scispace - formally typeset
Search or ask a question

Doed ELS-induced OXT system alterations result in depression susceptibility? 


Best insight from top research papers

Early life stress (ELS) has been identified as a significant risk factor for the development of depression, with alterations in the oxytocin (OXT) system playing a crucial role in mediating this susceptibility. Research indicates that ELS, particularly in the form of emotional abuse during infancy or adolescence, can significantly impact the risk of developing depressive symptoms, especially in individuals carrying specific genetic variants of the OXT and its receptor (OXTR) genes. This suggests a complex interplay between genetic predisposition and environmental stressors, where the presence of certain OXT/OXTR genetic variants increases the vulnerability to depression in those who have experienced ELS . The OXT system's involvement in depression is further supported by findings that link polymorphic variations and the methylation status of genes within the hypothalamic-pituitary-adrenal (HPA) axis, including OXT and OXTR, with phenotypic variations in depression levels. This association underscores the biological underpinnings of depression and highlights the potential of these genetic and epigenetic markers as biomarkers for depressive behavior . Moreover, the broader context of stress-induced alterations in the immune system, oxidative stress dysregulation, and neuroplastic changes also contributes to the understanding of depression's etiology. Dysregulation of innate immune responses, oxidative stress markers, and neuroplastic phenomena have all been associated with the development and recurrence of depression . These findings collectively suggest that ELS-induced alterations in the OXT system, in conjunction with genetic predispositions and other stress-related biological changes, significantly contribute to an individual's susceptibility to depression. This multifaceted approach underscores the importance of considering both genetic and environmental factors in the prevention and treatment of depression.

