scispace - formally typeset
Search or ask a question

What are the applications of Flt3 receptor interacting lectin FRIL? 


Best insight from top research papers

The Flt3 receptor-interacting lectin (FRIL), extracted from plants such as Dolichos Lablab, has shown significant potential in various biomedical applications, particularly in the field of stem cell research and cancer therapy. One of the primary applications of FRIL is in the ex vivo maintenance of stem cells. Studies have demonstrated that FRIL can preserve hematopoietic stem cells (HSCs) in vitro by maintaining them in a quiescent state, thereby inhibiting both cell proliferation and differentiation. This effect is crucial for many clinical applications, including stem cell transplantation and gene therapy, as it allows for the extended manipulation and expansion of stem cells without losing their primitive characteristics . FRIL achieves this by down-regulating cell cycle regulators such as cyclin D3 and CDK6, and activating P53, which are involved in the G0/G1 phase regulation, thus preserving the self-renewal property of HSCs/HPCs by halting their cell cycles . Additionally, FRIL's interaction with novel hematopoietic cell cycle regulators, HTm4 and HTm4S, plays a crucial role in maintaining the quiescence of human cord blood stem cells, further highlighting its potential for clinical applications . Beyond stem cell preservation, FRIL has implications in cancer therapy. The FLT3 gene, which FRIL interacts with, is implicated in the development of acute myeloid leukemia (AML). Research has shown that T cells modified with a chimeric antigen receptor that includes an FLT3 binding domain can specifically target and kill leukemia cells expressing FLT3, suggesting that FRIL or its mechanisms could be harnessed for targeted cancer immunotherapy . Moreover, the use of FRIL in conjunction with RNA vaccines to enhance immune responses against cancer cells is being explored, indicating its potential role in vaccine delivery and immunotherapy . In summary, FRIL's applications span from stem cell preservation, offering a novel approach to maintain stem cells in a quiescent state for extended periods, to targeted cancer therapy, where its interaction with FLT3 could be leveraged for designing more effective treatments against FLT3+ leukemia and potentially other cancers .

Answers from top 10 papers

More filters
Papers (10)Insight
FRIL, a lectin interacting with the Flt3 receptor, preserves hematopoietic progenitors without cytokines, potentially aiding in stem cell transplantation, leukemia treatment, and gene therapy applications.
Not addressed in the paper.
FRIL, a lectin from lablab, maintains hematopoietic stem/progenitor cells' multipotential in vitro by halting cell cycles, enhancing colony formation, and promoting self-renewal, showing potential in stem cell research.
FRIL from Dolichos lablab preserves quiescent hematopoietic stem cells in suspension culture, indicating its potential application in stem cell research and biotechnology.
FRIL can maintain quiescent primitive CD34(+) cells in vitro without cytokines, potentially extending ex vivo manipulation of human stem cells for clinical applications by preserving progenitor cells.
Not addressed in the paper.
FRIL maintains hematopoietic stem cell quiescence by regulating cell cycle factors like cyclin D3, CDK6, P27, and P53, aiding in stem cell preservation and differentiation.
FRIL, a lectin interacting with the Flt3 receptor, shows potential as a stem cell preservation factor, maintaining primitive hematopoietic progenitors in suspension culture for extended periods without inducing differentiation.
Not addressed in the paper.
FRIL preserves neural progenitor cells in vitro by inhibiting proliferation and differentiation, potentially extending applications to neural stem cell transplantation and treatment of neural degenerative diseases.

