scispace - formally typeset
Search or ask a question

What is the role of IL-1b in regulating metabolism and energy balance? 


Best insight from top research papers

IL-1β plays a crucial role in regulating metabolism and energy balance by influencing various physiological processes. It has been found that IL-1β contributes to postprandial insulin secretion, adjusts glucoregulation in the brain, and enhances glucose uptake into macrophages. Additionally, IL-1β can stimulate brain metabolism, affect butyrate oxidation in colorectal cancer cells, and is required for maintaining steady-state glucose homeostasis. These findings highlight the multifaceted impact of IL-1β on glucose metabolism, insulin secretion, and energy provision to neural cells, underscoring its significance in maintaining metabolic homeostasis and energy balance.

Answers from top 5 papers

More filters
Papers (5)Insight
Journal ArticleDOI
Marit Hjorth, Mark A. Febbraio 
15 Feb 2017-Nature Immunology
5 Citations
IL-1β plays a dual role in metabolism; it is associated with inflammation and obesity but also aids in maintaining glucose homeostasis by enhancing postprandial insulin secretion.
IL-1β diminishes butyrate oxidation in colorectal cancer cells, suppressing NADH levels and mitochondrial respiration. It shifts cancerous colonocytes away from utilizing and oxidizing butyrate, impacting energy metabolism.
IL-1β, secreted by macrophages postprandially, stimulates insulin secretion, promoting glucose uptake. IL-1β and insulin synergistically regulate glucose disposal and inflammation, playing a crucial role in metabolism and immunity.
IL-1β adjusts glucoregulation, stimulates brain metabolism, and provides fuel to neural cells in an autocrine/paracrine manner, supporting synaptic plasticity, learning, and memory.
IL-1β plays a crucial role in glucose homeostasis by promoting postprandial insulin secretion, regulating glucose disposal, and alerting the immune system to prevent microbial dissemination from food.

