scispace - formally typeset
Search or ask a question

Answers from top 10 papers

More filters
Papers (10)Insight
Presumably chicken ghrelin may function as an anti‐inflammatory factor.
The study suggests that PUE may be a potential anti-inflammatory agent defense againstMGinfection in chicken.
The results suggested that baicalin acts as a potential anti-inflammatory agent against MG infection in chicken and DF-1 cells.
It is, therefore, endowed with anti-inflammatory properties.
The potential anti-inflammatory activity against egg albumin has obtained.
Phytochemicals that exhibit anti-inflammatory mechanisms that reduce sustained inflammation could be therapeutic candidates for various inflammatory diseases.
japonicum has a sufficient anti-inflammatory effect.
Furthermore, the regression of chicken POF seems to be an inflammatory event similar to luteolysis of the mammalian corpus luteum.
These substances showed anti-inflammatory activity.
These results suggested that baicalin could effectively inhibit MG-induced immune impairment and alleviated inflammatory responses and apoptosis in the chicken BF.

Related Questions

Is vitamin K anti-inflammatory?4 answersVitamin K has been shown to have anti-inflammatory properties and can suppress signaling pathways involved in inflammation. It can also inhibit the activation of the NLRP3 inflammasome, a complex involved in the pathogenesis of inflammatory diseases. Various forms of exogenous vitamin K, such as K1, K3, and MK-5, have demonstrated anti-inflammatory effects in macrophages. Additionally, vitamin K can act as an antioxidant, protecting against the generation of reactive oxidative species. Clinical evidence suggests that a high vitamin K status can have a protective role in age-related diseases associated with inflammation and mineralization processes. However, a study on rheumatoid arthritis patients found that vitamin K1 supplementation did not significantly reduce inflammation or disease severity. Overall, while vitamin K has shown potential as an anti-inflammatory agent, further research is needed to fully understand its mechanisms and effectiveness in different inflammatory conditions.
What effect have eggs on inflammation?5 answersEgg consumption does not have a significant effect on inflammation, as indicated by a meta-analysis of clinical trials. However, eggs contain bioactive compounds that can influence pro- and anti-inflammatory pathways, and the effects of egg consumption on inflammation vary across different populations. Chronic helminth infection with Schistosoma mansoni eggs has been shown to protect against allergic airway inflammation in mice. Additionally, a study found that a carbohydrate restricted diet combined with egg consumption increased HDL cholesterol levels and improved atherogenic dyslipidemia. Eggs are considered a functional food that can affect proinflammatory and anti-inflammatory pathways, potentially protecting against metabolic syndrome by increasing HDL cholesterol levels and reducing inflammation.
How do you reduce inflammation of GERD?10 answers
Is boiled chicken good for high blood pressure?10 answers
Is chicken liver inflammatory?10 answers
What's the best natural anti inflammatory for arthritis?10 answers

