scispace - formally typeset
Search or ask a question

Do N2A neuroblastoma cells have greater glutamate uptake than COS-7 cells? 


Best insight from top research papers

N2A neuroblastoma cells have been shown to have a concentration-dependent response to L-glutamate, as well as other glutamate analogues, resulting in an inward current . These cells have also been used as a model for studying excitatory amino acid properties . However, there is no information available in the provided abstracts regarding the glutamate uptake in N2A neuroblastoma cells compared to COS-7 cells.

Answers from top 4 papers

More filters
Papers (4)Insight
The provided paper does not mention N2A neuroblastoma cells or COS-7 cells. Therefore, there is no information in the paper to answer the question about the glutamate uptake of these specific cell types. The paper is about the presence of functional NMDA receptors in a human neuroblastoma cell line.
The provided paper does not mention anything about the glutamate uptake of N2A neuroblastoma cells compared to COS-7 cells.
The provided paper does not mention anything about the glutamate uptake of N2A neuroblastoma cells compared to COS-7 cells. The paper focuses on the evaluation of calcium oscillations and neural spikes in mouse neuroblastoma N2A cells in an epileptic condition.
The provided paper does not mention anything about the glutamate uptake of N2A neuroblastoma cells or COS-7 cells.

Related Questions

What is the role of glutamate in neurons as an energetic substrate?5 answersGlutamate plays a crucial role in neuronal energy metabolism by serving as an energetic substrate. It can be utilized for ATP synthesis, especially during hypoxia/reoxygenation injury, where glutamate supplementation can improve cell survival and ATP levels. Glutamate influences neuronal energy metabolism by regulating the expression of key mitochondrial proteins like AQP9, MCT2, and LDHA. Additionally, the enzyme glutamate dehydrogenase (GDH) is essential for connecting amino acid, carbohydrate, neurotransmitter, and oxidative energy metabolism in neurons. Glutamate metabolism is intricately linked to the glutamate-glutamine cycle, highlighting its significance in sustaining glutamatergic activity and neurotransmission. Overall, glutamate serves as a vital energy substrate in neurons, contributing to their metabolic functions and survival under various conditions.
What is the relationship between GABA and Glutamate?5 answersThe relationship between GABA and Glutamate is crucial for maintaining physiological brain functions. Both neurotransmitters play significant roles in various processes, including the sleep-wake cycle. Glutamate is primarily involved in excitatory synaptic functions, plasticity, and excitotoxicity, while GABA underlies inhibition and can impact psychophysical functions. Studies have shown that disturbances in GABAergic and glutamatergic neurotransmission can lead to serious health issues, affecting neurodegeneration, affective disorders, and sleep problems like obstructive sleep apnea (OSA). In the context of glaucoma, a decline in GABA levels, rather than glutamate, has been linked to reduced neural specificity in the visual cortex, suggesting the importance of GABA in maintaining neural function. Additionally, changes in glutamate levels have been associated with motor learning, highlighting the intricate interplay between GABA and glutamate in cognitive processes.
Do extracellular glutamate levels depend on vesicular release of glutamate?5 answersExtracellular glutamate levels are influenced by vesicular release mechanisms. Studies suggest that glutamate release can occur through full fusion of vesicles with the membrane or via 'kiss-and-run' mechanisms, where glutamate is released through a fusion pore. During brain ischemia, the cystine/glutamate antiporter plays a crucial role in elevating extracellular glutamate levels, leading to neuronal damage through overactivation of extrasynaptic NMDA receptors. Research on cultured neurons indicates that glutamate release can deplete vesicular stores, with sustained release dependent on Ca2+ levels and vesicular mechanisms. Furthermore, vesicular filling of glutamate, removal of Synapsin I, and Ca2+-dependent exocytosis are key steps in the release process. Additionally, satellite glial cells have been shown to release glutamate in a calcium-dependent manner, with potential modulation by botulinum neurotoxin type A.
Does glutamate play a role in ALS?5 answersGlutamate plays a role in ALS. Increased extracellular glutamate in ALS and cerebral ischemia stimulates the utilization of glutamate as metabolic fuel, suggesting a beneficial role for glutamate in these conditions. Glutamatergic excitotoxicity or dysregulation of glutamatergic signaling is considered a mechanism underlying motor neuron death in ALS. Altered glutamate homeostasis is believed to be involved in ALS pathogenesis, and the approved treatment for ALS, riluzole, modulates glutamate. Glutamate metabolism disorders are connected to TDP-43 proteinopathy, a pathological mechanism in ALS. Glutamate-mediated corticomotor neuronal hyperexcitability is correlated with ALS, and disruption of the neuron-astrocyte glutamate-glutamine cycle may retard ALS progression.
Why when transfecting gene into Neuro-2a from the media not direct transfer still occurs?5 answersWhen transfecting genes into Neuro-2a cells, it is more common to use a transfection reagent in the culture medium rather than directly transferring the gene. This is because using a transfection reagent, such as TransFectin™, can significantly improve the transfection efficiency of Neuro-2a cells, resulting in higher rates of successful gene transfer. Direct transfer of genes into Neuro-2a cells without a transfection reagent can lead to lower transfection efficiency, making it more difficult to achieve successful gene transfer. Using a transfection reagent optimizes the transfection conditions, including factors such as pH, which can greatly enhance the efficiency of gene transfer into Neuro-2a cells. Additionally, using a transfection reagent-based system does not cause detectable cytotoxicity to the cells, making it a safe and effective method for gene transfection in Neuro-2a cells.
Are there NMDA receptors on GABA neurons?3 answers

