scispace - formally typeset
Search or ask a question

What is the effect of phytol on tumor growth? 


Best insight from top research papers

Phytol, a phytochemical derived from plants, has been shown to have various effects on tumor growth. It has been found to inhibit cancer initiation, promotion, progression, and metastasis by employing antioxidant and anti-inflammatory effects mediated via NF-kκB, Nrf2, and AP-1 signaling . Additionally, phytol induces apoptosis in tumor cells and inhibits cancer growth . It has also been observed that phytol induces autophagy and activates the cytoprotective Nrf2 pathway in cancer cells . Furthermore, phytol has been reported to enhance the activity of natural killer cells and promote macrophage functions in immunity . In a mouse skin initiation-promotion model, phytol has been found to have weak tumor promoter activity compared to 12-O-tetradecanoyl phorbol-13-acetate (TPA) . Overall, phytol exhibits potential as a chemopreventive and chemotherapeutic agent in cancer treatment.

Answers from top 5 papers

More filters
Papers (5)Insight
The effect of phytol on tumor growth is mentioned in the paper. Phytol has a weak tumor promoter activity compared to TPA and is a non-TPA-type tumor promoter in the mouse skin carcinogenesis model.
Open accessJournal ArticleDOI
15 Apr 2018
18 Citations
The provided paper does not mention the effect of phytol on tumor growth. The paper focuses on the inhibitory effect of phytol on cellular senescence in HaCaT keratinocytes.
The provided paper does not mention the effect of phytol on tumor growth.
The provided paper does not mention the effect of phytol on tumor growth. The paper discusses the role of phytochemicals in preventing cancer initiation, promotion, progression, and metastasis, but does not specifically mention phytol or its effects on tumor growth.
The paper does not provide information about the effect of phytol on tumor growth.

Related Questions

Which foods help in cancer treatment?4 answersVarious foods have been identified for their potential role in cancer treatment. Bioactive foods and functional foods derived from plant, animal, and microbial sources have shown significant anticancer effects with diverse mechanisms of action, such as antimetastatic, antioxidant, and induction of apoptosis. Additionally, a diet rich in fruits, vegetables, whole grains, nuts, and legumes, while low in red and processed meats, is recommended for cancer prevention. These foods provide essential nutrients, vitamins, minerals, and protective phytochemicals that help defend the body against cancer by diffusing carcinogens, eliminating toxins, facilitating cellular repair, inhibiting cancer cell growth, and inducing cell apoptosis. Functional foods, beyond basic nutrition, have been highlighted for their potential to prevent and treat various types of carcinoma, making them crucial in the fight against cancer.
What is the role of polyphenols in cancer treatment?5 answersPolyphenols play a significant role in cancer treatment by counteracting drug resistance, enhancing the efficacy of chemotherapy, and reducing side effects. They have the potential to inhibit drug efflux transporters, alter immunological responses, trigger apoptosis in cancer cells, and inhibit tumor growth and metastasis. Polyphenols can be used as anticancer drugs alone or in combination with conventional drugs, leading to more efficient therapies with fewer side effects. To improve the therapeutic potential of polyphenols, nanotechnology approaches have been applied to encapsulate these compounds, resulting in enhanced antitumor activity and decreased tumor growth. Plant polyphenols can simultaneously affect multiple processes involved in malignant cell properties, making them a promising option for cancer treatment. Flavonoids, a type of polyphenol, have shown various mechanisms of action in cancer prevention and treatment, including carcinogen deactivation, apoptosis, and suppression of multidrug resistance.