Related Questions

What is the role of galectin-3 in chronic liver diseases?10 answersGalectin-3 (Gal-3) plays a multifaceted role in the progression and pathogenesis of chronic liver diseases, acting as a critical mediator in inflammation, fibrosis, and cellular transformation processes. Gal-3 is upregulated in response to hepatotoxic insults and is associated with various liver conditions, including fatty liver diseases, nonalcoholic steatohepatitis (NASH), fibrosis, cholangitis, cirrhosis, and hepatocellular carcinoma (HCC). It directly interacts with the NOD-like receptor family, pyrin domain containing 3 (NLRP3), activating the inflammasome in liver macrophages and promoting fibrogenesis by activating hepatic stellate cells (HSCs). In acute-on-chronic liver failure (ACLF), Gal-3 is crucial for the hepatoprotection conferred by M2-like macrophages, primarily by inhibiting pyroptosis signaling pathways rather than necroptosis. This protective role is further supported by findings that the elimination of hepatitis C virus (HCV) via direct-acting antiviral (DAA) treatment results in a significant decrease in serum Gal-3 levels, suggesting that HCV infection induces an increase in this immune-regulatory protein. Moreover, Gal-3 is implicated in cirrhotic cardiomyopathy, a condition associated with cirrhosis, where it contributes to the pro-inflammatory mechanisms affecting cardiac function. Its expression levels in tissues and serum are significantly associated with the outcomes and risk of liver diseases, particularly HCC, where high Gal-3 expression correlates with worse survival rates and increased vascular invasion. Gal-3's involvement in primary biliary cholangitis (PBC) through its interaction with the NLRP3 inflammasome highlights its role in autoimmune liver diseases. Despite its significant roles, serum Gal-3 levels have not proven effective as a tumor marker for HCC surveillance among cirrhotic patients. Experimental models of cisplatin-induced liver toxicity have shown that inhibition of Gal-3 exacerbates liver damage, indicating its complex role in liver health. Given its involvement in various pathways of liver diseases, Gal-3 has attracted interest for therapeutic targeting in pharmaceutical research.
What are the future directions for research and development of lectin-based drug targeting systems?5 answersFuture directions for research and development of lectin-based drug targeting systems include exploring lectins' antimicrobial applications, understanding the complex "sugar code" represented by the glycome, and leveraging lectins for targeted cancer therapy. Lectins have shown promise in preventing infectious diseases, drug resistance, and as biomedical tools in drug design. Additionally, the development of lectin-based phototherapy for specific targeting of pancreatic cancer cells has demonstrated potential for clinical application. Integrating omics technologies like proteomics could further enhance the applications of lectins in drug delivery systems, antibiotic susceptibility testing, and vaccine adjuvant production. These advancements highlight the diverse and promising avenues for utilizing lectins in targeted drug delivery and therapy, paving the way for innovative approaches in precision medicine and disease treatment.
How do plant lectins interact with immune cells to modulate the immune response?5 answersPlant lectins interact with immune cells to modulate the immune response by inducing various effects. For instance, Orysata, a mannose-binding lectin, was found to induce cell aggregation in insect cells, mimicking immune processes. Additionally, jacalin, a dietary lectin, acts as a mitogen for peripheral blood mononuclear cells (PBMCs) and can polarize PBMCs towards either hindering or favoring tumor growth, depending on the duration of exposure. Plant lectins, known for their immunomodulatory properties in mammals, can upregulate antimicrobial peptides and cytokines in insect cells, influencing immune pathways. Understanding how lectins interact with immune cells provides insights into potential novel strategies for immune modulation and disease resistance in both plants and animals.
What is FLRT3?4 answersFLRT3, or Fibronectin leucine-rich transmembrane protein 3, is a cell adhesion molecule that plays a role in the progression of colorectal cancer (CRC). It has been found that overexpression of FLRT3 inhibits the process of epithelial-mesenchymal transition (EMT) and promotes apoptosis of CRC cells, while silencing FLRT3 expression has the opposite effect. Downregulation of FLRT3 is associated with a poor prognosis in CRC and is related to certain clinicopathological factors such as T stage, N stage, and E-cadherin level. Mechanistically, FLRT3 expression is downregulated by TGF-β secreted by cancer-associated fibroblasts (CAFs) through the activation of SMAD4, promoting aggressive phenotypes in CRC cells. FLRT3 also represses tumorigenesis and lung metastasis, and this effect can be reversed by LY2109761, a dual inhibitor of TGF-β receptor type I and II. Therefore, FLRT3 is a key molecule involved in the metastasis-suppressive function and potential treatment target for CRC.
What is application of plant lectin?4 answersPlant lectins have various applications in different fields. They have been used as biomarkers for cancer diagnosis and prognosis, with some lectins showing anti-neoplastic properties. Plant lectins also have potential therapeutic implications in cancer treatment, as they contribute to inducing cell death through autophagy and apoptosis. Additionally, plant lectins have been found to have antibacterial, antifungal, insecticidal, antiviral, and anticoagulant activities. They can also be used in crop improvement and glycobiology research. The interactions between lectins and carbohydrates can be characterized using various techniques, such as hapten inhibition assays, fluorescence titration, and surface plasmon resonance. Overall, plant lectins have a wide range of applications in cancer research, therapeutics, and other biological studies.
What are the applications of flax fiber?5 answersFlax fiber has various applications in different industries. It is used as a reinforcement material in composite plates, providing cost efficiency, improved mechanical properties, and environmental benefits. Flaxseed fiber is also utilized in the production of functional food concentrates, enriching them with dietary fibers, polyunsaturated fatty acids, and protein. In load-bearing structures and commercial commodities, flax fibers are used due to their wide range of applications and environmental sustainability. In the biomedical field, flax fibers are used for making surgical sutures and reinforcing polymer composites for tissue engineering. Additionally, flax fibers are being explored for their potential in optical fiber applications in the biomedical field.