See what other people are reading

What is the molecular structure of a prion protein?
5 answers
The molecular structure of a prion protein involves key aspects such as metal ion binding, beta-sheet formation, glycolipid anchors, and intermolecular beta sheets. Prion proteins like PrPC can bind Cu(II) and Zn(II) ions, leading to structural transitions from polyproline II helices to beta-turn and beta-sheet structures. Additionally, infectious prions are amyloid fibrils with parallel in-register intermolecular beta sheets, where residues 95-227 form the ordered fibril core, while glycans and glycolipid anchors project from the fibril's surfaces. The globular domain of PrPC can dimerize through domain swap or non-covalent association, with potential implications for the conversion process to the pathogenic form. Understanding these structural features is crucial for developing anti-prion therapies targeting the stabilization of the native conformation of PrPC.
What is the current understanding of the physiological effects of piretrine and pyrethroid exposure on humans?
5 answers
Current research indicates that exposure to pyrethroids like allethrin and prallethrin can induce various physiological effects in humans. Pyrethroids, synthetic derivatives of natural pyrethrins, primarily target the sodium channels in insect neurons, potentially leading to toxicity in vertebrates, including humans. Acute exposure to pyrethroids can enhance the late sodium current in the heart, causing cardiac arrhythmias, while long-term exposure may result in oxidative stress and tissue remodeling. Furthermore, studies have shown that exposure to pyrethroids equivalent to human consumption levels can lead to systemic and reproductive toxicity, impacting organ function, antioxidant status, and anatomical structures. These findings emphasize the importance of understanding the potential health risks associated with pyrethroid exposure in humans.
What is the effect of incubation time of a dye on the fluorescent signal in microscopy?
5 answers
The effect of incubation time of a dye on the fluorescent signal in microscopy varies depending on the specific dye used. Longer incubation times can lead to brighter and more stable fluorescence signals in certain dyes. For instance, the hydrophobic fluorescence dye NAO shows bright green fluorescence in mitochondria with an incubation time of less than 1 minute, and this fluorescence can last longer than 7 days. Similarly, calcium dyes used in neuroscience studies remain functional and exhibit similar calcium dynamics even after more than 24 hours of loading, indicating the effectiveness of prolonged incubation periods. On the other hand, optimizing the illumination process by allowing dark states to relax between molecular absorption events can significantly increase fluorescence yield before bleaching, as observed with GFP and Rhodamine dyes.
How does gadolinium retention differ between healthy individuals and those with neurological disorders, such as Alzheimer's disease?
5 answers
Gadolinium retention differs between healthy individuals and those with neurological disorders like Alzheimer's disease. In healthy individuals, gadolinium-based contrast agents (GBCAs) can lead to gadolinium retention in various tissues, including the brain, liver, skin, and bone. Studies have shown that gadolinium can be deposited in the brain, particularly in deep gray matter regions, even in patients without renal impairment. However, in patients with multiple sclerosis (MS), gadolinium retention in the brain, specifically in the dentate nuclei, does not seem to impact long-term motor or cognitive outcomes. Furthermore, in an experimental autoimmune encephalomyelitis (EAE) model, neuroinflammation was found to enhance gadolinium retention in the brain, with differences observed between macrocyclic and linear GBCAs in terms of retention profiles and potential neurotoxicity.
How does in silico exploration aid in the discovery of potential anti-Parkinson effects for Punica Granatum?
5 answers
In silico exploration plays a crucial role in identifying potential anti-Parkinson effects for Punica Granatum by leveraging computational tools to predict interactions between bioactive compounds and target proteins. Studies have shown that Punica Granatum, rich in polyphenols like ellagitannins, exhibits neuroprotective properties through antioxidant, anti-inflammatory, and mitochondrial regulatory mechanisms. Computational analyses aid in understanding how these compounds interact with neuronal and glial cells, influence blood-brain barrier function, and restore redox balance in the brain. By simulating molecular interactions, in silico approaches provide insights into the mechanisms through which Punica Granatum polyphenols may mitigate Parkinson's disease symptoms, offering a promising avenue for the development of novel anti-Parkinson agents.
How does decreased intracellular free calcium concentration contribute to the development and progression of SMA disease?
5 answers
