Next Article in Journal
Selenoprotein P in a Rodent Model of Exercise; Theorizing Its Interaction with Brain Reward Dysregulation, Addictive Behavior, and Aging
Previous Article in Journal
Follistatin as a Potential Biomarker for Identifying Metabolically Healthy and Unhealthy Obesity: A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

IL-8 (CXCL8) Correlations with Psychoneuroimmunological Processes and Neuropsychiatric Conditions

by
Anton Shkundin
and
Angelos Halaris
*
Department of Psychiatry and Behavioral Neurosciences, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2024, 14(5), 488; https://doi.org/10.3390/jpm14050488
Submission received: 18 March 2024 / Revised: 26 April 2024 / Accepted: 30 April 2024 / Published: 3 May 2024
(This article belongs to the Section Disease Biomarker)

Abstract

:
Interleukin-8 (IL-8/CXCL8), an essential CXC chemokine, significantly influences psychoneuroimmunological processes and affects neurological and psychiatric health. It exerts a profound effect on immune cell activation and brain function, suggesting potential roles in both neuroprotection and neuroinflammation. IL-8 production is stimulated by several factors, including reactive oxygen species (ROS) known to promote inflammation and disease progression. Additionally, CXCL8 gene polymorphisms can alter IL-8 production, leading to potential differences in disease susceptibility, progression, and severity across populations. IL-8 levels vary among neuropsychiatric conditions, demonstrating sensitivity to psychosocial stressors and disease severity. IL-8 can be detected in blood circulation, cerebrospinal fluid (CSF), and urine, making it a promising candidate for a broad-spectrum biomarker. This review highlights the need for further research on the diverse effects of IL-8 and the associated implications for personalized medicine. A thorough understanding of its complex role could lead to the development of more effective and personalized treatment strategies for neuropsychiatric conditions.
Keywords:
IL-8; CXCL8; CXCR1; CXCR2; SNPs

1. Introduction

The intricate balance between the neuroendocrine and immune systems is critical for maintaining an organism’s homeostasis. During times of stress, the redistribution of immune cells across various immune compartments becomes crucial, ensuring the efficiency of cell-mediated immune responses [1]. The activation of the immune system in the periphery impacts the central nervous system (CNS), and disturbances in systemic immune functions can significantly contribute to the onset of neuropsychiatric conditions [2]. Dysregulation in neuroimmune interactions may lead to dysfunction in vital organs and result in the widespread impairment of neuromodulation and symptoms across multiple body systems [3]. Peripheral afferent neurons activated by the immune system, along with circulating cytokines and microbial products, stimulate neurons and glial cells in the hypothalamus and medulla, triggering sympathetic and humoral responses [3,4]. The gaining of insight into the comorbidities between psychiatric conditions and cardiovascular, cerebrovascular, and neurological disorders has been greatly enhanced through the perspective of psychoneuroimmunology, revealing the complex interconnections between the brain and the immune system [5,6].
Neuroinflammatory processes influence the health of the nervous system, exerting control over the development and viability of brain cells and their connections [7]. CNS immune responses can lead to synaptic dysfunction, neurotransmitter imbalances or deficiencies, neuronal loss, and the exacerbation of brain-related pathologies [5]. Neuroinflammation, implicated in a diverse array of CNS disorders, including autoimmune and degenerative pathologies, often manifests as a dysregulated chemokine system [7,8].
Chemokines, also referred to as chemotactic cytokines, comprise a diverse family of relatively small proteins, ranging from 8 to 12 kDa. These proteins are secreted and play a critical role in inducing chemotactic responses in immune cells, and participate in numerous inflammatory processes [9,10,11]. Their distinctive features lie in the presence of three to four conserved cysteine residues, which leads to their categorization into four families (CXC, CC, CX3C, and C) based on the arrangement of these N-terminal cysteine residues [12,13,14,15].
Chemokines play a key role in controlling the development and equilibrium of the immune system. They actively participate in all aspects of both protective and detrimental immune and inflammatory reactions [10]. These proteins can attract and activate a wide range of cells, including both immune and non-immune varieties [12,14]. Beyond their role in inducing chemotaxis, chemokines also exert control over cell proliferation, survival, and differentiation [16].
Chemokines and their receptors play a prominent role in facilitating communication between neurons and inflammatory cells, a crucial aspect of normal neuronal function [7]. These molecules and their receptors are naturally expressed in the brain, under physiological conditions. Their presence suggests their potential involvement in intercellular communication and the modulation of neuronal activity, in addition to their well-known immunological functions. Furthermore, chemokines are actively engaged in brain development and in the maintenance of brain homeostasis, influencing synaptic activities, as well as the processes of migration, differentiation, and proliferation in both glial and neuronal cells [8,14,17,18,19,20].
The recruitment of inflammatory cells is a well-recognized driver of the secondary damage cascades commonly observed in CNS injuries. Changes in the expression of chemokines drive the activation and infiltration of cells to the injury site. This post-traumatic infiltration of inflammatory cells has been linked to secondary tissue damage, cell death, and the demyelination of axons [21]. Furthermore, the potential of chemokines to induce neuronal death, either directly through the activation of neuronal chemokine receptors or indirectly by triggering microglial destroying mechanisms, underscores their significant role in CNS injury [7,8,13].