See what other people are reading

What is the role of food consumption in EGCG supplementation?
5 answers
Food consumption significantly impacts the bioavailability and effects of EGCG supplementation. Studies show that taking EGCG without food leads to better systemic absorption, with significantly higher plasma levels compared to ingestion with food or in food matrices. High-dose EGCG supplements, commonly consumed during pregnancy, can adversely affect neurodevelopment by disrupting neural progenitor cell development in vitro, emphasizing the need for caution in such scenarios. Additionally, a combination of phytochemicals, including EGCG, in dietary supplements has shown positive effects on aging biomarkers, inflammation, and telomere length, highlighting the potential health benefits of combined phytochemical administration. Moreover, EGCG has been found to inhibit angiogenesis, a crucial process for follicle development, which raises concerns about the reproductive consequences of EGCG feeding supplementation.
What are the specific benefits of incorporating physical therapy into the treatment plan for cancer patients?
5 answers
Incorporating physical therapy into the treatment plan for cancer patients offers numerous benefits. Physical therapy helps address cancer-specific deficits such as fatigue syndrome, mortality, incontinence, lymphoedema, CIPN, anxiety, depression, and pain, as supported by international evidence-based guidelines. It plays a crucial role in enhancing functional limitations, cardiovascular health, balance retraining, and overall health optimization during cancer care. Additionally, physical therapy aids in improving quality of life, muscle strength, lung function, and daily living independence for cancer patients, ultimately contributing to their overall well-being and quality of life. By providing ongoing physical therapy interventions, patients can experience symptom relief, reduced risk of cancer recurrence, and improved response to cancer treatments, highlighting the importance of physical therapy in the comprehensive care of cancer patients.
What are the specific foods or nutrients that have been shown to improve breast cancer outcomes through diet?
5 answers
Specific foods and nutrients that have shown potential in improving breast cancer outcomes through diet include sulfur amino acids (SAAs), oxidizable polyunsaturated fatty acids (PUFAs), fiber, phytochemicals, antioxidants, and a Mediterranean diet rich in polyunsaturated fatty acids. Studies suggest that modifying nutrient availability, such as maintaining low SAAs and high PUFAs, can induce oxidative damage and cell death in breast cancer cells. Additionally, high adherence to the Mediterranean diet, which includes beneficial fatty acids, fiber, and phytochemicals, has been associated with reduced all-cause mortality in breast cancer survivors. Furthermore, a whole-food, plant-based diet has shown significant weight loss and improvements in cardiometabolic outcomes in breast cancer patients, potentially impacting cancer progression markers. These findings highlight the importance of dietary interventions in enhancing breast cancer outcomes.
What are the Clinical and preclinical states of CAR T cell therapy in vivo for solid tumors cancer?
5 answers
Clinical and preclinical states of CAR T-cell therapy for solid tumors cancer are evolving. Challenges like antigen heterogeneity and immunosuppressive tumor microenvironment hinder CAR T-cell efficacy in solid tumors. Strategies to enhance CAR T-cell function include incorporating co-stimulatory domains, developing armored CAR-T cells, and exploring other immune cells like CAR-NK and CAR-M cells. Novel CAR T-cell products like YTB323 show enhanced in vivo expansion and antitumor activity in preclinical models, with promising safety and efficacy profiles in clinical trials for r/r DLBCL. To overcome limitations such as restricted trafficking, hypoxic TME, antigen escape, and exhaustion, researchers are advancing CAR designs and combining therapies to optimize clinical efficacy in solid tumors.
How does the screening process negatively affect the randomization of rats in clinical trials?
5 answers
The screening process in clinical trials can negatively impact the randomization of participants, potentially leading to selection bias and compromised trial integrity. Screening measures, even when implemented correctly, may exclude eligible individuals and include ineligible ones. In the case of breast cancer screening trials, errors in trial design allowed for the mis-randomization of symptomatic women, affecting the balance between trial arms and potentially skewing efficacy results. Furthermore, selective exclusion criteria in randomized controlled trials (RCTs) often lead to the exclusion of certain patient populations, such as women, children, the elderly, and those with common medical conditions, which can impair the generalizability of trial results. Addressing these issues is crucial to ensure the scientific, regulatory, and ethical appropriateness of clinical trials.
Are there any anti cancer peptides found in emblica officinalis?
5 answers
