scispace - formally typeset
Search or ask a question

Answers from top 4 papers

More filters
Papers (4)Insight
We conclude therefore that entry is not the limiting step and that macrophages represent clinically relevant reservoirs for ‘non-macrophage tropic’ viruses.
Taken together, these results increase our understanding of how these viruses interact with human macrophages, at the cellular and molecular levels, and suggest mechanisms that may underlie their utility as recombinant vaccine vectors.
This suggests that virus might be shed from infected macrophages and then reinfect other macrophages.
IMPORTANCE Our studies clearly demonstrate that there are substantial biological differences in the patterns of cellular gene expression between macrophages infected with different poxvirus strains and that these changes are due specifically to infection with the distinct viruses.

Related Questions

What is the role of macrophages in vaccine response?5 answersMacrophages play a crucial role in the immune response to vaccines. Vaccination can enhance the phagocytosis and microbiocidal activity of macrophages, leading to a reduction in pathogen viability and an increase in the number of internalized pathogens. Activation of macrophages is observed after vaccination, as evidenced by the upregulation of genes associated with macrophage activation, such as interferon γ, interleukins, and macrophage numbers. Macrophages are also involved in antibody-mediated protection, as they play a dominant role in conferring protection provided by both broadly neutralizing and non-neutralizing antibodies. The interaction between protective antibodies and macrophages can induce inflammation and antibody-dependent cellular phagocytosis, contributing to the antiviral response. Overall, macrophages contribute to the immune response to vaccines by enhancing pathogen clearance, promoting inflammation, and mediating antibody-mediated protection.
How do tumor cells and macrophages interact?4 answersTumor cells and macrophages interact through various mechanisms in the tumor microenvironment. Tumor-associated macrophages (TAMs) are the most abundant stromal cells in the tumor microenvironment and can polarize into anti-tumorigenic (M1) or pro-tumorigenic (M2) phenotypes. Exosomes, which are small vesicles released by cells, play a crucial role in the communication between tumor cells and TAMs. Tumor-derived exosomal non-coding RNAs (ncRNAs) can promote macrophage polarization, while exosomal ncRNAs derived from TAMs can affect tumor proliferation, metastasis, angiogenesis, and chemotherapy resistance. Cancer stem cells (CSCs) can recruit macrophages into the tumor microenvironment and differentiate them into TAMs, which maintain CSC stemness and create niches favorable for CSC survival. Tumor cell-derived extracellular vesicles (T-EVs) can modulate the phenotypes and functions of macrophages, promoting tumor development. In glioblastoma, tumor cells release factors that recruit TAMs, which in turn create an immunosuppressive and tumor-supportive microenvironment.
How can tumor-associated macrophages be targeted with nanotechnology?5 answersTumor-associated macrophages (TAMs) can be targeted using nanotechnology. Nanoparticles have been developed for precise imaging and diagnosis of TAMs, allowing for tumor monitoring, surgical guidance, and efficacy evaluation. These nanoprobes and imaging agents include metal-based nanoprobes (iron, manganese, gold, silver), fluorine-19-based nanoprobes, radiolabeled agents, near-infrared fluorescence dyes, and ultrasonic nanobubbles. Additionally, nanocarriers have been designed to actively target TAMs and repolarize M2 TAMs into M1 TAMs, stimulating antitumor immune effects. Sonoresponsive nanoparticles have been used for antitumor sonodynamic therapy, inhibiting tumor growth and promoting the conversion of M2 TAMs into M1 TAMs in the tumor microenvironment. Furthermore, advanced nanostructures can deliver TAMs-modulating agents to enhance therapeutic efficacy and serve as modulators of TAMs. Macrophages and macrophage-derived components can also be exploited as tumor-targeting delivery vehicles. Overall, nanotechnology offers promising strategies for targeting TAMs in tumor immunotherapy.
Can macrophages become infected?4 answers
Do macrophages fight viruses?5 answers
How to target macrophages?1 answers

See what other people are reading

Does ATR inhibition activate the cGAS STING pathway?
