scispace - formally typeset
Search or ask a question

Answers from top 10 papers

More filters
Papers (10)Insight
CAF and phytosteryl ferulates probably have potentially anti-inflammatory properties.
Altogether, results show that this compound has potential as anti-inflammatory and anti-cancer agent.
In addition, this compound also exerts anti-inflammatory actions.
The formulation shows a statistically significant anti-inflammatory activity and is non-irritant to skin.
Therefore, it might be a candidate anti-inflammatory drug in patients with inflammatory bowel disease.
It is suggested that ES extract might be used to treatment anti-inflammatory skin disease.
Furthermore, SFN exhibits anti-inflammatory properties in vitro and in vivo.
This anti-inflammatory property might be the reason for its anti-tumor effects.
These results suggested that cavidine is a selective COX-2 inhibitor which possesses an anti-inflammatory activity.
These substances showed anti-inflammatory activity.

Related Questions

Why anti inflammatory drugs?4 answersAnti-inflammatory drugs are used because they can help prevent and treat age-related diseases, reduce cancer risk, interfere with the tumor microenvironment, and minimize tissue damage in inflammatory diseases. These drugs, including non-steroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids, have been widely used for their anti-inflammatory properties. NSAIDs, in particular, have been shown to reduce cell migration, increase apoptosis, and enhance chemo-sensitivity in cancer therapy. Additionally, plants have been studied for their potential as alternative anti-inflammatory agents with less toxicity. The development of new biological targets, such as NOD protein NLRP11, has also opened up possibilities for treating chronic inflammation diseases, autoimmune diseases, and tumors. Overall, anti-inflammatory drugs are important for managing inflammation and its associated complications in various diseases.
Is rOPINIRole an anti-inflammatory agent?5 answersRopinirole is not an anti-inflammatory agent. It is a nonergoline dopamine agonist primarily used for the treatment of Parkinson's disease and restless legs syndrome. Studies have shown that ropinirole effectively reduces the motor symptoms of restless legs syndrome and improves sleep quality. Additionally, ropinirole has been proven to be effective in both monotherapy and combination therapy for idiopathic Parkinson's disease, improving bradykinesia, rigor, tremor, daily living abilities, and depressive mood. However, there is no evidence to suggest that ropinirole has anti-inflammatory properties or is used as an anti-inflammatory agent.
Can paracetamol be used for the treatment of inflammation give reason?10 answers
Is Boroline anti inflammatory?10 answers
Is Safi anti inflammatory?10 answers
What are anti inflammatory analgesic drugs?10 answers