Decreased intracellular free calcium concentration plays a crucial role in the development and progression of Spinal Muscular Atrophy (SMA) by impacting presynaptic terminal function. In SMA, alterations in mitochondrial calcium signaling and defects in the secretion machinery significantly contribute to presynaptic terminal dysfunction. Calcium ions are essential for neurotransmitter release and synaptic plasticity. The impairment of endocytosis in SMA may be linked to functional changes in presynaptic mitochondria during action potential firing, affecting calcium dynamics. This disruption in calcium signaling within the nerve terminals can lead to reduced neurotransmitter release, ultimately contributing to the motor neuron degeneration characteristic of SMA.
How does glutamatergic neurotransmission affect the development and function of the central nervous system?
5 answers
Glutamatergic neurotransmission significantly influences the development and function of the central nervous system. Glutamate, the major excitatory neurotransmitter, plays crucial roles in various physiological functions. It is involved in processes like memory, learning, and motor planning, while also regulating peripheral nervous and endocrine systems. Dysfunctions in glutamatergic signaling are associated with neurodevelopmental disorders, impacting higher cognitive functions. Glutamate affects neural progenitor cells, promoting or inhibiting their proliferation, survival, migration, and differentiation through ionotropic and metabotropic receptors. The precise control of glutamatergic signaling is vital, as excessive transmission can lead to excitotoxicity and cell damage. Understanding the mechanisms of glutamatergic neurotransmission is essential for developing potential therapeutics for various neurological and psychiatric conditions.
How does the kindling effect contribute to the development of depression in people with ADHD?
5 answers
The kindling effect, a phenomenon of lasting brain alterations from repeated stimulation, plays a role in the development of depression in individuals with ADHD. Research suggests that individuals with both ADHD and depression exhibit more severe impairments than those with either condition alone. The N-methyl-d-aspartate (NMDA) receptor subtype is implicated in limbic kindling, affecting seizure development. Kindling involves tetanic activation in brain regions like the limbic system, leading to behavioral changes akin to learning processes. This effect can be utilized in laboratory settings for learning experiences related to brain stimulation and behavior observation. Therefore, the kindling effect, through its impact on brain function and behavior, contributes to the development of depression in individuals with ADHD.
What is the molecular mechanism behind the formation of vesicles on skin?
5 answers
The formation of vesicles on the skin involves various molecular mechanisms. One key mechanism involves the aggregation and transformation of micelles into vesicles induced by specific molecules like doxorubicin and surfactants. Additionally, the presence of stress vesicles in epidermal stem cells under mechanical stress conditions, regulated by Piezo1 ion channels and calcium dynamics, plays a crucial role in determining cell fate. Ultradeformable vesicles, composed of phosphatidylcholine and surfactants, enhance transdermal drug delivery through mechanisms like vesicle-skin adhesion, fusion, and potential penetration through hair follicles. Furthermore, extracellular vesicles derived from multipotent stromal cells have shown efficacy in skin wound healing, with the need for standardized processing methods to ensure consistent therapeutic outcomes. These studies collectively highlight the diverse molecular mechanisms involved in vesicle formation and their implications for skin health and drug delivery.
How does magnesium activate GABA receptors?
5 answers
Magnesium plays a crucial role in modulating GABA receptors by affecting their function in various ways. Studies have shown that physiologically relevant concentrations of magnesium can impact GABA responses on GABAA receptors, both in native and recombinant receptor subtypes. Additionally, in conditions where magnesium levels are low, such as in neurotic patients, there is a decrease in GABAergic cerebral system activity, highlighting the importance of magnesium in GABA receptor function. Furthermore, under Mg2+-free conditions, the activation of GABAA receptors during the neonatal period can lead to depolarization and the generation of epileptiform discharges, emphasizing the intricate relationship between magnesium and GABA receptor activation. Magnesium's ability to modulate GABA receptors is essential for maintaining the balance between excitation and inhibition in the central nervous system.
What reaction is H2S mainly involved in within the mitochondria?
4 answers
Hydrogen sulfide (H2S) is primarily involved in the mitochondrial sulfide oxidation pathway within the mitochondria. This pathway includes enzymes like sulfide quinone oxidoreductase (SQOR), which catalyzes the oxidation of sulfide to prevent H2S accumulation and generate reactive persulfide species. Additionally, H2S is metabolized in mitochondria by various enzymes like persulfide dioxygenase and sulfite oxidase. H2S oxidation in mitochondria produces reactive sulfur species that can modify proteins through sulfhydration, impacting processes like protein persulfidation and mitochondrial function. Furthermore, H2S has been shown to interact with cytochrome c in the electron transport chain, potentially influencing sulfide-based signaling pathways within the mitochondria.