamental biological importance. In the human genome, TNK1 shares about 45% amino acid homology with its family member TNK2 (also known as ACK1). While TNK2 primarily functions as an oncogene, TNK1 often acts as a tumor suppressor in normal cells, a distinction likely stemming from structural differences, including the absence of the Cdc42-binding CRIB domain in TNK1 and the presence of a unique C-terminal regulatory sequence. Understanding TNK1's structural features and regulatory mechanisms is key to elucidating its physiological and pathological roles.
Figure 1. Tnk1/Kos1 uncouples Grb2-Sos1 GEF complex. (May WS, et al., 2010)
TNK1 functions as a non-receptor tyrosine kinase, phosphorylating specific substrates to modulate multiple signaling pathways. Under basal conditions, TNK1 exists in a low-activity state, with activation dependent on mechanisms including 14-3-3 dissociation, ubiquitin binding, and interactions with other signaling proteins. The UBA domain can bind polyubiquitin chains, inducing conformational changes that expose the catalytic domain and increase kinase activity. Activated TNK1 participates in processes such as cell proliferation, differentiation, survival, and migration. In the Ras-MAPK pathway, TNK1 negatively regulates signaling by phosphorylating specific tyrosine residues on Raf1, suppressing ERK activity.
TNK1 also plays a dual role in apoptosis. Overexpression in immortalized human hepatocytes can trigger intrinsic apoptotic pathways, including mitochondrial outer membrane permeabilization, cytochrome C release, and caspase activation. TNK1 influences the balance of Bcl-2 family proteins, activating pro-apoptotic Bax and Bak while inhibiting anti-apoptotic Bcl-2 and Bcl-xL. It also participates in death receptor signaling by promoting caspase-8 and caspase-10 activation. Interestingly, TNK1 can promote cell survival in certain contexts, such as pancreatic cancer, reflecting cell type-specific functional variability. TNK1's expression in liver cancer is often epigenetically suppressed, impairing its pro-apoptotic role and potentially facilitating tumor cell survival.
TNK1 is involved in cellular stress responses, including DNA damage repair and oxidative stress. It can be activated by DNA-damaging agents, phosphorylate histone H2AX, and recruit DNA repair factors. TNK1 also regulates p53 stability and transcriptional activity, influencing cell cycle arrest. Under oxidative stress, TNK1 activates the Nrf2 pathway, enhancing antioxidant enzyme expression and protecting cells from reactive oxygen species. Additionally, TNK1 modulates autophagy by phosphorylating key autophagy-related proteins, impacting autophagosome formation. These diverse functions position TNK1 as a critical node in cellular stress response networks.
During development, TNK1 is essential for embryogenesis. In zebrafish, TNK1 knockout leads to defects in neural and cardiovascular development. In mammals, TNK1 contributes to axon growth, synaptic plasticity, and neuronal morphology, potentially by regulating cytoskeletal dynamics. In hematopoiesis, TNK1 is expressed in stem and progenitor cells, maintaining self-renewal capacity. These developmental roles are linked to TNK1's ability to regulate multiple signaling pathways, including Wnt, Notch, and Hedgehog.
TNK1 exhibits tissue-specific dual roles in cancer, functioning as both a tumor suppressor and a potential oncogene. As a tumor suppressor, TNK1 is downregulated in several cancers, which can result from promoter hypermethylation or gene deletion. Loss of TNK1 expression is associated with increased tumor aggressiveness and impaired apoptotic responses. Restoring TNK1 expression can inhibit cancer cell proliferation, migration, and invasion while promoting apoptosis through Ras-MAPK inhibition, caspase activation, and modulation of FAK/paxillin signaling.
Conversely, TNK1 can display oncogenic properties in specific contexts. Gene amplification in acute myeloid leukemia enhances TNK1 expression, activating STAT3 signaling and promoting leukemic cell survival. In multiple myeloma, TNK1 stabilizes BCL6 and represses differentiation-promoting factors, maintaining tumor cell undifferentiated states. In pancreatic ductal adenocarcinoma, TNK1 cooperates with KRAS mutations to activate PI3K/AKT/mTOR signaling, promoting tumor growth and metastasis. These tissue-specific roles are influenced by substrate specificity, isoform expression, and interactions with particular signaling pathways.
TNK1 also affects the tumor microenvironment. Tumor-derived TNK1 can influence macrophage polarization, favoring immunosuppressive M2 phenotypes, and promote angiogenesis through VEGFR2 phosphorylation, enhancing endothelial proliferation and migration. TNK1 contributes to metabolic reprogramming by activating HIF-1α and glycolysis-related genes, supporting the Warburg effect. These findings underscore TNK1's broader role beyond intrinsic tumor cell regulation, shaping a supportive microenvironment.
TNK1 is a promising target for therapeutic intervention. Small molecule inhibitors targeting the TNK1 kinase domain or SH3 domain have shown efficacy in preclinical cancer models, reducing tumor growth, enhancing apoptosis, and potentially improving immunotherapy outcomes. Gene therapy approaches, including viral vector-mediated TNK1 restoration and mRNA-based delivery systems, can reactivate TNK1 expression in tumors with epigenetic silencing. CRISPR activation strategies have also been explored to restore endogenous TNK1 function selectively.
Challenges for TNK1-targeted therapy include ensuring selectivity to minimize effects on normal tissues, particularly the nervous system, and addressing the dual roles of TNK1 across different cancers. Next-generation approaches are being developed, including conditionally activated prodrugs, PROTAC-based degraders, and tissue-specific delivery systems. With advancing understanding of TNK1 biology and therapeutic technology, TNK1-targeted interventions have the potential to enter clinical evaluation and provide new options for treating difficult-to-treat cancers.
Reference
Contact us today for a free consultation with the scientific team and discover how Creative Biogene can be a valuable resource and partner for your organization.
Inquiry