5 answers
Yes, ATR inhibition activates the cGAS/STING pathway. Studies have shown that inhibiting ATR leads to the activation of the cGAS/STING pathway, resulting in increased type I interferon signaling. This activation triggers downstream effects such as induction of the interferon pathway genes, enhanced infiltration of cytotoxic T-cells, and upregulation of MHC class I expression in small cell lung cancer (SCLC) models. Additionally, the cGAS/STING pathway was found to be upregulated and activated in lung ischemia/reperfusion (I/R) injury models, suggesting its involvement in triggering endoplasmic reticulum stress (ERS) responses. Therefore, ATR inhibition can indeed activate the cGAS/STING pathway, highlighting its potential as a therapeutic target for enhancing immune responses in cancer treatment.
What are the protective effects of Curcumin towards pancreatic cancer?
5 answers
Curcumin, a polyphenol derived from turmeric, has demonstrated significant potential in the fight against pancreatic cancer through various mechanisms and formulations. Its anticancer and chemopreventative properties are well-documented across preclinical models of major cancer types, including pancreatic cancer. However, the clinical application of curcumin has been limited by its poor systemic bioavailability. To overcome this, researchers have developed novel formulations such as polymeric nanoparticle-encapsulated curcumin (NanoCurc) and magnetic nanoparticle-loaded curcumin (MNP-CUR), which show enhanced systemic bioavailability and therapeutic efficacy in pancreatic cancer models. NanoCurc, in combination with gemcitabine, has been shown to significantly inhibit tumor growth and abrogate systemic metastases in orthotopic pancreatic cancer xenograft models, suggesting an additive therapeutic influence. Similarly, the MNP-CUR formulation has demonstrated effective growth inhibition of pancreatic cancer cells and tumors in xenograft mouse models, improving survival rates. These formulations also impact the expression of various proteins involved in cancer progression, such as PCNA, Bcl-xL, Mcl-1, and MUC1. Further studies have explored the combination of curcumin with other compounds, such as 10058-F4, showing enhanced antipancreatic cancer actions both in vitro and in vivo. Curcumin analogs, like C66, have been investigated for their anti-inflammatory and anticancer activities, demonstrating inhibition of pancreatic cancer cell proliferation and migration by targeting JNK-mediated inflammation. Additionally, curcumin has been found to regulate Beclin1 expression and inhibit the HIF-1α-mediated glycolytic pathway, thereby inhibiting pancreatic cancer cell proliferation. Research also highlights the importance of enhancing curcumin's bioavailability through novel analogs and nano-formulations for effective pancreatic cancer treatment. Herbal drugs and metabolites, including curcumin, have been reviewed for their potential in pancreatic cancer therapy due to their antioxidant, anti-angiogenic, and anti-metastatic activities. Despite challenges related to bioavailability and rapid elimination, curcumin's modulation of multiple molecular targets and its safety profile in high doses suggest a promising role in pancreatic cancer therapy.
Cancerous mechanism of hydroquinone
4 answers
Hydroquinone (HQ) has been implicated in cancerous mechanisms through various pathways. Studies have shown that HQ induces malignant transformation by altering the cell cycle, inhibiting the p16/pRb signaling pathway, and activating the JNK signaling pathway. Additionally, HQ exposure leads to increased DNA repair through homologous recombination (HR), with the NF-κB pathway playing a crucial role in this process. HQ's pro-apoptotic effects have been observed in leukemia cell models, further highlighting its potential in cancer development. However, mutagenic mechanisms do not seem to be the primary driver of HQ-induced carcinogenesis, as indicated by mutation assays in mice. These findings collectively suggest that HQ contributes to cancer development through complex interactions involving cell cycle dysregulation, DNA repair pathways, and apoptotic mechanisms.
What is the biogenesis of Extracellular vesicles?
