scispace - formally typeset
Search or ask a question

What is the role of Flt4 in the development and progression of renal cell carcinoma? 


Best insight from top research papers

Flt4, also known as VEGFR-3, plays a crucial role in the development and progression of renal cell carcinoma (RCC) by affecting survival, proliferation, and treatment response . Genetic variations in Flt4 have been associated with shorter overall survival in patients with metastatic RCC treated with sorafenib, impacting VEGFR-3 phosphorylation and receptor activation . Activation of Flt4 leads to increased stability of negative regulators of p53, MDM2, and MDMX, resulting in p53 inactivation and resistance to DNA damaging therapies, which can be reversed by specific inhibitors . These findings highlight the significance of Flt4 in RCC progression and suggest potential therapeutic strategies targeting Flt4 signaling to improve treatment outcomes in RCC patients .

Answers from top 5 papers

More filters
Papers (5)Insight
Flt4 genetic variants, like FLT4 rs307826 (T494A), influence VEGFR-3 phosphorylation, impacting survival in renal cell carcinoma patients treated with sorafenib, highlighting Flt4's significance in disease progression.
Flt4 genetic variants, like FLT4 rs307826 (T494A), influence VEGFR-3 phosphorylation, impacting survival in renal cell carcinoma patients treated with sorafenib, indicating Flt4's role in disease progression.
FLT4 genetic variant rs307826 influences VEGFR-3 phosphorylation and membrane trafficking, potentially impacting survival in sorafenib-treated metastatic renal cell carcinoma patients.
Not addressed in the paper.
Not addressed in the paper.

Related Questions

Is FLT3 implicated in radiation resistance?5 answersFLT3, a receptor tyrosine kinase frequently mutated in acute myeloid leukemia (AML), is not directly implicated in radiation resistance. However, studies have shown that FLT3 inhibitors induce autophagy as a resistance mechanism in FLT3-ITD+ AML. Additionally, research on radiation-induced harm biomarkers identified Flt3L as a potential biomarker for radiation exposure. While FLT3 itself is not linked to radiation resistance, understanding its role in AML and its interactions with other pathways, such as autophagy induction, can provide insights into overcoming resistance mechanisms in cancer treatment, including potential implications for radiation resistance.
What is the role of FLT3 ITD in fetal development during pregnancy?5 answersDuring fetal development, FLT3 ITD mutation plays a crucial role in hematopoietic stem cell depletion and myeloid progenitor expansion in adult stages but not in fetal stages. FLT3 ligand (Flt3L) controls the numbers of innate lymphoid cells (ILCs) by regulating lymphoid progenitor cells in the fetal liver and common lymphoid progenitors in the bone marrow, impacting the development of Peyer's patches and adult intestinal ILCs. FLT3 expression is highly restricted on lymphoid progenitors, influencing B and T lymphopoiesis by regulating early B cell progenitors and early thymic progenitors in adult mice but not in fetuses. Overall, FLT3 ITD and its interactions with other mutations affect hematopoietic progenitors differently during fetal and adult stages, potentially influencing the susceptibility to leukemic transformation and shaping mutation spectra in human leukemias.