How can nutrition help to prevent cancer?3 answersNutrition plays a crucial role in preventing cancer. Unhealthy diets and behaviors increase the risk of cancer onset, while healthy dietary practices can help avoid 30-40% of cancer cases. Following a diet high in fiber and soy, limiting the consumption of fats, particularly saturated fatty acids, and maintaining a healthy body weight can increase overall survival after cancer diagnosis. Cancer-protective foods such as whole grains, fruits, non-starchy vegetables, fish, and dairy products contain nutrients and bioactive components that enhance immunologic function, induce apoptosis, and donate electrons to free radicals, thus preventing cancer. A balanced plant-based diet with limited amounts of fast foods, sugar-sweetened drinks, red meat, and alcohol is recommended for enhancing health and reducing the risk of cancer and other noncommunicable diseases. Understanding how nutrition influences cancer risk profiles and communicating and supporting individuals in making dietary and lifestyle changes are important for preventing cancer.
What are the Natural product anti cancer drug?5 answersNatural products have been widely used as a source of therapeutic ingredients for the development of anti-cancer drugs. Several plant-based drugs, such as vincristine, etoposide, topotecan, and paclitaxel, have shown promising anti-tumor activity and are currently used in clinical practice. Additionally, natural products with immunomodulatory properties, such as curcumin, resveratrol, zerumbone, quercetin, genistein, and betulinic acid, are being explored as adjuvant therapies in cancer treatment. Natural products and their derivatives have demonstrated potential anti-cancer properties, including cytotoxicity, proliferation inhibition, apoptosis induction, metastasis retardation, angiogenesis suppression, and improved chemotherapy. Plant-derived anti-cancer agents in late preclinical development or early clinical trials include taxol, vincristine, vinblastine, topotecan, irinotecan, etoposide, paclitaxel, and docetaxel. Furthermore, the conjugation of natural products with other bioactive molecules has led to the development of novel, bifunctional drugs for cancer therapy, such as ADCs, PDCs, ApDCs, PROTACs, and AUTOTACs.
What are the cancer chemo preventive effects of certain herbal medicines?5 answersCertain herbal medicines have shown cancer chemo preventive effects. Studies have reported the beneficial effects of herbal medicines on the survival, immune modulation, and quality of life of cancer patients when used in combination with conventional therapeutics. Natural compounds in herbal medicines have been found to inhibit tumor formation and are well tolerated by patients at high doses. Herbal extracts, such as SH003, have been effective in treating lung and respiratory diseases and reducing symptoms induced by particulate matter. Herbal therapeutics have also shown potential in preventing and treating ROS-induced cancer through their chemopreventive and antioxidant properties. Additionally, natural products and their combinations with chemotherapeutics have been found to mediate anticancer effects by modulating signaling pathways involved in apoptosis, cellular proliferation, and immune response. The concomitant use of herbal medicines with chemotherapy has been demonstrated to be beneficial in multiple malignant tumors, improving the quality of life of patients and alleviating side effects.
What are the effects of combining a medical plant extract with a cancer drug in treating breast cancer?5 answersCombining a medical plant extract with a cancer drug in treating breast cancer can have positive effects. Natural compounds extracted from plants, such as green tea, berberine, thymoquinone, and cannabidiol, have shown promising results in inhibiting cancer cell growth and inducing apoptosis. These compounds have also demonstrated anti-inflammatory and hormonal modulation properties, making them potential adjuvant treatments for breast cancer. Additionally, the combination of phytomedicine or natural products with synthetic drugs has the potential to enhance therapeutic efficacy and overcome drug resistance. Various herbal compounds, including Ginseng, garlic, black cohosh, turmeric, and green tea, have been identified as having anti-cancer properties and may be effective in combination with conventional treatment regimens. However, further clinical trials and studies are needed to provide more evidence of their medical benefits.