See what other people are reading

What is the role of cholangiocyte in NAFLD setting?
5 answers
Cholangiocytes play a crucial role in non-alcoholic fatty liver disease (NAFLD) by contributing to disease progression through various mechanisms. In NAFLD patients, cholangiocytes can be affected, leading to ductular reaction and activation of the progenitor cell compartment, ultimately promoting portal fibrosis and disease advancement. Additionally, cholangiocytes are involved in modulating bile composition and transportation, and their secreted cytokines (cholangiokines) can drive ductular cell proliferation, inflammation, fibrosis, and even carcinogenesis in liver diseases. Furthermore, in the context of the "Gut-Liver Axis," changes in gut bacterial homeostasis or impaired intestinal barrier functions can expose cholangiocytes to inflammatory responses triggered by gut-derived microorganisms, exacerbating liver fibrosis. Understanding the intricate role of cholangiocytes in NAFLD pathogenesis is essential for developing targeted therapies.
What are the specific factors that affect the 3D printed PCL polymer scaffolds in bone tissue engineering?
5 answers
The mechanical properties of 3D printed PCL polymer scaffolds in bone tissue engineering are influenced by various factors. Scaffold design parameters such as height and internal structure play a crucial role. Additionally, the incorporation of nano-hydroxyapatite (nHAp) into PCL scaffolds affects their mechanical and biological performance. The pore size and internal structure of the scaffold also impact osteoblast performance, with staggered scaffolds showing better support for cellular activities. Furthermore, the addition of bioceramics like hydroxyapatite (HA) and β tricalcium phosphate (β-TCP) along with carbon nanotubes (CNTs) in PCL-based composite scaffolds influences gene expression related to osteogenesis and inflammatory responses, ultimately affecting tissue formation and mineralization. These factors collectively determine the effectiveness of 3D printed PCL polymer scaffolds in bone tissue engineering applications.
Is nn hexane a pestisied?
5 answers
n-Hexane is not a pesticide itself but is used in the production of pesticide-grade solvents. Various methods have been developed to purify n-hexane to meet the stringent requirements of pesticide residue grade solvents. These purification processes involve steps such as decoloring, rectifying, and filtering to achieve ultra-high purity levels with low impurity content, making the n-hexane suitable for pesticide residue analysis. Additionally, the industrialization process of producing pesticide residue grade n-hexane has been optimized and validated by several government organizations in China, confirming its competitiveness with imported n-hexane products. Therefore, while n-hexane itself is not a pesticide, it plays a crucial role in the production of high-quality solvents used in pesticide applications.
What are the common methods used to prepare chitosan-based film-forming solutions for in vitro testing of antifungal properties?
4 answers
Chitosan-based film-forming solutions for in vitro testing of antifungal properties are commonly prepared using methods such as nebulization, mixing with organic acids followed by gassing process and solution preparation, and by mixing chitosan with other compounds like 2-amino-4,5,6,7-tetrahydrobenzo[b]thiophene-3-carbonitrile (6CN). These solutions can also involve the incorporation of additional antifungal agents like nystatin and propolis dispersed in glycerin. The film-forming solutions are characterized by their physicochemical properties using techniques like FTIR, DSC, TGA, XRD, and SEM. The resulting films have shown effective antifungal activity against various fungi species, making them promising candidates for topical antifungal formulations.
Why do plants produce gamma-linolenic acid (GLA)?
4 answers
Plants produce gamma-linolenic acid (GLA) as it is a bioactive fatty acid with various health benefits. GLA is an omega-6 polyunsaturated fatty acid that plays a crucial role in inflammation modulation. GLA is known for its anti-inflammatory properties, which can be beneficial in conditions involving inflammation. Additionally, GLA is essential for the production of prostaglandins and leukotrienes, which are important in inflammatory responses and immune regulation. Some plant species, such as Ribes taxa and cultivars, are particularly rich in GLA, making them valuable sources for obtaining this beneficial fatty acid. Therefore, plants produce GLA to contribute to the synthesis of anti-inflammatory compounds and support overall health and well-being.
How does LPA regulate cell recruitment and migration?
4 answers
Lysophosphatidic acid (LPA) plays a pivotal role in cell recruitment and migration through various mechanisms involving its interaction with specific G-protein coupled receptors (GPCRs) and subsequent activation of downstream signaling pathways. LPA induces the expression and secretion of inflammatory cytokines such as CXCL13, which is crucial for T cell recruitment in inflammatory diseases, highlighting its role in the inflammatory response and leukocyte recruitment. It is produced by vascular endothelial and other stromal cells within lymph nodes, regulating lymphocyte migration across high endothelial venules (HEVs) and within the interstitial spaces, demonstrating its importance in immune cell trafficking. In the context of regenerative engineering, LPA enhances human mesenchymal stem cells (hMSCs) migration towards wound sites, indicating its potential in tissue repair and regeneration. This process involves ATX-mediated LPA production, which increases hMSC migration through LPA receptor 1/3 activation, calcium influx, and PKC phosphorylation, leading to cytoskeletal rearrangement and E-cadherin reduction. LPA also directly influences T cell motility within lymph nodes, with specific LPA receptors like LPA2 playing non-redundant roles in intranodal T cell migration, suggesting receptor-specific functions in immune surveillance. In cancer, LPA promotes tumor cell migration and invasion through the activation of the LPA1 receptor, leading to Rho kinase-mediated phosphorylation of nonmuscle myosin II, which is essential for cytoskeleton-related signaling in tumor cell migration. Similarly, in oral carcinoma cells, LPA stimulates migration through LPAR3, mediated by PKC activity and possibly EGFR transactivation, indicating a complex signaling network that varies among cell types. The Hippo-YAP signaling pathway, known for its role in cancer, is regulated by LPA through GPCR ligands, affecting cell migration and invasion in epithelial ovarian cancer cells via a novel signaling pathway involving G13, RhoA-ROCK, and PP1A, leading to YAP dephosphorylation and nuclear translocation. Furthermore, LPA signaling through LPA4 is essential for the radial migration and morphogenesis of newborn cortical neurons, indicating its role in brain development by regulating the actin cytoskeleton and stabilizing proteins like filamin A. In pancreatic cancer, LPA induces FAK and paxillin activation, enhancing cell motility through LPA1–3 receptors, which is crucial for metastasis. Lastly, in rat aortic smooth muscle cells, LPA stimulates migration via a Gi-protein-coupled LPA1 receptor and a PTX-sensitive Gi/p38MAPK pathway, showcasing the diversity of LPA's effects across different cell types.