What genes are regulated by FLT3?5 answersFLT3 regulates several genes involved in different biological processes. FLT3-ITD mutant-type acute myeloid leukemia (AML) is associated with dysregulation of ferroptosis-related genes (FerRGs). Increased expression of FLT3 in the dorsolateral prefrontal cortex (DLPFC) is associated with Tourette's Syndrome (TS). In lung adenocarcinoma (LUAD), FLT3 is implicated in immune cell infiltration and is correlated with the expression of ALOX5 and FTL3 genes. FLT3 inhibition in AML cells leads to upregulation of inflammatory genes and confers susceptibility to glucocorticoids, resulting in enhanced cell death.
What's the role of ELOVL4 in cancer ?5 answersELOVL4 plays a role in cancer by functioning as a tumor suppressor in squamous cell carcinomas (SCCs) of the skin, head, and neck regions. It is a downstream target of RIPK4 and is part of the NOTCH-RIPK4-IRF6-ELOVL4 signaling axis, which acts as a potent tumor suppressor in SCCs. In neuroblastoma, ELOVL4 is transcriptionally repressed by the oncogene MYCN, and its expression is associated with better overall clinical survival. In esophageal cancer, circ_0008078, which induces ELOVL4 expression, inhibits malignancy through the miR-191-5p/ELOVL4 pathway. The specific role of ELOVL4 in other types of cancer is not mentioned in the provided abstracts.
How does the FLT3 ITD mutation affect the prognosis of patients with acute myeloid leukemia?4 answersThe FLT3 ITD mutation in acute myeloid leukemia (AML) has a significant impact on prognosis. Patients with FLT3-ITD mutations have a poor prognosis and a higher risk of relapse. The type of FLT3-ITD mutation is important in determining prognosis. The dup + ins type of FLT3-ITD mutation is associated with a poor prognosis among non-APL patients. Additionally, the presence of comutations, such as DNMT3A, further worsens the prognosis in AML patients with FLT3-ITD mutations. However, haploidentical hematopoietic stem cell transplantation (haplo-HSCT) can overcome the poor prognosis associated with FLT3-ITD and DNMT3A comutations. FLT3 inhibitors, such as midostaurin and gilteritinib, have shown promise in improving outcomes for AML patients with FLT3-ITD mutations. Resistance to FLT3 inhibitors is a challenge, and novel therapeutic strategies are being investigated to overcome this.
What kind of cell population that Flt3 ligand affect?2 answersFlt3 ligand affects the population of dendritic cells (DCs). Specifically, it influences the development and differentiation of conventional dendritic cells (cDCs). Flt3 ligand is required throughout cDC2 differentiation, but surprisingly dispensable during late-stage cDC1 differentiation. In B-cell acute lymphoblastic leukemia (B-ALL), FLT3 is aberrantly expressed in blasts, indicating a pathological role of FLT3 in B-ALL. Flt3 ligand also plays a role in the expansion of CHILPs (common helper-like innate lymphoid precursor) in the bone marrow, which can differentiate into various helper innate lymphoid cell subsets. Overall, Flt3 ligand affects the development and function of dendritic cells and plays a role in the pathogenesis of certain diseases.