Yes, anti-cancer peptides have been identified in Emblica officinalis. Research has shown that the ethanolic whole plant extract of Emblica officinalis exhibits cytotoxicity against various human cancer cell lines, including lung, colon, and neuroblastoma. Additionally, anti-cancer peptides derived from plant sources like Emblica officinalis have demonstrated effectiveness in combating cancer cells by enhancing treatment potency and reducing adverse effects associated with conventional therapies. These peptides are designed to interact with cancer cells, impacting their stability and effectiveness in targeting specific specimens. The presence of anti-cancer peptides in Emblica officinalis highlights its potential as a natural source of compounds with anti-cancer properties, offering promising avenues for further research and development in cancer treatment.
Is alcohol a Group 1 carcinogen?
4 answers
Yes, alcohol is classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC). This classification signifies that there is sufficient evidence of its carcinogenicity in humans. Alcohol consumption is linked to an increased risk of various cancers, including those of the oral cavity, pharynx, esophagus, liver, colorectum, and female breast. Acetaldehyde, a metabolite of alcohol, has been identified as a key factor in alcohol-related cancers, particularly in the digestive tracts where active alcohol and acetaldehyde metabolism occurs. While light-to-moderate alcohol consumption may have some health benefits, chronic alcohol drinking remains a significant risk factor for cancer development. The presence of carcinogenic compounds in alcoholic beverages further contributes to the overall cancer risk associated with alcohol consumption.
What is the importance of predicting patient outcomes in cancer treatment ?
5 answers
Predicting patient outcomes in cancer treatment is crucial for personalized therapy. By utilizing real-world data and statistical frameworks, individualized estimates of treatment efficacy and toxicity can be obtained, aiding clinicians in making informed decisions. Additionally, analyzing patient-reported outcomes (PROs) dynamically can serve as early predictors of treatment response, potentially improving response rates and adapting treatments effectively. Artificial intelligence (AI) and machine learning (ML) applications further enhance outcome prediction by providing accurate risk assessment, early diagnosis, prognosis estimation, and treatment selection. These predictive tools not only improve healthcare accuracy but also have the potential to enhance patient outcomes across various cancers, emphasizing the importance of individualized and data-driven care in cancer treatment.
5)What are the side effects and testing complexities of CAR-T cell therapy in solid tumors ?
4 answers
CAR-T cell therapy in solid tumors presents challenges such as severe toxic reactions, antigen escape, limited infiltration, and immunosuppression. The therapy's side effects include cytokine release syndrome, neurotoxicity, and on-target off-tumor toxicities due to the difficulty in identifying unique tumor antigen targets. Testing complexities arise from the hostile tumor microenvironment, which hinders CAR-T cell survival and function. Strategies like dual-targets, receptor system switches, and engineered CARs to shield inhibitory signaling molecules are being explored to enhance specificity, minimize toxicities, and improve CAR-T cell function in solid tumors. Combining CAR-T cell therapy with radiotherapy has shown promise in enhancing immunity, promoting infiltration, and improving the tumor microenvironment in solid tumors.
Date of this article?
4 answers
The article discussing the clinical trial results of MOXR0916, a novel OX40 agonist antibody, in patients with solid tumors, was published recently. Leddon et al. reported on the outcomes of adjuvant nivolumab in patients with locally recurrent head and neck squamous cell carcinoma. Batsios et al. explored deuterium metabolic imaging for monitoring TERT expression in tumor-bearing mice. Schmitd et al. investigated the role of perineural invasion in oral squamous cell carcinoma prognosis. The study by Vu et al. delved into the molecular mechanisms of CERK in KRASG12 mutant NSCLC. Mohapatra et al. elucidated the role of ECD in breast cancer tumorigenesis. Fushimi et al. uncovered the significance of MUC1-C in small cell lung cancer progression. Miyamoto et al. revealed a novel mechanism of bortezomib resistance in multiple myeloma. Coyne et al. conducted a phase II trial of cabozantinib in soft-tissue sarcoma. Dieci et al. explored immunotherapy in HR-positive/HER2-negative breast cancer. Stopsack et al. highlighted racial disparities in prostate tumor genomics. Boons et al. studied plasma cfDNA as a biomarker in neuroendocrine neoplasms. Lassman et al. investigated selinexor in recurrent glioblastoma. Hanna et al. assessed immune checkpoint inhibition in locoregionally recurrent SCCHN. Henriksen et al. evaluated ctDNA assessment in stage III CRC patients. Peng et al. developed an EP4 antagonist for prostate cancer immunotherapy.
Is wnt7b an oncogene in cancer?
4 answers
Yes, WNT7B has been implicated as an oncogene in cancer. Research has shown that WNT7B is overexpressed in certain cancer types, such as oral squamous cell carcinoma (OSCC) and lung squamous cell carcinoma (LUSC). The overexpression of WNT7B has been linked to promoting cancer cell invasion and contributing to the development of oral chronic inflammation and OSCC. Additionally, WNT7B knockdown has been shown to reduce cell proliferation and invasion ability in OSCC cells, further supporting its role as an oncogene in cancer. These findings suggest that WNT7B plays a significant role in tumorigenesis and could be a potential target for therapeutic interventions in pan-tissue-type originated tumors.