Related Questions

What role does IL1B play in senescence?4 answersIL-1β plays a significant role in inducing senescence in various cell types. Studies have shown that IL-1β can induce cellular senescence in vascular smooth muscle cells (VSMCs), rat astrocytes, and bovine oviduct epithelial cells (OECs). This induction of senescence is associated with increased expression of senescence markers, such as p21, and activation of senescence-associated pathways, including NF-κB/p53/p21. IL-1β-induced senescence can lead to cellular dysfunction and the development of a senescence-associated secretory phenotype (SASP), characterized by the secretion of inflammatory cytokines. Furthermore, IL-1β has been implicated in promoting inflammatory responses and immune cell migration, contributing to chronic inflammation and tissue dysfunction. Overall, IL-1β emerges as a key mediator of senescence and inflammation in various cell types.
Whats the influence of IL-1 on growth hormone?5 answersInterleukin-1 (IL-1) plays a significant role in modulating the effects of growth hormone (GH) on various physiological processes. Studies have shown that IL-1 can induce GH resistance during catabolic illness or inflammation, leading to decreased circulating insulin-like growth factor-I (IGF-I) levels despite elevated GH levels. IL-1 inhibits the expression of GH-inducible genes like serine protease inhibitor (Spi 2.1) and IGF-I through the NFκB pathway, contributing to GH resistance. Additionally, IL-1 has been found to dose-dependently inhibit the growth of normal rat pituitary cells, indicating a direct influence on pituitary cell growth. Central administration of IL-1β has also been shown to alter peripheral IGF-I levels, further highlighting the intricate relationship between IL-1 and the GH/IGF-I axis.
What is the role of DKK1 on IL-1b secretion?5 answersDKK1 plays a crucial role in IL-1β secretion by modulating inflammatory responses. Research indicates that DKK1 maintains an elevated inflammatory tone in cancer cells and is essential for cytokine responses triggered by toll-like and cytokine receptors. In conditions like neonatal-onset multisystem inflammatory disease (NOMID), IL-1β reduces DKK1 production, promoting Wnt signaling activation and potentially contributing to dysregulated bone formation. Moreover, in Epstein-Barr virus-infected cells, DKK1 influences NF-kappaB activation, with IRAK1 playing a critical role in mediating p65/RelA S536 phosphorylation, a key step in NF-kappaB activation. In chronic obstructive pulmonary disease (COPD), decreased DKK1 levels are associated with airway inflammation, and overexpression of DKK1 attenuates inflammatory factors, suggesting a role in COPD pathophysiology.
What is the function of IL-1β in rats?5 answersIL-1β has various functions in rats. It plays a key role in the occurrence and development of vascular dementia (VD) by regulating cell apoptosis and improving learning ability. IL-1β is also involved in ovulation and female reproduction, as its plasma concentrations are higher in rats treated with the ovulation-inducing agent clomiphene citrate. In the context of global ischemia, inhibiting IL-1β with an antibody can have protective effects on cognitive function recovery, even when administered 24 hours after the ischemic event. Additionally, IL-1β is associated with neuropathic pain after nerve injury, and its concentration in the intrathecal space correlates with the extent of mechanical allodynia, providing a molecular biomarker for pain. Finally, during bladder outlet obstruction, NLRP3-mediated inflammation triggered by IL-1β leads to bladder denervation, indicating the involvement of IL-1β in this process.
What is the mechanism of IL-1beta secretion?4 answersIL-1β secretion involves two main mechanisms: pyroptosis and unconventional protein secretion. Pyroptosis is a form of cell death that leads to the release of IL-1β and other inflammatory cytokines. It is triggered by the activation of inflammasomes, which are supramolecular platforms that respond to damage-associated molecular patterns and pathogen-associated molecular patterns. The other mechanism, unconventional protein secretion, is a process by which leaderless proteins, such as IL-1β, are secreted without going through the ER/Golgi pathway. The exact mechanism of unconventional protein secretion is still not well understood, but it has been proposed that caspase-1, the protease responsible for the maturation of IL-1β, plays a role in this process. Calcium and calmodulin have also been identified as important factors in IL-1β secretion, as their presence is required for nigericin-induced IL-1β release. Further research is needed to fully elucidate the mechanisms of IL-1β secretion and to explore potential therapeutic opportunities for targeting this process in various diseases.
Does interleukin-1 cause metabolic syndrome?5 answersInterleukin-1 (IL-1) is implicated in the pathophysiology of metabolic syndrome. Elevated IL-1β expression is associated with chronic low-grade inflammation in patients with metabolic syndrome. Polymorphisms in the IL-1 gene family are associated with central obesity and metabolic syndrome. IL-1β levels are significantly higher in patients with metabolic syndrome compared to healthy controls. Visceral obesity, a component of metabolic syndrome, is associated with low-grade chronic inflammation and oxidative stress, which are synergized by IL-1β. IL-1β plays a role in diseases associated with metabolic syndrome, such as Type 2 diabetes, atherosclerosis, and chronic heart failure. Therefore, IL-1β is involved in the development and progression of metabolic syndrome, contributing to inflammation, insulin resistance, and other metabolic abnormalities.

