scispace - formally typeset
Search or ask a question

What are the functions of cytokines produced by cells? 


Best insight from top research papers

Cytokines are biologically active proteins that regulate various processes in the body, including immune response, cell growth, differentiation, and functional activity . They are produced by all types of cells and play a crucial role in immune regulation, inflammatory responses, and homeostasis . Cytokines can elicit a range of responses such as cell activation, proliferation, differentiation, angiogenesis, hematopoiesis, and inflammation . They act as messengers between cells, modifying cellular functions and allowing communication between different cell types . Cytokines also have an impact on neuronal structure and function, influencing synaptic scaling, plasticity, and synapse structure . Overall, cytokines have diverse functions in the body, playing a vital role in immune regulation, inflammation, cellular communication, and neuronal processes.

Answers from top 3 papers

More filters
Papers (3)Insight
The paper does not explicitly mention the functions of cytokines produced by cells.
The functions of cytokines produced by cells include regulating immune response, cell growth, differentiation, and functional activity. They can both stimulate and suppress these processes.
Book ChapterDOI
R.A. Prell, J.M. Tarrant 
01 Jan 2018
Cytokines have various functions including immune regulation, inflammatory responses, cell activation, proliferation, differentiation, angiogenesis, hematopoiesis, and inflammation.

Related Questions

What are cytokines/?5 answersCytokines are small proteins or peptides that act as immunomodulating agents, released mainly by cells of the immune system in response to various stimuli. They play a crucial role in regulating immune responses, inducing inflammation, and modulating immune system activity. Cytokines can have pleiotropic effects, influencing cell differentiation, proliferation, and activation in nearby or distant cells. Classified based on structure and function, cytokines include interleukins, chemokines, interferons, and growth factors. While essential for host defense, certain cytokines like IL-1, IL-6, and TNF-α can also contribute to inflammatory and chronic diseases. Moreover, cytokine storms, characterized by excessive cytokine release, can lead to severe conditions like sepsis and acute respiratory distress syndrome. The skin serves as a model system for studying cytokine biology, with implications for dermatology and therapeutic interventions.
What role do cytokines play in the immune system?5 answersCytokines play a crucial role in the immune system by regulating inflammation, immunity, and immune cell growth and maturation. They act as intercellular messengers, mediating the complex interactions between different immune cells. Cytokines are produced by almost every cell in the body except red blood cells and have evolved to include various types such as interferons, interleukins, chemokines, and adipokines. They have pleiotropic effects on immune cells and are involved in immune cell differentiation, inflammation, tumorigenesis, and viral pathogenesis. Cytokines also have a significant role in autoimmune diseases and can serve as diagnostic, prognostic, and therapeutic biomarkers. In the context of cancer, cytokines are key components of the immune system and are being studied for their potential as therapeutic agents or targets in cancer immunotherapy.
