scispace - formally typeset
Search or ask a question

How the high calloric diets affect neuroinflamation on hippocampus? 


Best insight from top research papers

High-caloric diets, particularly those high in fat and sugar, have been shown to induce neuroinflammation in the hippocampus. These diets can lead to cognitive decline, memory impairment, and alterations in neurotransmission systems involved in memory regulation. The neuroinflammatory response in the hippocampus includes glial activation, alterations in glutamatergic function, and increased levels of pro-inflammatory molecules. Additionally, high-caloric diets have been linked to a reduction in neurogenesis and an increase in markers of cellular senescence, potentially contributing to cognitive decline. The effects of these diets on the hippocampus can be exacerbated in aging individuals, leading to impaired spatial learning, memory, and increased anxiety levels. Overall, high-caloric diets can have detrimental effects on hippocampal function through the induction of neuroinflammation and related processes.

Answers from top 5 papers

More filters
Papers (5)Insight
High-fat diet induces hippocampal neuroinflammation, reducing IRS2 expression and increasing IRS1, TNFα, and APP levels, while decreasing BDNF and neurogenesis. Abscisic acid supplementation rescues these alterations.
High fat diets induce neuroinflammation in the hippocampus through mechanisms like ER stress, mitochondrial dysfunction, and insulin resistance, negatively impacting cognitive function and glucose transport mechanisms.
High-fat high-sugar diets increase neuroinflammation in the hippocampus of aged rats, as evidenced by elevated levels of glial fibrillary acidic protein (GFAP) cells.
Chronic consumption of a hypercaloric diet induces neuroinflammation in the hippocampus, contributing to cognitive decline in middle-aged female Wistar rats, as shown in the study.
Chronic hyperpalatable diets can induce hippocampal glutamatergic dysfunction and memory impairment without causing neuroinflammation, possibly through modulation of fractalkine levels to counter glutamatergic toxicity.

Related Questions

What are the links between allergies and neuroinflammation?5 answersAllergies 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.
How does neuroinflammation affects metabolism in various tissues?5 answersNeuroinflammation significantly impacts metabolism in various tissues. Studies highlight that neuroinflammation leads to metabolic alterations in brain cells, particularly glial cells, affecting energy homeostasis. Chronic neuroinflammation in conditions like multiple sclerosis triggers metabolic dysfunction in microglia and macrophages, potentially offering therapeutic targets. Moreover, in neurodegenerative diseases such as Alzheimer's, microRNAs like miR-146a play a crucial role in regulating cellular energy metabolism, with upregulation contributing to mitochondrial dysfunction and disease progression. Additionally, obesity-induced neuroinflammation in the hypothalamus exacerbates intracellular disturbances, oxidative stress, and autophagic defects, disrupting neurohormonal signaling and energy balance. Overall, neuroinflammation induces a neurometabolic shift, emphasizing the intricate link between inflammation and metabolism in various tissues.
Does diet effective on the relationship between adverse childhood experiences and brain morphological changes?5 answersResearch indicates that both a diet high in fat and sugar and experiences of early adversity can independently impact brain morphology and function. High energy diets and early life stress have been associated with memory impairments and increased peripheral inflammation, potentially affecting hippocampal neurogenesis. Adverse childhood experiences, such as maltreatment, have been linked to alterations in brain morphology, including gray matter volume and cortical thickness, which can lead to psychopathology and cognitive deficits. Furthermore, childhood adversity, specifically events during sensitive periods, has been shown to be associated with reductions in gray matter in brain regions susceptible to chronic stress, supporting the "sensitive periods" model of brain development. Therefore, diet and adverse childhood experiences can interact to influence brain morphological changes, emphasizing the importance of understanding their combined effects.
How do different fad diets impact the brain?5 answersDifferent fad diets can have varying impacts on brain health. Consuming a Western-style diet (WS-diet) rich in saturated fats, sugar, and salt has been linked to cognitive impairment, neuroinflammation, oxidative stress, and increased risk for neurodegenerative diseases like Alzheimer's and Parkinson's. In contrast, a Mediterranean diet, Okinawan diet, and vegetarian diet have shown to produce neuroprotective effects by reducing oxidative stress and inflammation in the brain. High-fat diets can have both positive and negative effects on the brain, as fat and fatty acids are essential for brain tissue nourishment, but a high-fat diet can also influence astrocyte metabolism and physiology. Additionally, a study on Alzheimer's disease in mice fed a Western diet demonstrated disrupted glucose and fatty acid metabolism, along with T cell recruitment in the brain, highlighting the role of the brain-liver-fat-axis and the adaptive immune system in disrupting brain homeostasis.
How does neuroinflamation effect emotion and behaviour?5 answersNeuroinflammation has a significant impact on emotions and behavior by influencing neural plasticity, neurotransmitter metabolism, and neuroendocrine systems. Studies suggest that neuroinflammation can lead to dysregulation of the hypothalamus-pituitary-adrenal axis, impaired neuroplasticity, and changes in neurotransmitter levels, ultimately affecting cognitive and emotional functions. In conditions like depression and anxiety disorders, dysregulated cytokine networks resulting from neuroinflammation may contribute to the pathogenesis of these affective disorders. Furthermore, neuroinflammation-induced changes in synaptic and non-synaptic transmission in brain regions like the amygdala can lead to anxiety- and depressive-like behaviors. Overall, the complex interplay between neuroinflammation and various brain functions underscores its role in shaping emotions and behaviors in health and disease.
Can neurons become inflamed?9 answers