Answers from top 4 papers

More filters

Related Questions

How does genetics influence depression?5 answersGenetics plays a significant role in the development of depression. Various genetic factors, such as gene polymorphisms, have been associated with depressive disorders, particularly in adolescents. Additionally, genetic factors can influence drug metabolism and contribute to increased drug resistance in patients with depression. The study of genetic nurture, which considers the effects of parental genotypes on the environment experienced by their children, is essential in understanding the genetic and environmental influences on depression. Preliminary evidence suggests that genetic nurture may be associated with depression and neuroticism in adults. However, despite extensive research, no single genetic variation has been identified to significantly increase the risk of depression. Further studies are needed to unravel the complex genetic mechanisms underlying depression and to identify pathways involved in its development.
What is the effect of DNA methylation on the oxytocin receptor gene in depression?5 answersDNA methylation of the oxytocin receptor (OXTR) gene appears to have an effect on depression. Studies have shown that increased methylation levels of the OXTR promoter region are associated with deficits in social, cognitive, and emotional functioning. In patients with major depression, stressful life events were negatively associated with OXTR methylation, suggesting a dysregulation of the oxytocin system due to life stress. Additionally, OXTR methylation patterns were found to change through psychotherapy, and differences in methylation were observed between depressed patients who remitted and those who did not. In pregnant women, higher oxytocin plasma levels were associated with a favorable methylation profile of clock genes, which may have a protective role in the development of perinatal depression. Epigenetic alterations in OXTR methylation have also been found in individuals with recent-onset schizophrenia, particularly in relation to negative symptoms such as anhedonia-asociality.
How does the Nrf2 pathway contribute to oxidative stress in depressive disorders and what are potential therapeutic targets?5 answersThe Nrf2 pathway plays a crucial role in oxidative stress in depressive disorders. Studies have shown that Nrf2 levels are lower in depression and that antidepressant treatments can increase Nrf2 expression. Oxidative stress-induced neuronal damage is implicated in the pathogenesis of neurodegenerative disorders, including depression. Nrf2 activation can regulate the expression of antioxidant genes, reducing oxidative stress and its damages. In major depressive disorder (MDD), oxidative stress biomarkers, such as modifications to antioxidant enzymes and non-enzymatic antioxidants, are altered. Antidepressant treatment can correct these changes and normalize oxidative stress markers. Targeting Nrf2 has potential therapeutic benefits in depression, and compounds that activate Nrf2 have been identified as potential treatment options. Overall, Nrf2 activation and the modulation of oxidative stress-related pathways hold promise as therapeutic targets for depressive disorders.
How is pcos and depression related?5 answersPCOS and depression are related, with studies showing a higher incidence of depression in women with PCOS compared to those without the condition. The exact mechanism of this relationship is still unknown, but several factors may play a role, including increased BMI, infertility, high cortisol levels, body image, vitamin D deficiency, and elevated inflammation markers. Additionally, women with PCOS may experience poor mental health and a lower quality of life, which can contribute to the development of depression. The bidirectional two-sample Mendelian randomization analysis suggests a potential causal association between PCOS and an elevated risk of depression. It is important to tailor PCOS management to women's life stage and provide age-appropriate and patient-centered healthcare, including screening for depressive symptoms and lifestyle counseling. Early psychological intervention for PCOS may also have anti-depression benefits. Further research is needed to fully understand the pathogenesis and treatment of depression in PCOS.
What is the relationship between SNPs and the risk of developing depression?4 answersSingle nucleotide polymorphisms (SNPs) have been found to be associated with the risk of developing depression. In a study conducted in a Chinese population, the rs155979 SNP in the lncRNA NONHSAT102891 was significantly associated with an increased risk of depression. Another study conducted a genome-wide association meta-analysis and identified 243 risk loci for major depression (MD), including genes involved in glutamate and GABA receptors. Additionally, a study investigating gene-environment interactions found that certain genetic factors were only relevant in individuals with moderate and/or high stress exposure, suggesting a gene-environment interaction in the development of depression. Furthermore, a genetic-risk score (GRS) combining multiple depression risk SNPs was found to be significantly associated with a higher risk of depression, and the predictive ability for depression was improved when non-genetic risk factors such as body mass index (BMI) were included in the model. These findings suggest that SNPs play a role in the risk of developing depression, and their interaction with environmental and non-genetic factors may further influence this risk.
Do neural alterations associated with the familial risk of depression manifest in a similar manner throughout development?5 answersNeural alterations associated with familial risk of depression manifest in a similar manner throughout development. Studies have shown that youth at high familial risk for depression exhibit weaker functional connectivity (FC) patterns in brain regions implicated in reward and emotional processing, such as the caudate and angular gyrus. These alterations are primarily driven by maternal risk and are evident even in youth who do not currently have depression. Additionally, asymptomatic offspring at high familial risk for depression show structural and functional brain changes similar to those found in adults with depression, including reduced cortical thickness, altered white matter integrity, and altered striatal reward processing. Furthermore, in a study involving both healthy and depressed relatives of individuals with major depressive disorder (MDD), widespread increased fractional anisotropy (FA) was found in healthy adults at familial risk for MDD, potentially reflecting compensatory neural mechanisms associated with resilience.