See what other people are reading

What role has the hypothalamus?
5 answers
The hypothalamus plays a crucial role in various physiological functions, including metabolic regulation, glucose homeostasis, endocrine control, and integration of signals for maintaining overall homeostasis. It acts as a central regulatory center in the brain, influencing energy balance, food intake, body temperature, fluid balance, and blood pressure. Dysfunction in the hypothalamus can lead to a range of disorders, such as obesity in cases of hypothalamic dysfunction or surgery. Additionally, alterations in the hypothalamic nuclei have been linked to conditions like essential hypertension, highlighting the role of the hypothalamus in blood pressure regulation. Understanding the mechanisms and pathways controlled by the hypothalamus is crucial for addressing metabolic diseases, diabetes, and other related health issues.
How does high-fiber diet associate with body composition?
5 answers
A high-fiber diet is closely linked to favorable body composition outcomes. Research indicates that higher dietary fiber intake is associated with lower body mass index (BMI), reduced body weight, decreased waist circumference, and lower body fat percentage. Additionally, consuming diets rich in fiber can lead to increased lean mass, improved glucose homeostasis, and enhanced skeletal muscle strength. Notably, individuals with high physical activity levels who also have high fiber intake exhibit significantly lower values for body composition parameters, particularly in males. These findings suggest that incorporating a high-fiber diet into one's lifestyle, especially when combined with physical activity, can contribute to maintaining a healthy body composition by reducing fat mass and promoting lean muscle mass.
What are the nutritional benefits of Parmesan cheese compared to other types of cheese?
5 answers
Parmesan cheese, like other hard Italian cheeses such as Parmigiano Reggiano, offers various nutritional benefits. These cheeses are rich in protein, vitamins (such as A, B12, and D), and minerals like calcium, phosphorus, iron, zinc, and selenium. They also contain bioactive components like conjugated linoleic acid, phytanic acid, and peptides with antihypertensive, antioxidant, and anti-inflammatory effects. Parmesan cheese is low in lactose, making it suitable for lactose-intolerant individuals. Additionally, Parmigiano Reggiano cheese has been found to have high protein content with a favorable biological value, making it a nutritious and functional food suitable for various age groups and potentially beneficial for conditions like diabetes, osteoporosis, and dyslipidemias.
How much sodium butyrate use to incraese expression in Bacmam?
5 answers
Sodium butyrate has been shown to increase gene expression in various studies. In the context of BacMam-mediated gene expression, the optimal concentration of sodium butyrate varies depending on the specific gene and cell type. Research suggests that sodium butyrate can enhance gene expression in different systems. For instance, in the context of the human NIS gene, sodium butyrate treatment led to increased expression of the gene in A549 cells, resulting in higher iodine accumulation. Similarly, sodium butyrate was found to stimulate HBsAg expression in human hepatoma cells in a dose-dependent manner. Moreover, sodium butyrate induced the expression of the CEA gene in colorectal cancer cells through both transcriptional and post-transcriptional mechanisms. Therefore, the appropriate concentration of sodium butyrate to enhance gene expression in BacMam systems may need to be determined based on the specific gene and cell line used.
Concentration of sodium butyrate in Bacmam?
5 answers
The concentration of sodium butyrate (SB) varies depending on the specific application. In the context of glioblastoma (GB) cell studies, physiological concentrations of SB ranging from 0.25 mM to 4 mM were used to demonstrate inhibitory effects on cell proliferation, invasion, and induction of senescence. In a study involving B16 melanoma cells, concentrations of 1 mM, 2 mM, 3 mM, and 5 mM of SB were utilized to observe significant reductions in cell viability and tumor suppression effects. The effectiveness of SB in bovine macrophages to alleviate inflammatory responses was demonstrated using concentrations up to 1 mM. Therefore, the concentration of sodium butyrate in Bacmam would depend on the specific experimental requirements and the desired outcomes, with concentrations ranging from 0.25 mM to 5 mM based on the different studies cited.
What is the typical range of hormone levels in rats?
5 answers
The typical range of hormone levels in rats varies depending on the specific hormone and age group. For instance, in male rats, testosterone levels start low and increase to adult levels by day 62, then decline gradually with age. In female rats, serum LH concentrations can vary significantly, with peaks observed before ovulation. Growth hormone levels in female rats show considerable variation with age, with peaks and declines observed at different stages of development. Additionally, diurnal variations in hormone levels are noted, with fluctuations in FSH, LH, oestradiol, prolactin, and testosterone levels in both male and female rats at different stages of prepubertal development. Overall, hormone levels in rats exhibit dynamic changes throughout development and are influenced by factors such as age, sex, and circadian rhythms.