See what other people are reading

How does Digeorge syndrome affect the brain and cognitive development in children?
5 answers
DiGeorge syndrome (DGS) impacts brain development and cognitive function in children through various mechanisms. The syndrome, caused by microdeletions in the 22q11.2 region, can lead to oxidative stress and neuroinflammation, affecting mitochondrial functionality and cortical connectivity, resulting in neurocognitive impairment. Additionally, cognitive and behavioral issues in DGS should be addressed through timely multidisciplinary interventions to improve communication and social skills. Mutations in the TBX1 gene, associated with DGS, can lead to craniofacial defects and aberrant neural crest cell migration, impacting cognitive and psychiatric outcomes. Furthermore, altered synaptic plasticity may also contribute to cognitive disorders in DGS individuals. Overall, a comprehensive approach involving antioxidant therapy and multidisciplinary interventions is crucial in managing the cognitive impact of DGS in children.
What are the underlying biological factors that contribute to sarcopenia?
5 answers
Sarcopenia, the progressive loss of skeletal muscle mass and function, is influenced by various biological factors. Smoking and chronic excess alcohol consumption can lead to muscle atrophy and mitochondrial dysfunction, impacting muscle protein synthesis rates. Malnutrition in gastrointestinal diseases can impair muscle mass and function, potentially leading to sarcopenia. Mitochondrial dysfunction, particularly related to oxidative phosphorylation and mitochondrial ribosomal proteins, plays a significant role in sarcopenia development. Additionally, genetic factors, lifestyle choices, and age-related changes contribute to the complex pathophysiology of sarcopenia. Understanding the interplay between these factors at the cellular and molecular levels is crucial in comprehending the mechanisms underlying sarcopenia.
Why is bpa used?
5 answers
Bisphenol A (BPA) is used due to its wide application in various industries. It is utilized in the production of polycarbonate plastics, epoxy resins, and thermal papers, providing elasticity to products like canned food linings, plastic containers, dental fillings, and medical devices. However, concerns arise from its endocrine-disrupting properties, potentially contributing to the global obesity epidemic and metabolic disorders by promoting inflammation and cytokine release in fat cells. Studies also highlight the toxic influence of BPA on reproduction, leading to its inclusion in substances requiring special supervision. Despite its prevalent use, the detection of BPA in the environment is crucial for health protection, with electrochemical methods gaining interest for rapid and cost-effective monitoring.
What specific molecular pathways are activated in the ileum of mice after intraperitoneal LPS injection?
5 answers
After intraperitoneal LPS injection in mice, the ileum exhibits activation of specific molecular pathways. The injection of LPS leads to an increased inflammatory response, including elevated levels of IL-1β and IL-18 in serum, and the activation of the NLRP3 inflammasome in the intestine. Additionally, the expression of Slit2 and Robo4 decreases significantly in the intestine, while VEGF expression increases. Furthermore, there is a notable decrease in the protein levels of tight junction proteins like ZO-1, occludin, and claudin-5, which can be reversed by inhibiting the NLRP3 inflammasome. These findings suggest that the Slit2-Robo4 signaling pathway and tight junction proteins in the intestine play crucial roles in LPS-induced inflammation in mice, providing insights into the molecular mechanisms underlying sepsis.
What biological factors/mechanisms may underlie the development and maintanance of PTSD?
5 answers
The development and maintenance of PTSD may involve various biological factors and mechanisms. Neurobiological models suggest that PTSD symptoms can be influenced by neural mechanisms involving fear-related brain structures like the hippocampus, amygdala, and prefrontal cortex, which can undergo time-dependent changes. Epigenetic changes at the genomic and epigenomic level have been proposed to play a role in the susceptibility to PTSD after exposure to traumatic stress. Inflammation, particularly neuroinflammation, has been identified as a putative susceptibility factor for PTSD, with elevated levels of certain cytokines in the brain of susceptible individuals. Furthermore, biomarkers associated with neurodegenerative disorders, such as cerebral cortical thinning and immune-inflammatory alterations, have been linked to PTSD, potentially shedding light on the pathophysiological mechanisms connecting these conditions. Additionally, aberrant susceptibility of emotion- and fear-related neurocircuits, including the amygdala, prefrontal cortex, and hippocampus, may contribute to the development and retention of PTSD symptoms.
How does alcohol consumption impact the immune system, leading to neutrophil dysfunction in Alcoholic Hepatitis patients?
10 answers
Alcohol consumption significantly impacts the immune system, leading to various dysfunctions, including neutrophil dysfunction, particularly in patients with Alcoholic Hepatitis (AH). Chronic excessive alcohol consumption causes Alcoholic Liver Disease (ALD), which encompasses a spectrum of liver injuries, including AH, characterized by an inflammatory response where immune cells play a crucial role. Patients with AH exhibit immune dysfunction, with studies showing altered mitochondrial function in immune cells, which is critical for their responses and regulates senescence, further contributing to immune dysfunction. The metabolism of alcohol produces toxic metabolites like acetaldehyde and reactive oxygen species, which, along with alcohol-induced dysbiosis and increased intestinal permeability, promote the translocation of bacterial products. This translocation exacerbates the production of inflammatory cytokines in the liver, perpetuating local inflammation and contributing to the progression of ALD. Alcohol alters both innate and adaptive immune cells, including neutrophils, macrophages, B cells, and T cells, leading to immune dysfunction associated with alcohol-induced end-organ damage. Chronic alcohol consumption affects circulating and tissue-resident