See what other people are reading

How a glutathione precursor impact infant cancer?
5 answers
The glutathione precursor, gamma-L-glutamyl-L-cysteine (γGlu-Cys), plays a crucial role in inhibiting oxidative injury, which is significant in cancer development. Glutathione (GSH), the primary antioxidant in humans, is closely linked to cancer progression and treatment outcomes. Studies have shown that the glutathione-S-transferase (GST) polymorphism, involved in detoxifying chemotherapeutic agents, impacts the outcome of childhood B-precursor acute lymphoblastic leukemia (ALL). Disruptions in GSH synthesis and changes in the GSH/GSSG ratio are common in malignant neoplasms, affecting tumor cell viability, drug resistance, and tumor progression. Therefore, by influencing oxidative stress and detoxification processes, a glutathione precursor like γGlu-Cys can potentially impact infant cancer by modulating cellular responses to oxidative damage and chemotherapeutic agents.
Does low NCOA4 levels induce ferroptosis in macrophages?
5 answers
Low levels of NCOA4 have been associated with inducing ferroptosis in macrophages. Studies on lung adenocarcinoma and COPD emphysema have shown that decreased NCOA4 expression leads to increased ferroptosis in macrophages, particularly the M2 phenotype, contributing to the pathogenesis of these conditions. Additionally, in clear cell renal cell carcinoma (ccRCC), low NCOA4 expression is linked to enhanced ferroptosis and reduced infiltration of immune cells, including CD8+ T cells, which can impact disease progression and patient prognosis. Furthermore, research on sepsis has revealed that the interaction between STING and NCOA4 can trigger ferritinophagy-mediated ferroptosis in macrophages, exacerbating multiple organ injury during sepsis. Therefore, low NCOA4 levels indeed play a role in inducing ferroptosis in macrophages across various pathological conditions.
How does nutrition impact the immune system in obesity?
5 answers
Nutrition plays a crucial role in modulating the immune system in obesity. The Western diet and sedentary lifestyle contribute to the rising obesity rates, leading to chronic inflammation and altered tissue functions. Obesity-related inflammation is linked to cellular senescence, characterized by irreversible cell cycle arrest and the secretion of pro-inflammatory factors. Both undernutrition and overnutrition have significant impacts on immunity, with undernutrition causing immunosuppression and overnutrition leading to chronic inflammation, increasing the risk of metabolic and cardiovascular diseases. An anti-inflammatory diet can reduce markers of inflammation and improve immune status in individuals with obesity, highlighting the importance of dietary interventions in managing immune responses in obesity.
What is known about opioids and chlamydia infections?
5 answers
Opioids have been associated with an increased risk of infections, including bacterial infections like chlamydia. Studies have shown that opioids, particularly those with immunosuppressive properties like morphine, can increase susceptibility to serious infections, with morphine specifically linked to gut barrier dysfunction and altered gut microbiome, promoting bacterial dissemination and virulence. Opioids, including morphine, have been found to suppress the immune system, affecting innate and adaptive immunity cells, which are crucial in defending against bacterial infections like chlamydia. Furthermore, opioids and their receptors have been shown to impact viral infections, indicating a complex relationship between opioids and infectious diseases, including bacterial infections like chlamydia.
Is phagocytic receptor binding promotes receptor clustering which is the key initiator of phagocytosis an accurate interpretation?
4 answers
Phagocytic receptor binding does indeed promote receptor clustering, which is a crucial step in initiating phagocytosis. The clustering of phagocytic receptors generates signals that trigger the actin-dependent engulfment of particles. Specifically, when ligands are clustered closer together, it enhances engulfment efficiency by increasing receptor phosphorylation and downstream signaling. Additionally, the activation of receptors after binding immunoglobulin G (IgG) leads to the formation of the phagocytic cup, culminating in the ingestion of foreign particles. Moreover, the intracellular domains of receptors play a role in cytoskeletal coupling during phagocytosis, with tyrosine phosphorylation facilitating this process. Therefore, it is accurate to interpret that phagocytic receptor binding promotes receptor clustering, which is a key initiator of phagocytosis.
What affect does formaldehyde have on mitochondria and intracellular calcium?
5 answers