Explain how cytokine signalling lead to interleukin production?4 answersCytokine signaling leads to interleukin production through a series of events. When a cytokine binds to its receptor on the cell surface, it triggers dimerization or conformational change of the receptor, which activates Janus kinase proteins (JAKs). The activated JAKs phosphorylate tyrosines on the receptor and themselves, initiating intracellular signaling pathways. These pathways often involve a cascade of kinases, leading to the activation of substrate proteins within the cell. In the case of IL-6, it can induce IL-6 production in monocytes when stimulated by IL-1. Additionally, IL-6 can increase STAT3 phosphorylation, and inhibition of JAKs, MEK, and PI3K interferes with IL-10 production. Overall, cytokine signaling activates intracellular pathways that ultimately result in the production of interleukins.
What are the mechanisms of action of cytokines produced by cells?3 answersCytokines are low-molecular-weight molecules that play a vital role in intercellular communication and regulate various physiological and pathological events. They are secreted by immune and glial cells and can induce and maintain pain. Cytokines act through specific receptors, such as cytokine receptors, ionotropic glutamate receptors, transient vanilloid receptors, and sodium channels, to modulate pain sensation. Imbalances in pro- and anti-inflammatory cytokine expression may contribute to individual pain susceptibility. Cytokines also have effects in the central nervous system, including fever, sickness behavior, and activation of the hypothalamic-pituitary-adrenal axis. Cytokines can be packaged into extracellular vesicles (EVs) and their secretion may be regulated by cytokines. The packaging of cytokines into EVs is influenced by the biological state of the cell and the EV biogenesis pathway involved. Understanding the mechanisms of cytokine release and packaging into EVs may have therapeutic implications for regulating innate and adaptive immunity.
Cytokines and chemokines?4 answersCytokines and chemokines are small biologically active molecules that play crucial roles in immune responses, cell-to-cell communication, and immune cell trafficking. Cytokines are soluble mediators that act as messengers of the immune system and are involved in processes such as host defense, inflammation, wound healing, and angiogenesis. They can have both proinflammatory and anti-inflammatory potential, depending on the immune cells present and their state of responsiveness. Chemokines, on the other hand, attract specific populations of inflammatory cells and regulate the trafficking of leukocytes during immune responses. They are divided into different families based on their structure and have diverse functions, including regulating immune cell migration and contributing to autoimmunity. The cytokine and chemokine milieu in the wound microenvironment plays a crucial role in the healing process, and dysregulation of these molecules is involved in the pathogenesis of chronic diabetic wounds.
14)What are therapeutic use of cytokine?3 answersCytokines have therapeutic uses in various fields such as cancer treatment, infectious diseases, autoimmunity, and heart disease. They play a crucial role in regulating the immune system and can provoke or mitigate immune responses. In cancer therapy, cytokines are used as therapeutic agents or targets to enhance the anticancer immune response. Efforts have been made to improve the delivery efficiency, stability, and availability of cytokines in therapeutic applications. In the treatment of melanoma, innovative technological approaches have been developed to improve the safety and efficacy of cytokines, resulting in encouraging clinical activity. Cytokines have also been explored as drugs in immunotherapy for bacterial and viral infections, autoimmune diseases, transplantation, and veterinary medicine. In the field of heart disease, cytokine-based therapies have shown promise in reducing post-infarct heart failure and chronic ischemia by stimulating cell proliferation and mobilization.