IS hyaluronic acid beneficial for helth?
5 answers
Hyaluronic acid (HA) is indeed beneficial for health due to its versatile properties. HA plays a crucial role in wound healing, acts as a scaffold in regenerative medicine, and exhibits anti-inflammatory and water-binding capacities in various applications, including periodontal therapyand lung disease management. Its biodegradability, biocompatibility, and nonimmunogenicity make it valuable in biomedical fields like tissue engineering and drug delivery. Clinical evidence supports HA's effectiveness in tissue repair, wound healing, and improving quality of life in patients with periodontal issues. Moreover, HA's ability to retain water contributes to maintaining fluid balance in tissues, making it beneficial in treating lung diseases. Overall, HA's diverse benefits make it a valuable biomaterial with significant potential for enhancing health outcomes.
What are asc specks?
4 answers
ASC specks are micron-sized complexes formed by the polymerization of the inflammasome adaptor protein ASC. These specks play a crucial role in inflammasome activation, particularly in the maturation of IL-1 cytokines and caspase-1 activation. ASC specks are released into the extracellular space during pyroptosis, a lytic cell death process, and can accumulate in tissues during chronic inflammation. They have been linked to the propagation of inflammasome activation-associated α-synuclein pathology in Parkinson's disease, exacerbating neurodegeneration and motor deficits. ASC specks are also used as a readout for inflammasome activation in various diseases, aiding in the identification of inflammasome-activated monocytes in patient samples. Overall, ASC specks are key players in the inflammatory response and have implications in various pathological conditions.
How is mTOR inhibition proposed to improve longevity?
10 answers
Inhibition of the mechanistic target of rapamycin (mTOR), specifically the mTORC1 complex, has emerged as a promising strategy to improve longevity and healthspan across various model organisms. The mTOR pathway plays a crucial role in promoting anabolic processes and inhibiting catabolism, and its downregulation has been associated with lifespan extension, mimicking the effects of calorie restriction. Rapamycin, an FDA-approved therapeutic, has been shown to extend the lifespan and survival of wild-type mice and mouse models of human diseases by inhibiting mTORC1. However, concerns regarding the side effects of rapamycin, such as impacts on immunity and metabolism, have limited its use as a geroprotective therapy. Recent studies have suggested that these adverse effects are primarily due to off-target inhibition of mTORC2, and strategies like intermittent rapamycin treatment, specific dietary regimens, and the development of new molecules targeting mTORC1 more selectively, could mitigate these concerns. For instance, the discovery of TKA001, a novel small molecule inhibitor of mTOR, through machine learning, has shown potential in extending lifespan in C. elegans and inhibiting human cancer cell proliferation, indicating its capability to slow aging in vivo. Moreover, targeting mTORC1 in specific tissues, such as neurons, has been demonstrated to uncouple the longevity benefits from the negative effects on growth and reproduction, highlighting the potential for tissue-specific aging therapeutics. Additionally, the role of mTORC1 inhibition in promoting longevity has been linked to the activation of anti-aging mechanisms and increased production of hydrogen sulfide (H2S), which enhances protein persulfidation, a protective modification against oxidative stress. In summary, mTOR inhibition, particularly through selective targeting of mTORC1, offers a multifaceted approach to improving longevity by balancing metabolic processes, enhancing stress resistance, and reducing the incidence of age-related diseases, while minimizing adverse side effects.
Are there studies showing obesity is related to proinflammatory cytokines and reactive oxygen species?
5 answers
Obesity has been consistently linked to proinflammatory cytokines and reactive oxygen species (ROS) in various studies. Research has shown that individuals with normal weight obesity (NWO) exhibit higher levels of proinflammatory markers like C-reactive protein (CRP), interleukin 6 (IL6), and tumor necrosis factor-alpha (TNFα). Additionally, studies comparing obesity management strategies, such as sleeve gastrectomy (SG) and lifestyle interventions (LS), have demonstrated reductions in inflammatory cytokines like IL-6 and TNF-a post-intervention. Furthermore, investigations into oxidative stress in obese individuals have revealed increased levels of ROS and proinflammatory markers like TNFα and IL-6 in plasma and peripheral blood mononuclear cells (PBMCs). These findings collectively emphasize the association between obesity, proinflammatory cytokines, and oxidative stress markers.
What are the most common underlying causes of chronic kidney disease (CKD)?
5 answers
The most common underlying causes of chronic kidney disease (CKD) include diabetes mellitus, hypertension, glomerulonephritis, polycystic kidney disease, malformations, autoimmune diseases, obstructions, infections, high cholesterol, chronic pharmaceutical use, and cardiovascular disease. CKD is a global health burden with significant economic costs to health systems, often exacerbated by risk factors such as diabetes and hypertension. The onset of CKD is associated with oxidative stress, inflammation, impaired antioxidant defense, and molecular mechanisms affecting metabolic homeostasis, autophagy, apoptosis, and genetic conditions like gene polymorphisms. Understanding the diverse etiologies of CKD is crucial for effective disease management, which may involve multidisciplinary collaboration and nutritional therapy.