Formaldehyde has significant effects on mitochondria and intracellular calcium levels. Exposure to formaldehyde leads to cytotoxicity, apoptosis, and necrosis, affecting cellular physiology. Formaldehyde induces misfolding of neuronal tau protein, hippocampal neuronal apoptosis, and cognitive impairment. It also triggers oxidative stress, reduces mitochondrial membrane potential, inhibits mitochondrial respiration, and induces apoptosis in sensitive neuronal cells. Mitochondrial formaldehyde levels can damage mitochondria by decreasing membrane potential and inhibiting respiration. Additionally, formaldehyde exposure can increase intracellular calcium levels in cultured hippocampal neurons, potentially involving NMDAR and T-type Ca2+ channels. In summary, formaldehyde disrupts mitochondrial function, leading to structural rearrangements, DNA double-strand breaks, and activation of mitophagy, while also impacting intracellular calcium signaling pathways.
Does phagocytic receptor signalling require both ligand binding or clustering? or just one of the 2?
4 answers
Phagocytic receptor signaling necessitates both ligand binding and clustering for an effective response. Studies show that reducing ligand spacing enhances engulfment efficiency by increasing engulfment-initiation processes. Furthermore, the clustering of multiple receptors is crucial for enhancing receptor phosphorylation and downstream signaling, indicating that both ligand density and clustering play vital roles in phagocytosis decisions. Additionally, simultaneous activation of multiple opsonic receptors leads to enhanced particle binding and internalization, highlighting the importance of engaging multiple receptors for efficient phagocytosis. The process of phagocytosis involves complex signaling mechanisms that are triggered by both ligand binding and receptor clustering, emphasizing the significance of both aspects in the engulfment process.
How does the duration and quality of sleep affect muscle growth and recovery?
5 answers
The duration and quality of sleep play crucial roles in muscle growth and recovery. Research indicates that sleep deprivation can lead to ultrastructural changes in skeletal muscles, including tissue degeneration and muscle atrophy. Additionally, poor sleep quality and short sleep duration are associated with decreased muscle strength in university students. Studies on Chinese adults have shown that both short and long sleep durations, as well as insomnia, are linked to low muscle mass and strength. Furthermore, sleep restriction has been found to reduce skeletal muscle protein synthesis in humans, potentially impacting muscle quality, although regular high-intensity interval exercise during sleep restriction may help maintain muscle protein synthesis rates. Overall, adequate sleep duration and quality are essential for optimal muscle growth, repair, and maintenance.
How the immune system works?
5 answers
The immune system functions by distinguishing between self and non-self entities, aiming to eliminate the latter. It comprises the innate immune system, offering a rapid response against common pathogens, and the adaptive immune system, evolving to combat new invaders. Specialized cells present antigens to T cells, activating them for defense. T cells orchestrate immune responses, while B cells produce antibodies for protection. The immune system maintains self-tolerance and memory for efficient defense. It can be modulated by agents like azathioprine and monoclonal antibodies, enhancing responses or treating diseases. Understanding the immune system is crucial for pharmacists due to the increasing use of biotechnological products in healthcare.
How does fatty diet affect immunity?
5 answers
A high-fat diet (HFD) impacts immunity by inducing inflammation and altering immune cell populations in adipose tissue. Studies show that HFD consumption leads to increased infiltration of pro-inflammatory Th17 cells, dendritic cells, and macrophages, while reducing regulatory T cells. This results in elevated levels of inflammatory cytokines and chemokines, along with changes in key signaling pathways like PPAR-γ. Additionally, dietary fatty acids, particularly saturated fatty acids like palmitic acid (PA), can act as inflammatory stimuli, inducing a hyper-inflammatory response and altering immune memory. PA exposure enhances inflammation in macrophages through ceramide synthesis, contributing to long-lived innate immune memory. Conversely, oleic acid reverses PA-induced inflammation. These findings highlight the significant impact of fatty diets on immunity, influencing immune cell function and inflammatory responses.
What is the controversial effect of streptococcus mutans on health and dental plaque?
4 answers
Streptococcus mutans (S. mutans) has a controversial role in oral health and dental plaque formation. Research indicates that S. mutans dexA gene deletion alters biofilm structure and microbiota composition, potentially reducing cariogenicity. Conversely, S. mutans abundance is associated with oral squamous cell carcinoma (OSCC) development and progression, suggesting a detrimental impact on oral health. Furthermore, the absence of S. mutans in some cases of severe early childhood caries (SECC) highlights a potential protective effect against caries, with other bacteria like Leptotrichia, Selenomonas, and Prevotella_7 identified as biomarkers in S. mutans-low SECC cases. Additionally, the rhamnose-glucose polysaccharide (RGP) of S. mutans plays a crucial role in biofilm formation and stress response, with implications for oxidative stress tolerance and iron uptake.