2. IL-8 and CXCR1/2 Receptors

Within the chemokine family, the CXC or α chemokines, are characterized by a single amino acid separating the first two cysteine residues, denoted as cysteine-X amino acid-cysteine (CXC). Among the CXC chemokines, there is a subgroup distinguished by the presence of a specific three-amino-acid motif near their N-terminal region, known as the glutamic acid-leucine-arginine (ELR) motif [12].
Interleukin-8 (IL-8), also recognized as neutrophil-activating peptide 1 (NAP1) and CXC chemokine ligand 8 (CXCL8) [22], is a proinflammatory CXC chemokine that plays a prominent role in inducing neutrophil chemotaxis, the release of intracellular granule contents, and the upregulation of cell surface adhesion molecules [23,24,25].
IL-8 belongs to the ELR+ CXC chemokine family, known for its diverse biological functions. It serves a critical role in guiding neutrophils and promoting angiogenesis [26]. IL-8 exists in two forms, a monomeric and a dimeric form, and this distinction can lead to different effects on its CXCR1/2 receptors, including desensitization and receptor internalization [26,27].
The action of IL-8 involves several cellular responses, including alterations in cytoskeletal structure, fluctuations in intracellular calcium concentrations, the activation of integrins, the release of granule proteins through exocytosis, and the initiation of a respiratory burst [28].
The production and release of IL-8 can be modulated by multiple factors that influence its expression and levels. For instance, IL-8 may be induced by several cytokines and substances, including IL-1α [29,30,31,32], IL-1β [30,33,34,35], IL-7 [35,36], IL-17 [35,37,38,39,40,41,42], IL-22 [39,40,41,43], tumor necrosis factor-alpha (TNF-α) [29,30,33,35,44,45,46], histamine [46,47,48,49,50], stromal cell-derived factor-1 (SDF-1, CXCL12) [51,52,53,54,55], lipopolysaccharides (LPSs) [33,35,44,56], reactive oxygen species (ROS) [57,58,59,60], cadmium (Cd) [61,62,63,64], phytohemagglutinin (PHA) [35,56], prostaglandin E2 (PGE2) [65,66,67,68], polyinosinic-polycytidylic acid (poly I:C) [35,69,70], concanavalin A (ConA) [35,71], NaCl [72,73,74,75], thrombin [49,76,77,78], all-trans-retinoic acid (ATRA) [79,80,81], and other cellular stressors (Figure 1).
Additionally, numerous other cytokines and compounds demonstrate the ability to reduce IL-8 levels, such as IL-4 [29,33,35,44,45,82], IL-10 [33,45,73,82,83], IL-35 [84,85,86], transforming growth factor-beta 1 (TGF-β1) [33,87,88], interferon-alpha (IFN-α) [82,89,90], interferon-beta (IFN-β) [82,89,90,91,92], glucocorticoids (GCs) [29,35,44,45,82,93], lipoxins [94,95,96], vitamin D [29,35,44,97,98,99], lipoxygenase (LOX) inhibitors [29,35,100,101], antcin K [102], tannins [103,104], glycyrrhizin (GL) [50,105,106,107] and N-acetylcysteine (NAC) [31,108,109,110] (Figure 2). It is important to note that these are not the only examples of IL-8 modulators, and their impact on IL-8 levels can vary depending on factors such as their concentration, duration of exposure, and the specific cellular context.
IL-8 displays remarkable resilience when exposed to temperature variations and proteolytic enzymes, and it maintains a relative resistance to acidic conditions, making IL-8 an exceptional candidate for deployment at sites experiencing inflammation, where it must endure harsh and adverse surroundings [111,112]. Unlike most inflammatory cytokines, which have a brief lifespan in vivo, IL-8 remains active for days or even weeks after its early production in the inflammatory response [111,112].
IL-8 exerts its effects by binding to specific G protein-coupled receptors known as CXCR1 and CXCR2 [25]. These receptors, belonging to the γ subfamily of G-protein coupled receptors with seven transmembrane domains, play an essential role in mediating IL-8’s effects [113]. Both CXCR1 and CXCR2 interact with IL-8, sharing significant amino acid sequence similarity and exhibiting a binding affinity for IL-8 [114,115,116].
However, these receptors show distinctions in their second extracellular loop, fourth transmembrane domain, C-terminal (intracellular), and N-terminal (extracellular) regions [114,115,116]. Moreover, their desensitization processes differ significantly, with CXCR2 internalizing more rapidly and at lower ligand concentrations compared to CXCR1 [114,116,117]. Additionally, CXCR2 undergoes recycling back to the cell surface at a significantly slower pace than CXCR1 [114,116,117].
Upon binding to CXCR1 and CXCR2, IL-8 induces calcium flows, chemotaxis, and degranulation. However, only CXCR1 is responsible for activating phospholipase D and the stimulation of the superoxide production through the NADPH oxidase enzyme, contributing to the respiratory burst and generation of reactive oxygen species (ROS) in neutrophils [27,114,118,119,120,121,122].