See what other people are reading

How is antioxidant capacity important for cancer prevention?
5 answers
Antioxidant capacity plays a crucial role in cancer prevention by maintaining redox homeostasis and counteracting the damaging effects of reactive oxygen species (ROS) and reactive nitrogen species (RNS). While antioxidants are known for their ability to quench free radicals and reduce oxidative stress, their role in cancer is complex and context-dependent. Studies suggest that antioxidants can interact with various cancer targets, inhibiting cell growth, proliferation, and metastasis, as well as neutralizing free radicals involved in cancer initiation and progression. However, the dual nature of antioxidants is evident, as they can also potentially promote cancer under certain conditions. Understanding the intricate balance of antioxidant activity in cancer cells is essential, as exploiting vulnerabilities related to antioxidant dependence may offer therapeutic opportunities for selective cancer cell elimination.
When is CXCR4 receptor discovered?
5 answers
The CXCR4 receptor was first identified as a chemokine receptor in 1996, marking its discovery as a key co-factor for T-lymphotropic HIV infection and subsequently sparking significant research interest in its role in HIV pathophysiology. CXCR4's importance extends beyond HIV, as it has been found to be a crucial chemokine receptor in the metastasis of various cancers, including breast cancer. Recent studies have highlighted the significance of CXCR4 in physiological and pathological processes, emphasizing its interaction with the chemotactic ligand CXCL12 in cancer progression and metastasis. The continuous exploration of CXCR4's functions has led to the development of potent small molecule inhibitors targeting CXCR4, such as HFX51116, showing promising therapeutic potential in cancer treatment and hematopoietic stem cell mobilization.
What are the resideus of akt ative site?
5 answers
The active site of the Akt protein involves critical regulatory residues, specifically threonine 308 and serine 473, which undergo phosphorylation to induce a conformational change leading to Akt activation. Additionally, Akt activation is crucial for cell proliferation, survival, and metabolism. Interestingly, ATP-competitive inhibitors of Akt can paradoxically increase the phosphorylation of threonine 308 and serine 473 by stabilizing a conformation where these sites are inaccessible to phosphatases. Moreover, Akt activation can be modulated by posttranslational modifications, such as phosphorylation at these key residues, which play a role in oncogenesis and therapeutic resistance. Therefore, understanding the residues at the Akt active site and their phosphorylation status is essential for elucidating the mechanisms underlying Akt activation and its implications in cancer biology.
What are the potential health benefits of Echinacea purpurea?
4 answers
Echinacea purpurea offers a range of potential health benefits based on recent research. It has been found to stimulate the immune system, reduce blood sugar levels, anxiety, and inflammation, improve skin health, possess anti-cancer properties, and potentially act as an alternative to chlorhexidine. Additionally, Echinacea extracts have shown promise in treating chemotherapy-induced neuropathic pain through their alkamides and polyphenols content. The plant's immunomodulating properties make it useful in topical preparations for wound treatment, with extracts demonstrating antioxidant, antiaging, and wound healing effects. Furthermore, Echinacea extracts have been shown to possess anti-inflammatory properties, reducing pro-inflammatory cytokines and reactive oxygen species production, potentially aiding in the treatment of inflammatory diseases. Overall, Echinacea purpurea presents a versatile herbal remedy with various health benefits.
How does the disruption of circadian rhythms affect pancreatic cancer?
5 answers
Disruption of circadian rhythms has significant implications for pancreatic cancer progression. Studies have shown that circadian clock dysfunction, as evidenced by attenuated or dysregulated clock genes, accelerates tumor growth, worsens survival rates, and promotes chemoresistance in pancreatic cancer models. Furthermore, disruption of the circadian rhythm can lead to an exacerbated fibrotic phenotype in tumors, including pancreatic cancer, through mechanisms involving cancer-associated fibroblasts and the TGF-β signaling pathway. Epidemiological studies also suggest a link between circadian disruption, light at night exposure, and an increased risk of pancreatic cancer, highlighting the potential role of circadian disturbances in carcinogenesis. Additionally, circadian disruption can enhance tumor growth by promoting immunosuppression through the accumulation of myeloid-derived suppressor cells within the tumor microenvironment.
Is BLTR the same as cysLT1R?
4 answers
BLTR and cysLT1R are not the same. BLTR refers to the leukotriene B4 receptor, while cysLT1R specifically stands for the cysteinyl leukotriene receptor 1. The research papers provide insights into the distinct roles of cysLT1R in various pathophysiological conditions. Specifically, cysLT1R has been implicated in inflammatory responses in sepsis, platelet-induced breast tumor cell survival, and Alzheimer's disease (AD) pathogenesis. The studies highlight the significance of cysLT1R as a potential therapeutic target in conditions like sepsis, cancer metastasis, and AD. Therefore, while BLTR and cysLT1R are both involved in inflammatory processes, they represent different receptors with unique functions and implications in various diseases.
What specific proteins contribute to organ-specific metastasis liver cancer?
5 answers
Ferritin light chain (FTL), lactate dehydrogenase A (LDHA), and long-chain-fatty-acid-CoA ligase 1 (ACSL1) are specific proteins that contribute to organ-specific metastasis in liver cancer. Additionally, Actin Related Protein 2/3 Complex Subunit 2 (ARPC2) has been identified as highly expressed in extracellular vesicles (EVs) derived from metastatic hepatocellular carcinoma (HCC) cells, enhancing cancer cell growth and metastasis. Moreover, the activation of SREBP2-dependent cholesterol biosynthesis pathway, involving the up-regulation of SREBP2, is crucial for the colonization and growth of metastatic colorectal cancer (CRC) cells in the liver, highlighting a specific cholesterol metabolic aberration in CRC liver metastasis. These proteins play essential roles in driving organ-specific metastasis in liver cancer, offering potential targets for therapeutic interventions.