See what other people are reading

What specific neurotransmitters and hormones does sesamin modulate to improve sleep quality?
5 answers
Sesamin, a lignan found in sesame oil, modulates neurotransmitters and hormones to potentially improve sleep quality. It enhances dopamine biosynthesis by increasing intracellular dopamine levels and tyrosine hydroxylase activity in PC12 cells. Additionally, sesamin has been shown to protect neuronal cells from oxidative stress, potentially impacting neurotransmitter levels. It increases superoxide dismutase activity and reduces inducible nitric oxide synthase expression, suggesting a role in modulating oxidative stress-related pathways. Furthermore, sesamin activates endothelial nitric oxide synthase (eNOS) and nitric oxide production in endothelial cells through TRPV1-calcium signaling, which could influence vascular function and potentially impact sleep quality. These combined effects on dopamine, oxidative stress, and eNOS signaling pathways highlight sesamin's potential role in improving sleep quality through various neurochemical mechanisms.
What is the signal cascade used in the activation of the cholinergic receptors in rat ileum?
5 answers
The activation of cholinergic receptors in rat ileum involves a complex signal cascade. Muscarinic cholinoceptors are stimulated by interferon γ, leading to contractility in the ileum through G protein-regulated pathways involving phosphoinositide turnover and cytoskeletal structures. Additionally, the ascending reflex contraction of the small intestine predominantly relies on cholinergic neurotransmission, with muscarinic M2 receptors playing a key role in the reflex contraction process. These receptors are involved in both prejunctional and postjunctional sites, contributing significantly to the contractile response in the rat ileum. The intricate interplay of muscarinic receptors and their downstream signaling pathways underscores the importance of cholinergic mechanisms in regulating intestinal function and contractility in rats.
How do GSDME and Bcl2 interact to influence neuronal survival?
5 answers
GSDME and Bcl-2 play crucial roles in influencing neuronal survival. Bcl-2, a member of the BCL-2 family, regulates mitochondrial outer membrane permeabilization (MOMP) and impacts neuronal apoptosis and axon degeneration. On the other hand, GSDME is involved in apoptosis induction. Bcl-2 has been linked to cellular glutathione (GSH) status and mitochondrial GSH transport, showing interactions with the mitochondrial GSH transporter OGC. Additionally, Bcl-2 is essential for the survival of immature neurons, acting upstream of BAX to regulate the survival of doublecortin-expressing cells. These interactions highlight the intricate balance between pro-survival (Bcl-2) and pro-apoptotic (GSDME) factors in determining the fate of neurons, emphasizing the importance of understanding their interplay for therapeutic strategies aimed at enhancing neuronal survival.
How is oxidative stress in progeria?
5 answers
Oxidative stress plays a significant role in progeria, particularly Hutchinson–Gilford progeria syndrome (HGPS). The presence of progerin, a mutant form of lamin A, leads to disrupted nuclear transport, affecting the Ran GTPase and inducing oxidative stress. This oxidative stress in HGPS is linked to elevated levels of reactive oxygen species (ROS) and reduced nuclear Ran levels, impacting cellular functions. Additionally, the sequestration of NRF2 by progerin contributes to chronic oxidative stress in HGPS, exacerbating aging defects. The imbalance between oxidants and antioxidants, as seen in oxidative stress, is a key factor in the pathophysiology of progeria, highlighting the importance of redox biology in understanding and potentially targeting age-related diseases like HGPS.
Cons of older fishers dominating the fishing industry?
5 answers
Older fishers dominating the fishing industry can pose several challenges. Research indicates that older fishermen may struggle to keep up with the physical demands of the job due to declining work capacity. Additionally, older fishermen are more prone to occupational accidents and injuries, such as falls, animal bites, and boat accidents, which can impact their safety and well-being. Studies also show that younger fishermen have lower mortality rates compared to older fishermen, suggesting a potential socioeconomic component to the higher death rates among younger crew members in the fishing industry. Therefore, the dominance of older fishers in the industry may lead to safety concerns, decreased productivity, and higher mortality rates among crew members, highlighting the need for age-diverse workforces and targeted safety measures.