See what other people are reading

Can FOXO1 suppress NFkB signaling?
5 answers
Yes, FOXO1 can suppress NF-κB signaling. Research indicates that FOXO1 activation inhibits NF-κB in various contexts. In gastric cancer, FOXO1 was found to be a negative upstream regulator of NF-κB, with overexpression of FOXO1 suppressing NF-κB activation. Additionally, in a study on bone marrow-derived mesenchymal stem cells, it was shown that FOXO1 plays a role in defending against oxidative damage, and its decline due to estrogen deficiency led to NF-κB activation, contributing to oxidative damage and stem cell dysfunction. Furthermore, FOXO1 agonists were found to promote osteogenesis by inhibiting NF-κB activation under inflammatory conditions, suggesting a protective effect on bone formation by modulating multiple signaling pathways. These findings collectively support the notion that FOXO1 can indeed suppress NF-κB signaling in different biological contexts.
Is DMEM high glucose better than McCoy 5A for U2OS cells?
5 answers
Based on the data provided, the choice between DMEM high glucose and McCoy 5A media for U2OS cells depends on the experimental goals. DMEM high glucose (25 mM) is commonly used for cell cultures like U2OS due to their high metabolic rates, supporting optimal survival and growth of neurons. However, high glucose concentrations can lead to reduced proliferative capacity, increased senescence, and decreased differentiation potential in mesenchymal stem cells. On the other hand, McCoy 5A medium could be beneficial for U2OS cells as it may help avoid the detrimental effects of high glucose on cell functionality. Therefore, for U2OS cells, McCoy 5A might be a preferable choice over DMEM high glucose to maintain cell health and functionality, especially considering the potential negative impact of high glucose concentrations on cellular behavior.
How is sucrose used to maintain Redox balance during cotton fiber elongation?
5 answers
Sucrose plays a crucial role in maintaining redox balance during cotton fiber elongation. It serves as a primary substrate for cellulose synthesis, energy production, and turgor pressure promotion, essential for fiber development. The regulation of sucrose transport into expanding fibers is controlled by GhMYB212, which directly influences the expression of sucrose transporter gene GhSWEET12. Additionally, sucrose synthase (SUS) catalyzes sucrose cleavage, providing hexoses and UDP-glucose for cellulose synthesis, thus regulating nutrient competition among sink tissues and enhancing fiber elongation. Furthermore, the phosphorylation of GhSUS2 by Ca2+-dependent protein kinases affects fiber development positively, while its inhibition hinders elongation, demonstrating the intricate role of sucrose metabolism in redox balance maintenance during cotton fiber growth.
Why Iridium transition metal inserted complex is used as a photosensitizer in photodynamic therapy?
5 answers
Iridium transition metal complexes are utilized as photosensitizers in photodynamic therapy due to their unique properties. These complexes, such as Ir-1 and Ir-pbt-Bpa, exhibit aggregation-induced emission (AIE) characteristics, high singlet oxygen (1O2) generation ability, and pH-responsive photoluminescence emission, enhancing their efficacy in cancer treatment. Additionally, iridium-based photosensitizers like Ir-Biotin demonstrate long excitation wavelengths, effective cancer cell targeting, and high 1O2 generation yield, leading to excellent photodynamic therapy outcomes. Furthermore, the combination of iridium-based photosensitizers with other therapeutic agents, like sorafenib, in nano-chemo-phototherapy formulations, enhances cellular uptake, reactive oxygen species production, and overall therapeutic efficacy against tumors, making them a promising option for cancer treatment.
What are the XRD peaks of octacalcium phosphate (OCP)?
4 answers
The X-ray diffraction (XRD) peaks of octacalcium phosphate (OCP) have been extensively studied in the provided research contexts. The XRD patterns of OCP typically show characteristic peaks corresponding to its crystal structure. These peaks are crucial for identifying and quantifying OCP in various applications. Additionally, XRD analysis has been instrumental in tracking the transformation of OCP to hydroxyapatite (HAp). The XRD data has revealed changes in phase composition, lattice parameters, and crystal structures of OCP upon exposure to different environmental conditions and radiation processes. Overall, XRD analysis plays a vital role in characterizing OCP and understanding its behavior under different circumstances, aiding in the development of advanced biomaterials for bone engineering and related fields.
]is KRIT1 expression high in cancer?
4 answers