What the noninvasive method for tumor measurement using luciferase?
5 answers
A noninvasive method for tumor measurement utilizing luciferase is bioluminescence imaging (BLI). BLI allows for sensitive detection of tumors and disease progression. It provides a qualitative analysis of tumor burden and treatment response, enabling the calculation of a luminoscore for comparing animals in experiments. Additionally, BLI can be used for assessing engineered tumor models in vivo, such as in the chick chorioallantoic membrane assay, allowing for specific detection and quantitative monitoring of tumors over time. This method has been further enhanced by combining BLI with tomographic imaging of the sodium iodide symporter (NIS), providing improved spatial resolution, precise tumor localization, and noninvasive quantification of radiotracer uptake. Overall, BLI offers a valuable tool for noninvasive and comprehensive evaluation of tumor responses to therapies.
What are the epigenetic changes associated with ovarian cancer ?
10 answers
Epigenetic changes play a crucial role in the development, progression, and therapeutic resistance of ovarian cancer (OC). These changes include DNA methylation alterations, histone modifications, and RNA modifications, which affect gene expression and activity without altering the DNA sequence itself. DNA methylation patterns differ significantly between ovarian cancer cells with low and high metastasis potentials, with hypermethylation and downregulation of specific genes like SFRP1 and LIPG being associated with worse prognosis and increased metastasis potential. RNA modifications, such as m6A, m1A, and m5C, also contribute to the complexity of OC pathogenesis by affecting mRNA stability, translation efficiency, and the nuclear export of RNAs, thereby influencing the disease's occurrence and development. The use of DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi) has shown promise in targeting the ovarian carcinoma epigenome, enhancing immune signaling and potentially improving therapeutic outcomes. These epigenetic therapies aim to reverse aberrant epigenetic modifications in cancer cells and the tumor microenvironment, making OC cells more susceptible to standard chemotherapy. Furthermore, RNA methylation modifications have been identified as significant factors in OC progression and metastasis, offering potential targets for therapy. Research has demonstrated that blood DNA methylation alterations are highly consistent with tissue DNA methylation alterations in OC, suggesting that cell-free DNA (cfDNA) methylation could serve as a reliable biomarker for early detection. Epigenetic modifications also contribute to chemoresistance in OC through various mechanisms, including upregulating multidrug resistance proteins and remodeling the tumor microenvironment. Studies have identified epigenetic signatures associated with platinum drug resensitization, highlighting the potential of epigenomic targeting to improve therapeutic outcomes in platinum-resistant and recurrent OC by affecting DNA repair and antitumor immune responses. Lastly, epigenetic changes, being stable and disease subtype-specific, hold promise as biomarkers for diagnosing and treating OC, with the potential for serum-based detection offering an efficient and accurate diagnostic approach.
What are the haemotoxicity of Carbon Dots in vitro?
4 answers
The haemotoxicity of Carbon Dots (CDs) and Carbon Quantum Dots (CQDs) in vitro, as derived from the provided contexts, reveals a nuanced understanding of their interaction with biological systems, particularly concerning their impact on blood components. The studies collectively indicate that while CDs and CQDs exhibit promising biocompatibility and potential therapeutic applications, their haemotoxic effects, when present, are minimal and highly dependent on specific conditions such as concentration, cell type, and exposure duration. From the provided contexts, only one directly addresses haemotoxicity, indicating that the antibacterial functional CDs prepared from chitosan derivatives exhibit low haemolytic effects, suggesting minimal haemotoxicity to human normal hepatocytes. This finding is crucial as it highlights the potential of CDs for biomedical applications, including antibacterial treatments, without significant adverse effects on blood cells. The low haemolytic effect implies that these CDs do not significantly disrupt the integrity of red blood cells, a critical factor in evaluating the safety of nanomaterials for medical applications. Other contexts, while not directly addressing haemotoxicity, contribute to the overall understanding of CDs' biocompatibility and safety profile. For instance, studies have shown that CDs possess high biocompatibility, with no significant cytotoxic effects observed in various cell types, including liver cells, macrophages, and breast cancer cell lines, under certain conditions. Additionally, the therapeutic application of CQDs in immunomodulation without cytotoxicity or in vivo damage further supports their safety. Moreover, the absence of significant toxic effects in acute and subacute toxicity experiments with mice underscores the potential non-toxic nature of CDs. In summary, the haemotoxicity of CDs and CQDs appears to be minimal, with specific studies indicating low haemolytic effects under certain conditions. This, combined with their high biocompatibility and safety profile in various biological systems, suggests that CDs and CQDs could be safe for biomedical applications, although the specific conditions under which they are used must be carefully considered to ensure safety.
Should PBMCs contain Porphobilinogen?
5 answers
Porphobilinogen is not typically found in peripheral blood mononuclear cells (PBMCs). Instead, enzymes involved in the heme biosynthetic pathway, such as porphobilinogen synthase (PBGS) and porphobilinogen deaminase (PBGD), are present in PBMCs. PBGS is essential for tetrapyrrole biosynthesis and can utilize different metal ions like zinc and magnesium for catalytic function. On the other hand, PBGD activity has been linked to certain conditions like metastatic spread in cancer patients, with elevated levels observed in some cases. Therefore, while PBMCs do not contain porphobilinogen, they do harbor enzymes like PBGS and PBGD that play crucial roles in heme synthesis and related pathways.