Why excessive energy intake is important for managing type 2 diabetes?
5 answers
Excessive energy intake plays a crucial role in managing type 2 diabetes due to its association with insulin resistance and the risk of developing the disease. Studies have shown that high energy intake is strongly linked to increased insulin resistance, which is a key factor in the development of type 2 diabetes, especially in childhood. Furthermore, the prevalence of type 2 diabetes is closely related to lifestyle factors such as excessive energy intake, lack of physical exercise, and obesity. Effective management of diabetes involves controlling energy intake, as reducing caloric intake can lead to weight loss, improved insulin sensitivity, and better blood glucose control in type 2 diabetes patients. Therefore, monitoring and regulating energy intake are essential components of managing type 2 diabetes and reducing associated risks.
Is this sentence true: SC-islets respond poorly to glucose, and more importantly enter a hyperinsulinemic state when stimulated pyruvate?
5 answers
SC-islets derived from patients with diazoxide-unresponsive diffuse KATPHI exhibit altered responses to glucose and pyruvate. These SC-islets, carrying mutations in the ABCC8 gene, show increased insulin secretion in low glucose conditions and fail to respond to KATP-channel-acting drugs, indicating poor glucose responsiveness. On the other hand, when stimulated with pyruvate, SC-islets from patients with KATPHI mutations display hyperinsulinemia, suggesting a dysregulated response to pyruvate. This hyperinsulinemic state in SC-islets is likely influenced by the very low threshold for glucose-induced insulin secretion observed in islets from genetically obese mice, which can lead to exaggerated insulin secretion responses. Therefore, the statement that SC-islets respond poorly to glucose and enter a hyperinsulinemic state when stimulated by pyruvate is supported by the data from the research contexts.
How do prebiotics, like FOS, affect the gut microbiome of infants?
5 answers
Prebiotics, such as fructooligosaccharides (FOS), play a crucial role in shaping the gut microbiome of infants. Studies have shown that FOS supplementation can lead to bifidogenic effects in both children and adults, promoting the growth of beneficial Bifidobacterium species. Additionally, prebiotic oligosaccharides have been proposed as a strategy to reduce the risk of necrotising enterocolitis (NEC) and associated mortality in very preterm or very low birth weight (VLBW) infants, although the evidence on their effectiveness remains uncertain. Furthermore, the addition of 2'-fucosyllactose (2'-FL) and 3-fucosyllactose (3-FL) has been shown to increase the relative abundance of Bifidobacterium species in infant gut microbiota, indicating a beneficial impact on microbial composition and metabolites.
Does age impact the patient awareness of pre analytical requirements?
5 answers
Age does impact patient awareness of pre-analytical requirements. Studies have shown that inadequate information provided by clinicians when requesting histopathology investigations can lead to errors in specimen collection. Furthermore, research indicates that incorrect test results are often due to patient and specimen collection considerations rather than laboratory variability. Proper patient preparation, clear communication of requirements, and correct sample handling are crucial in ensuring accurate test results. Therefore, educating patients, especially older individuals, about the importance of following pre-analytical guidelines can significantly improve the quality of diagnostic testing and patient care. Clinicians must also be diligent in providing complete information and using appropriate specimen containers to minimize errors in the pre-analytical phase.
What is the function of G-Protein-coupled receptors in insuline resistance?
4 answers
G-Protein-coupled receptors (GPCRs) play a crucial role in insulin resistance, a key factor in metabolic disorders like type 2 diabetes mellitus (T2DM) and metabolic syndrome. These receptors are directly involved in the development of insulin resistance, β-cell dysfunction, and inflammation leading to T2DM. Specific GPCR signaling pathways, such as Gq signaling in skeletal muscle (SKM), have been shown to enhance glucose uptake and improve glucose homeostasis, suggesting that targeting GPCRs could be beneficial in treating insulin resistance and diabetes. Additionally, GPR120, a GPCR for free fatty acids, influences proteins related to glucose metabolism, indicating its involvement in insulin resistance. Overall, understanding the role of GPCRs in insulin resistance is crucial for developing novel pharmacological interventions for metabolic disorders.