See what other people are reading

How do M cells contribute to the development of autoimmune diseases?
5 answers
M cells, specialized epithelial cells in mucosal lymphoid tissues, play a crucial role in autoimmune disease development. They transport luminal antigens to initiate immune responses, potentially contributing to systemic autoimmunity beyond mucosal responses. Dysbiosis of intestinal microbiota, taken up by M cells, may be implicated in autoimmune disease pathogenesis. In autoimmune diseases, monocytes and macrophages, key innate immune cells, are involved in disease initiation and progression, with their infiltration being a hallmark of autoimmune conditions. Additionally, NK cells, part of the innate immune system, have been linked to various autoimmune diseases, showcasing both protective and pathogenic roles depending on subsets and disease context. Understanding the roles of M cells, monocytes, macrophages, and NK cells in autoimmune diseases could lead to more effective therapeutic strategies in the future.
What are the links between allergies and neuroinflammation?
5 answers
Allergies have been associated with neuroinflammation through the involvement of mast cells (MCs). MCs, known for their role in allergic reactions, can contribute to neuroinflammation by releasing inflammatory mediators, affecting the blood-brain barrier (BBB) integrity, and interacting with glial cells and neurons. Studies suggest that MC activation may play a part in the pathogenesis of neurodevelopmental disorders like attention deficit hyperactivity disorder (ADHD). Furthermore, MCs have been implicated in exacerbating CNS damage in conditions such as brain ischemia and hemorrhage, while also potentially playing a protective role in traumatic brain injury. The intricate relationship between allergies, MC activation, and neuroinflammation underscores the complex interplay between the immune system and the central nervous system in various pathological processes.
What is the healthiest bean for a cat to eat?
5 answers
The healthiest bean for a cat to eat would be red kidney beans (RKBs) due to their beneficial effects on lipid and glucose metabolism, as well as their potential to prevent hypercholesterolemia, improve glucose tolerance, and enhance gut health. Research suggests that processed RKBs, particularly roasted RKBs, can positively impact the cat's health by increasing fecal IgA and mucin content, suppressing liver injury markers, and potentially enhancing bile acid biosynthesis. Additionally, kidney beans, both white and dark red varieties, have shown anti-inflammatory effects and the ability to enhance microbial activity, gut barrier integrity, and antimicrobial defense responses in healthy mice, which can translate to a beneficial anti-inflammatory effect during colitis. Therefore, incorporating red kidney beans into a cat's diet may offer various health benefits.
Is vaccination an effective control against COVID?
4 answers
Vaccination has shown to be an effective control measure against COVID-19. Studies have demonstrated that vaccines can induce robust antibody and T cell responses, providing protection against the virus. Furthermore, research on recovered individuals and vaccinated cohorts indicates that immunity against SARS-CoV-2 can be long-lasting, with antibodies evolving to be more potent and broad over time. While some vaccines have faced controversies and questions over side effects, the overall consensus is that vaccination significantly reduces the risk of developing COVID-19. However, the protective effect of vaccines against long COVID remains debated in various studies. In conclusion, vaccination plays a crucial role in controlling the spread of COVID-19 by eliciting strong immune responses and reducing the severity of the disease.
What causes covid 19?
4 answers
COVID-19 is primarily caused by the severe acute respiratory syndrome-associated coronavirus-2 (SARS-CoV-2) virus, leading to a systemic illness with various manifestations. The disease progresses through different stages of infection, starting with mild symptoms targeting nasal epithelial cells and advancing to severe stages involving cytokine storm syndrome and acute respiratory distress syndrome (ARDS). The pathophysiology involves oxidative stress, endothelial dysfunction, and lipid peroxidation in endothelial cells, particularly in patients with severe outcomes. The exaggerated inflammatory response in COVID-19 results in tissue damage, proinflammatory cytokine release, and vascular hyper-permeability, leading to multiorgan failure. Factors like obesity, hypertension, diabetes, and aging contribute to a poor prognosis, possibly due to persistent inflammation. Understanding these mechanisms is crucial for developing effective treatments targeting specific components of the immune system to combat COVID-19.
What are the underlying mechanisms linking diabetes and atherosclerosis?
5 answers
Diabetes and atherosclerosis share intricate connections through various biological mechanisms. Endothelial dysfunction, inflammation, and oxidative stress are key features in both conditions, contributing to their pathogenesis. Factors like hyperglycemia, endothelial activation, inflammation, mitochondrial oxidative stress, and alterations in microRNAs play crucial roles in the progression of atherosclerosis in diabetic individuals. The nuclear factor κB is highlighted as a central regulator influencing these shared mechanisms, suggesting a common pathophysiological basis for diabetes and atherosclerosis. Additionally, extracellular vesicles, microRNAs, and metabolic reprogramming of macrophages have been identified as significant players in the development and progression of atherosclerosis in the context of diabetes. Understanding these underlying mechanisms is vital for identifying potential therapeutic targets to manage diabetic atherosclerosis effectively.