10 answers
The biogenesis of extracellular vesicles (EVs) involves complex cellular processes that result in the formation and release of these lipid bilayer-enclosed subcellular bodies into the extracellular space. EVs are produced by most, if not all, cells and play crucial roles in intercellular communication and the horizontal transfer of biological material, including proteins, lipids, nucleic acids, and other bioactive molecules. They are classified into three main types based on their size, biogenesis, and cargo: exosomes, microvesicles (MVs), and apoptotic bodies. Exosomes are the smallest type of EVs, originating from endosomal membranes. Their formation involves the inward budding of endosomal membranes to create multivesicular bodies (MVBs), which then fuse with the plasma membrane to release exosomes into the extracellular space. Microvesicles, or microparticles, are larger than exosomes and are formed through the outward budding and fission of the plasma membrane. Apoptotic bodies, the largest type of EVs, are released from cells undergoing apoptosis and contain a variety of cellular components. The release of EVs is a regulated process influenced by various external and biological stimuli, including circadian rhythms. The regulation of EV biogenesis and secretion is still not fully understood, but it is known to involve a variety of molecular mechanisms and pathways, including the assembly of local microdomains in endocytic membranes for exosomes and in the plasma membrane for ectosomes. Recent studies have also highlighted the role of alcohol in promoting exosome biogenesis and release from hepatocytes through the modulation of specific genes. Understanding the biogenesis of EVs is crucial for exploring their potential in clinical applications, such as drug delivery and diagnostics, as well as their role in physiological and pathological processes. Despite the progress made, much remains to be discovered about the specific mechanisms governing EV formation, cargo sorting, and secretion.
What is Co-culture of sperm?
5 answers
Co-culture of sperm refers to the practice of culturing sperm cells together with other cells or tissues to enhance sperm maturation and function. Various studies have explored the benefits of co-culture in different contexts. For instance, research has shown that co-culture with healthy Sertoli cells can contribute to the maturation and meiotic progression of spermatogonial stem cells. Additionally, studies have highlighted the role of testosterone and its metabolites in promoting the secretion of quiescin sulfhydryl oxidase 1 (QSOX1) in the epididymis, a key factor in sperm maturation. Furthermore, feeder cells like Sertoli cells have been found to promote spermatogonial proliferation through mechanisms such as exosomal exchange, indicating the importance of co-culture systems in expanding spermatogonia for potential clinical applications.
What is the main isoform of Leptin receptor expressed by Schwann cells?
4 answers
The main isoform of the Leptin receptor expressed by Schwann cells is the long isoform Ob-Rb. This isoform is crucial for signal transduction and the transmission of leptin's functions, playing a significant role in energy homeostasis and glucose regulation. While other isoforms exist, such as Ob-Ra, which has been found to mediate some effects of leptin, Ob-Rb stands out due to its full activation signal transduction capabilities and its presence in various tissues, including the hypothalamus where it regulates energy balance. Additionally, leptin receptor isoforms, including Ob-Rb, have been identified in peripheral blood mononuclear cells, with Ob-Rb being expressed alongside the shorter isoform Ob-Ra.
3.General functions and mechanisms of action of Zinc in cellular regulation?
4 answers
Zinc (Zn) is a trace element with multifaceted roles in cellular regulation, impacting a wide array of biological functions through its involvement in the modulation of Zn-dependent proteins, including transcription factors and enzymes pivotal for cell signaling pathways related to proliferation, apoptosis, and antioxidant defenses. It operates within a complex regulatory system, akin to calcium signaling, extending its signaling capabilities to lower concentrations and influencing both intracellular and extracellular regulatory functions. Over the last decade, Zn^2+ has been recognized as a major signaling metal ion, essential for the transmission of information within and between cells, thereby affecting virtually all aspects of cell biology. Zinc's role in male fertility, particularly in sperm motility, capacitation, and acrosomal exocytosis, underscores its importance in successful fertilization, mediated through specific cellular mechanisms and signaling pathways. Its immunomodulatory roles are significant, acting as an ionic regulator of immune responses and participating as an intracellular messenger in signaling pathways that regulate the activity and function of key immune cells. Zinc signaling also regulates immune reactions, with its homeostasis being crucial for preventing immunodeficiency and inflammatory diseases. Furthermore, zinc plays a critical role in energetic metabolism, mitochondrial function, and cytoprotection, essential for maintaining cellular homeostasis and metabolic physiology. Its involvement in neurodevelopment and cognitive function, through the regulation of neurotransmitters and synaptic function, highlights its importance in the nervous system, with dysregulation linked to various neurodegenerative diseases. Zinc signaling, particularly the "Zn wave" in mast cells, is crucial for immune response regulation, influencing inflammatory cytokine gene induction. Lastly, zinc's essentiality in growth, development, and host defense mechanisms, especially in the context of infectious diseases, underscores its critical role in human health.