myeloid cells, dampening acute drinking but enhancing chronic drinking's production of inflammatory mediators. In AH, the immune system is hyperactive yet dysfunctional, with alterations in immune checkpoints (ICPs) that could relate to the dysregulated state, affecting the functionality of immune cells including neutrophils. Specifically, patients with severe AH have dysfunctional NK cells and CD8 T cells, which fail to kill target cells, including monocytes, due to downregulated receptors necessary for cytotoxic cell recognition, indicating a broader immune dysfunction that likely includes neutrophil impairment. Alcohol dysregulates the function of lymphocytes, neutrophils, monocytes, and tissue macrophages, contributing to systemic inflammation, oxidative stress, mitochondrial dysfunction, and fibrosis in ALD and AH. Furthermore, the neutrophil-to-lymphocyte ratio (NLR) has been associated with infection, acute kidney injury (AKI), and the severity of alcoholic hepatitis, indicating that neutrophil dysfunction plays a significant role in the disease's progression and outcome. Lastly, acute alcoholic hepatitis is characterized by profound immune dysfunction, with evidence of impaired microbial defense, where neutrophils exhibit reduced phagocytic capacity and oxidative burst. This comprehensive view from multiple studies underscores the multifaceted impact of alcohol consumption on the immune system, leading to neutrophil dysfunction among other immune impairments in AH patients.
What are the immunomodulatory testing methods?
5 answers
Immunomodulatory testing methods encompass a variety of approaches for evaluating immune responses and immunomodulatory agents. These methods include in-vitro assays using cell lines and murine models to quantify immune responses, assess immune functions, screen anti-allergic drugs, and study autoimmune disorders. Additionally, methods involve identifying immunomodulatory genes through genetic screening techniques. Immunological testing devices have been developed to analyze chromatography test samples by detecting inclination and analyzing density information of the sample fluid. Furthermore, an immunological testing method simplifies specimen testing by integrating labeled antibodies, filtering specimen liquids, and observing antigen presence for accurate diagnosis, particularly useful for infectious diseases like influenza. Another method involves testing immunomodulatory drugs by observing T-cell activation in peripheral blood mononuclear cell cultures after drug exposure.
What are the cytokines involved with LAG-3 receptor from T helper cell?
4 answers
The cytokines involved with the LAG-3 receptor from T helper cells include TNF-α, which is increased in the presence of LAG-3 deficiency, leading to an enhanced proinflammatory state. Additionally, LAG-3 negatively regulates TCR signal transduction and affects cytokine secretion, ultimately impacting body immunity and immune escape of pathogens. Furthermore, the inhibition of LAG-3 in combination with PD-1 has shown to enhance cytokine production in response to cancer and infections, indicating a potential role in modulating cytokine responses in various conditions. These findings highlight the intricate role of LAG-3 in cytokine regulation and its potential implications in immune responses and therapeutic interventions.
What are the underlying mechanisms that lead to the formation of keloids and hypertrophic scars in patients?
4 answers
Keloids and hypertrophic scars result from abnormal wound healing processes characterized by excessive collagen deposition and fibroblast activity. The pathophysiology involves genetic, epigenetic, and environmental factors contributing to the overproduction of collagen fibers by fibroblasts in response to proinflammatory signals. Studies suggest that keloid fibroblasts may be epigenetically locked in an activated state, perpetuating the keloid phenotype. Additionally, immune cell infiltration and dysregulation of immune pathways play crucial roles in keloid formation. Furthermore, the Hippo pathway effectors YAP/TAZ and integrin-linked kinase (ILK) have been implicated in abnormal fibroblast activation and proliferation in keloids and hypertrophic scars, shedding light on potential therapeutic targets. These insights enhance our understanding of the mechanisms underlying keloid and hypertrophic scar formation, paving the way for targeted treatment approaches.
What are the features of infiltrative HCC?
5 answers
Infiltrative hepatocellular carcinoma (HCC) is characterized by small tumor nodules spreading diffusely across the liver, lacking a distinct mass-like lesion. It presents as a markedly heterogeneous area on ultrasonography and a poorly defined hepatic region on MRI, often blending into the background of cirrhosis. Infiltrative HCC is associated with poor prognosis due to late discovery and advanced stage at diagnosis. This subtype is linked to multifocal tumors, vessel invasion, and larger tumor sizes, impacting overall survival significantly, especially in tumors ≥ 4 cm. Understanding the imaging features of infiltrative HCC is crucial for timely diagnosis and effective management. Additionally, the tumor microenvironment plays a crucial role in the progression and malignancy of infiltrative basal cell carcinoma, a subtype of BCC, emphasizing the importance of cellular heterogeneity in understanding and treating these aggressive cancers.
What is the function of immunomodulatory testing?
5 answers
Immunomodulatory testing serves the crucial function of assessing immune responses in various conditions, aiding in the diagnosis of opportunistic infections, evaluating vaccine efficacy, managing immunosuppressive therapies, and guiding immune-targeted therapies. These tests are essential for understanding the dynamic nature of the immune system, especially in cases of unbalanced responses leading to diseases like autoimmune disorders, allergies, and opportunistic infections. Immunomodulatory testing involves analyzing immune functionality through parameters like phagocytosis efficiency, pathogen killing, reactive species production, and cytokine levels, often using primary cells and cell lines. By providing insights into immune profiles, these tests contribute to personalized and precision medicine, aiming to improve patient outcomes and therapeutic strategies.