How hmg2b regulated in progeria?
5 answers
HMG2B, a high mobility group protein, is regulated in various contexts such as early development, DNA recombination, and adult neurogenesis. In the context of progeria, which is characterized by accelerated aging, the regulation of HMG2B may be impacted. Studies suggest that factors like miRNAs, protein interactions, and chromatin modifications play crucial roles in regulating HMG2B. Additionally, in the context of BRCA2 deficiency, interactions with HMG20b have been implicated in cell division processes, shedding light on potential regulatory mechanisms of HMG2B in conditions like progeria. Understanding how these regulatory pathways intersect and influence HMG2B expression and function could provide insights into its dysregulation in progeria and other aging-related conditions.
What about schizophrenia?
5 answers
Schizophrenia is a severe psychiatric disorder affecting up to 24 million people globally, characterized by cognitive impairments, altered perception, and social dysfunction. It results from a complex interplay of genetic and environmental factors, with substance use disorder, particularly cannabis, being a significant risk factor. Treatment involves antipsychotics, with early intervention crucial for reducing long-term disability. Nonadherence to medication is a common issue, leading to progressive difficulties in symptom management and cognitive function. Building a strong therapeutic alliance between patients and clinicians, emphasizing empathy and compassion, positively impacts treatment adherence and outcomes. Additionally, treatment modalities for schizophrenia include psychotherapy, cognitive behavioral therapy, and pharmacological interventions with antipsychotics. Schizophrenia prevalence is higher in migrants and areas of social deprivation.
Are neurogenesis and neurotrophic theories of depression related?
4 answers
Yes, the neurogenesis and neurotrophic theories of depression are closely related, as evidenced by a comprehensive review of recent research findings. The neurotrophic theory of depression posits that a decrease in neurotrophic factors, particularly Brain-Derived Neurotrophic Factor (BDNF), in response to stress contributes significantly to the pathophysiology of depression. This reduction in BDNF and other neurotrophic factors is believed to lead to the atrophy of critical brain structures such as the hippocampus and prefrontal cortex, which are essential for synaptic plasticity and neuronal survival. BDNF plays a crucial role in neuronal growth, proliferation, synaptic neurotransmission, and neuroplasticity, and its impairment is associated with the development and progression of depression. The neurogenesis theory, on the other hand, suggests that impaired neurogenesis within the hippocampus, a region of the brain associated with mood and cognition, is a contributing factor to the onset of depressive disorders. This theory is supported by findings that defects in hippocampal activity and volume, indicative of reduced neurogenesis, are associated with depression-related behaviors in humans and animals. Moreover, antidepressants, which are known to increase BDNF levels, have been shown to induce the recruitment and integration of newborn neurons into the dentate gyrus, leading to the remission of major depressive disorder (MDD) symptoms. Research has also highlighted the importance of neurotrophic factors in maintaining cell function and their role in the growth, survival, differentiation, and cell cycle of nerve cells. Furthermore, studies have indicated that increasing the survival rate and incorporation of new neurons can alleviate depressive-like behaviors and promote stress resilience, supporting the neurogenic reserve hypothesis. The expression of genes involved in neurogenesis, such as NGF, BDNF, and GDNF, has been found to differ between depressed patients and healthy controls, suggesting their potential as biomarkers for depression. In summary, the neurogenesis and neurotrophic theories of depression are interrelated, with both pointing towards the crucial role of BDNF and neurogenesis in the pathogenesis and treatment of depression. These theories underscore the importance of neuroplasticity and the potential therapeutic benefits of enhancing neurotrophic signaling and neurogenesis in combating depressive disorders.
How does the enteric nervous system contribute to the development and progression of CRC?
10 answers