Where is the soluble reguion of leptin receptor?
4 answers
The soluble region of the leptin receptor is found in the extracellular domain of the receptor. This soluble leptin receptor (sLepR) acts as the main leptin-binding protein in plasma, contributing to the activation of circulating leptin. It plays a crucial role in protecting leptin from degradation in the harsh conditions of the gastric juice. The sLepR binds to leptin, forming a complex that is resistant to gastric degradation, ensuring the stability and functionality of leptin. Additionally, the sLepR is involved in regulating serum leptin concentrations and can transduce signals into cells, impacting various physiological responses to leptin, such as energy homeostasis and immune function. Therefore, the soluble region of the leptin receptor plays a significant role in mediating leptin's actions and maintaining its bioavailability in different physiological contexts.
Is zinc plays a multiple role in cell signaling?
4 answers
Yes, zinc plays multiple roles in cell signaling, acting as a crucial element in both intracellular and extracellular signaling pathways across various biological systems. Zinc's involvement in cell signaling extends from acting as a structural component and cofactor for numerous proteins to functioning as an ionic regulator of immune responses and a signaling molecule within the central nervous system (CNS). Intracellularly, zinc influences kinase signaling pathways, notably ERK and Akt, through fluctuations in labile zinc concentrations, which are not toxic and do not activate stress-dependent signaling under certain conditions. This modulation of signaling pathways underscores zinc's role in healthy cell signaling and its potential impact on cell physiology. Furthermore, zinc has been identified as an intracellular signaling molecule, akin to calcium, influencing various cell functions by converting extracellular stimuli to intracellular signals. In the context of neurotransmission, zinc acts as a neurotransmitter, influencing the function of neurotransmitter receptors and activating specific receptors whose primary ligand is zinc itself, thereby regulating neuronal excitation. Zinc signaling also plays a pivotal role in immune responses, with its homeostasis and signaling mechanisms being essential for the regulation of inflammatory and allergic responses. Zinc signaling is not limited to human physiology; it also occurs in bacteria, fungi, nematodes, and fish, indicating its fundamental role across different organisms. The homeostatic control of cellular zinc metabolism and signaling ensures specificity in zinc's functions, avoiding interference by other metal ions. In the intestinal epithelium, zinc activates signaling pathways that are crucial for maintaining barrier function and overall intestinal health. Collectively, these roles highlight zinc's multifaceted contributions to cell signaling, impacting a wide range of physiological processes and disease states.
What is Imipramine?
5 answers
Imipramine is a tricyclic antidepressant with diverse therapeutic potentials. Research indicates its efficacy in various medical conditions. Imipramine has shown promise in inhibiting the progression of aggressive cancers like triple-negative breast cancer (TNBC) by inducing apoptosis and suppressing metastasis. Additionally, it has been identified as a potential treatment for familial Alzheimer's disease (fAD) and Down syndrome-related Alzheimer's pathology, inhibiting apolipoprotein E and targeting common pathological mechanisms in these conditions. However, imipramine's use may lead to adverse effects, exacerbating conditions like nonalcoholic fatty liver disease, diabetes, diabetic retinopathy, and kidney injury in obese individuals. In the context of malignant glioma, imipramine has demonstrated the ability to inhibit tumor proliferation by targeting the oncogene YAP, suggesting a potential role in combination therapy for glioma treatment.
How gene expression related to the resistance of cancer cells with chemotherapy?
4 answers
Gene expression plays a crucial role in the resistance of cancer cells to chemotherapy. Studies have identified specific pathways associated with resistance, such as the fibroblast growth factor receptor 4 (FGFR4) pathway. Additionally, gene set enrichment analysis has linked pathways like matrix metalloproteinase, p53, methionine metabolism, and the bcl-2 pathway to cytotoxic resistance in cancer cells. Furthermore, in HER2+ breast cancer patients, differentially expressed genes (DEGs) like ATF6B, S100B, and TNXB have been identified as predictors of treatment response and survival outcomes. These findings underscore the complexity of gene regulation in chemotherapy resistance and highlight the importance of understanding molecular signatures to improve treatment strategies and patient outcomes.