How does IL-34 contribute to angiogenesis of prostate cancer?
4 answers
IL-34 plays a significant role in promoting angiogenesis in prostate cancer (PCA) by influencing the tumor microenvironment. Endothelial cells, which express CD31 and CD34, are increased in PCA and contribute to metastasis by enhancing cancer cell invasion. IL-34, secreted by various cells including tumor cells, has been implicated in promoting cancer growth and metastasis in various cancers. Additionally, IL-34 is involved in regulating immune cells, particularly macrophages, in the tumor microenvironment, which can lead to immune suppression and cancer progression. The combination of IL-34 inhibition with immune checkpoint inhibitors (ICIs) has been proposed as a potential strategy to overcome resistance to ICIs and improve cancer treatment efficacy. Therefore, targeting IL-34 may offer a promising approach to hinder angiogenesis and enhance therapeutic outcomes in prostate cancer.
What are the most common biomarkers used for the diagnosis and monitoring of colorectal cancer?
4 answers
The diagnosis and monitoring of colorectal cancer (CRC) have significantly advanced with the identification and application of various biomarkers, which are crucial for early detection, prognosis, and monitoring therapeutic responses. Among the most common biomarkers, genetic and epigenetic alterations, including mutations in APC, TP53, KRAS, BRAF, and PIK3CA genes, are widely recognized for their roles in CRC pathogenesis and as diagnostic tools. Additionally, DNA methylation patterns, such as those in SEPT9, HLTF, NEUROG1, MLH1, ALX4, and LINE-1, serve as epigenomic biomarkers, offering insights into CRC's epigenetic landscape. Transcriptomic markers, particularly microRNAs (miRNAs) like miR-21, miR-92a, miR-103a-3p, and miR-127-3p, have emerged as valuable tools for CRC diagnosis due to their altered expression levels in CRC tissues compared to normal tissues. Furthermore, the role of non-coding RNAs, including long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and PIWI-interacting RNAs (piRNAs), is being explored for their diagnostic and prognostic potential in CRC. Proteomic markers, such as CEA, NDKA, calprotectin (S100A8/A9), CLU, TIMP1, and ECM1, have shown promise in CRC detection and monitoring, benefiting from advancements in mass spectrometry and other proteomic technologies. Liquid biopsy, analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and other components like extracellular vesicles, offers a non-invasive approach for CRC diagnosis, prognosis, and treatment monitoring, reflecting tumor heterogeneity and dynamics. Fecal biomarkers, including DNA, RNA, protein biomarkers, gut microbes, and volatile organic compounds, have also been identified for CRC screening and early diagnosis, providing a non-invasive, cost-effective method suitable for large-scale population screening. In summary, the landscape of CRC biomarkers encompasses a wide array of molecular signatures from genetic, epigenetic, transcriptomic, proteomic, and fecal biomarkers to liquid biopsy components, each contributing to the comprehensive understanding and management of CRC.
How liver could play a substantial role in cancer related cachexia development?
5 answers
The liver plays a significant role in cancer-related cachexia development by contributing to systemic inflammation and metabolic disruption. In cachexia, the liver shows alterations in pathways related to mitochondrial function, glycogen catabolism, and lactate metabolism, potentially leading to inefficient energy utilization. Additionally, hepatic dysregulation exacerbates the pro-inflammatory response initiated by tumors, further promoting muscle and adipose tissue wasting. Studies have shown that cancer cachexia is associated with an increase in myeloid cell infiltration in the liver, along with modulation of the inflammasome pathway, leading to elevated release of interleukin-1β and systemic inflammation. These findings highlight the liver's crucial involvement in the pathogenesis of cancer-related cachexia through metabolic and inflammatory mechanisms.
How does WBC increase T2D?
5 answers
A raised white blood cell (WBC) count is associated with a higher risk of developing type 2 diabetes (T2D). Studies have shown that individuals with T2D exhibit disease-associated changes in B-cell function, leading to a proinflammatory cytokine profile that promotes insulin resistance and systemic inflammation. Moreover, beta cells, which regulate insulin secretion, face increased metabolic demands in conditions like obesity and insulin resistance, potentially leading to T2D development. The correlation between WBC count and T2D risk is independent of other factors like weight, smoking, and lipid levels, particularly in middle-aged and elderly Chinese individuals. Overall, these findings suggest that inflammation, influenced by factors like WBC count and B-cell function, plays a crucial role in the pathogenesis of T2D.
Can microglia get infected with influenza a virus h7n7?
5 answers
Microglia, the brain's resident immune cells, can indeed get infected with Influenza A virus (IAV) subtype H7N7. Research has shown that H7N7 infection can lead to microglia-induced neuroinflammation, affecting the central nervous system (CNS). Additionally, studies have demonstrated that both human H1N1 and avian H5N1 influenza viruses can infect microglia, inducing apoptosis, cytopathy, and proinflammatory cytokine production in these cells. Furthermore, neuroinflammation induced by neurotropic H7N7 infection results in long-term impairments in the CNS, affecting hippocampal neuronal morphology and cognitive function. These findings highlight the potential of influenza viruses, including H7N7, to infect microglia and trigger detrimental neuroinflammatory responses in the brain.