The enteric nervous system (ENS) plays a multifaceted role in the development and progression of colorectal cancer (CRC), as evidenced by recent research findings. The ENS's interaction with CRC involves a complex interplay of neural, cellular, and molecular mechanisms that contribute to tumorigenesis and tumor growth. Firstly, the ENS contributes to CRC progression through the secretion of factors by enteric neurons, as demonstrated by the N-Myc Downstream-Regulated Gene 4 (NDRG4) research. The absence of Ndrg4 in enteric neurons was associated with enlarged intestinal adenoma development, suggesting that the ENS, via specific genes like NDRG4, influences intestinal tumorigenesis. The secretome of Ndrg4-/- ENS cells, enriched for Nidogen-1 and Fibulin-2, was found to enhance the migration capacities of CRC cells, indicating a direct contribution of ENS-derived factors to colorectal carcinogenesis. Moreover, the ENS's role in CRC is further highlighted by its interaction with the tumor microenvironment (TME). The neuroendocrine-immune axis, involving nerve fibers and signals, has been shown to contribute to various cancers, including CRC. Nerves may promote tumor development through mechanisms such as perineural invasion and neurotransmitter induction, underscoring the ENS's involvement in the TME and its impact on CRC progression. Additionally, the ENS influences CRC through its regulatory effects on immune responses and gut motility. For instance, the presence of a dense mucosal cholinergic innervation is associated with decreased secretion of pro-inflammatory IL-8 in the rectosigmoid of Hirschsprung disease patients, suggesting that cholinergic signals from the ENS can modulate inflammatory responses in the gut, which may have implications for CRC development. Furthermore, the ENS's ability to regulate gut homeostasis and immune responses, as well as its involvement in neurogenesis and tumor-driven neurogenesis, highlights its critical role in GI tumor initiation and progression. The activation of various oncogenic pathways downstream of neural receptors within cancer cells by neurotransmitters and neuropeptides underscores the importance of neural signaling pathways in GI tumor malignancy. In summary, the ENS contributes to CRC development and progression through a variety of mechanisms, including the secretion of tumor-promoting factors, modulation of the tumor microenvironment, regulation of immune responses, and the activation of oncogenic pathways via neural signaling.
How radiation can affect children brain development in having radiotherapy?
5 answers
Radiation therapy can significantly impact children's brain development during radiotherapy, leading to neurocognitive impairment. Young children are particularly vulnerable to radiation-induced neurocognitive dysfunction, with age playing a crucial role in the severity of cognitive deficits post-exposure. Studies show that ionizing radiation can trigger neuronal degeneration and cell death in neurogenic brain regions more prominently in young animals compared to adults, affecting neurocognitive function in a dose- and age-dependent manner. Additionally, factors like total radiation dose, age at treatment, and time since last treatment can influence the risk of late endocrine deficiency following cranial irradiation, further impacting brain development in children. Therefore, understanding the mechanisms and age-related effects of radiation on the brain is essential for developing strategies to mitigate neurocognitive impairments in pediatric patients undergoing radiotherapy.
How these microbiota-mediated gene expression shape neurodevelopmental processes‎?
5 answers
Microbiota play a crucial role in shaping neurodevelopmental processes through their influence on gene expression. The microbiota impact neurogenesis, neurotrophin expression, DNA methylation, and the transcriptome of intestinal epithelial cells, affecting critical signaling pathways in the brain during postnatal development. Studies show that microbiota-responsive genes are involved in immune pathways, metabolic processes, and neurodevelopmental pathways like Wnt signaling. Germ-free conditions lead to altered gene expression in the amygdala, affecting anxiety-related behaviors and social interactions. Microbiota-derived metabolites can rescue neural gene expression changes, highlighting the importance of these metabolites in early neural development. Overall, microbiota-mediated gene expression alterations influence various aspects of neurodevelopment, providing insights into potential mechanisms underlying neurodevelopmental disorders.