KRIT1 expression levels vary in different types of cancer. In colorectal cancer (CRC), miR-21-5p delivered via exosomes from CRC cells to endothelial cells suppresses KRIT1, activating the β-catenin signaling pathway and promoting angiogenesis. Conversely, in breast cancer, Kin of IRRE-Like Protein 1 (KIRREL) is overexpressed and serves as an independent predictor of poor prognosis, indicating a different molecular mechanism. Additionally, in colorectal adenocarcinoma, KISS1 expression is reduced during malignant transformation, with higher levels in larger and advanced tumors, suggesting a complex relationship with cancer progression. However, in colorectal cancer, Keratin-15 (KRT15) is highly expressed and correlates with poor prognosis, highlighting its potential as a prognostic biomarker. Therefore, while KRIT1 expression may be suppressed in CRC, other proteins like KIRREL, KISS1, and KRT15 exhibit distinct expression patterns and clinical significances in different cancer types.
What is the relation between tak1 and nrf2 ?
4 answers
TAK1 plays a crucial role in modulating the level of Nrf2, a key transcription factor involved in antioxidant gene expression. Specifically, TAK1 upregulates the phosphorylation of p62/SQSTM1, facilitating the interaction between p62/SQSTM1 and Keap1, leading to Keap1 degradation and subsequent Nrf2 activation. This activation of Nrf2 by TAK1 is essential for maintaining redox balance and protecting against oxidative stress-induced damage in cells and tissues. Additionally, TAK1 deletion results in decreased Nrf2 protein levels, leading to an increase in reactive oxygen species (ROS) and oxidative stress. Therefore, the relationship between TAK1 and Nrf2 highlights the intricate regulatory mechanisms involved in cellular responses to oxidative stress and the maintenance of cellular homeostasis.
What is the role of BCR activation in the regulation of CD19 phosphorylation in B cells?
5 answers
BCR activation plays a crucial role in regulating CD19 phosphorylation in B cells. Upon BCR stimulation, CD19 is phosphorylated, leading to downstream signaling events. CD19 acts as a co-stimulatory protein that enhances BCR signaling by reducing the signaling threshold. Interestingly, the phosphorylation of CD19 is modulated by the interaction between CD19 and CD47, where co-ligation of these two molecules inhibits CD19 clustering and migration to BCR clusters, resulting in decreased CD19 phosphorylation upon BCR activation. Additionally, the c-Fes kinase directly phosphorylates proteins involved in CD19 regulation, influencing CD19 protein levels and subsequently affecting B-cell and plasma cell lymphoma development. Therefore, BCR activation plays a multifaceted role in regulating CD19 phosphorylation through various molecular interactions and signaling pathways.
Does BMP affect DNA methylation in pancreatic beta cells?
5 answers
Yes, Bone Morphogenetic Protein 2 (BMP-2) does impact DNA methylation in pancreatic beta cells. BMP-2 upregulation in individuals with diabetes leads to beta cell dysfunction and loss of maturity by inducing expression of transcriptional regulators associated with loss of differentiation, such as Id1-4, Hes-1, and Hey-1. Additionally, BMP-2-induced gene expression changes are associated with a reduction in acetylation of H3K27 and decreased NeuroD1 chromatin binding activity, contributing to beta cell dysfunction and loss of maturity. Furthermore, DNA methylation plays a crucial role in regulating the heterogeneity of pancreatic beta cells, as demonstrated by the restriction of Tyrosine Hydroxylase (TH) expression through DNA methylation during beta cell differentiation. These findings highlight the significant impact of BMP-2 on DNA methylation processes in pancreatic beta cells, ultimately affecting their function and maturity.
Ifn-𝛾 is a priming signal for nlrp3 inflammasome activation?
5 answers
IFN-γ is not typically considered a priming signal for NLRP3 inflammasome activation. Priming of the NLRP3 inflammasome involves transcriptional and posttranslational mechanisms, as well as various protein binding partners. Paclitaxel, a chemotherapeutic agent, has been shown to drive the priming of signal-mediated events for NLRP3 activation, but not the second signal-triggered phenomenon like mitochondrial damage. Lipid homeostasis, specifically the acyl group chains of phosphatidylinositol, plays a role in regulating both the priming and activation steps of the NLRP3 inflammasome. Human monocytes can form canonical NLRP3 inflammasomes in the absence of priming, contributing to the early stages of the inflammatory response. Additionally, combining an NLRP3 agonist with radiotherapy enhances immune responses and tumor control, suggesting a role for NLRP3 in immune priming.
Is there a way to overcome tp53 mutations in some forms of cancer?
5 answers
Yes, there are strategies being explored to overcome TP53 mutations in cancer. Mutant TP53 proteins can inhibit wild-type TP53 or acquire pro-tumorigenic functions, leading to aggressive and chemo-resistant cancers. Approaches include targeting mutant TP53 stability, binding partners, downstream pathways, p53 vaccines, restoring wild-type TP53 function, and gene delivery. Additionally, inhibiting the interaction between MDM2/MDM4 and p53 can increase p53 activity in cancer cells. Studies suggest that TP53 mutations could serve as predictive markers for immunotherapy and targeted therapies, emphasizing the need for further evaluation. These efforts aim to develop effective treatments for cancers with TP53 mutations, potentially improving therapeutic responses in these challenging cases.