3. IL-8 and CNS

IL-8 plays a crucial role in the peripheral immune response, but it may also exert central effects and be involved in the regulation of neuroendocrine functions related to stress [123]. Glucocorticoids (GCs) have been shown to downregulate IL-8 mRNA expression [124] and decrease IL-8 serum levels [125]. Moreover, the CXCL8 gene features a glucocorticoid receptor binding core site situated at positions -330 to -325, making it susceptible to inhibition by glucocorticoids [123]. Interestingly, IL-8 mRNA expression was detected in the rat paraventricular nucleus of the hypothalamus (PVN), a key site for corticotropin-releasing hormone (CRH) synthesis, and the hippocampus, where negative feedback to CRH production is generated [17,123]. IL-8’s activation of the hypothalamic-pituitary-adrenal (HPA) axis increases cortisol production, which, in turn, helps protect the body from autoimmune disorders by suppressing proinflammatory cytokine production [126].
Additionally, IL-8 possesses the capability to influence glutamatergic synaptic transmission, impacting both presynaptic and postsynaptic processes. Increased IL-8 levels are implicated in notable alterations of synaptic transmission in the prefrontal cortex and may contribute to the development of persistent inflammatory pain [127].
Various cell types within the brain, including astrocytes, neurons, microglia, and endothelial cells, consistently express CXCL8 receptors [128]. The production of IL-8 has been noted upon the stimulation of microglia, resident brain tissue macrophages, and monocyte-derived macrophages (MDMs) [33]. These innate immune cells play crucial roles in the inflammatory response, the phagocytosis of cellular debris, and tissue repair following injury [129]. Furthermore, IL-8 is secreted by astrocytes, which are the most abundant cell types in the brain [33,130,131]. Astrocytes participate in numerous functions within the CNS, encompassing the regulation of glutamate, ion homeostasis (e.g., Ca2+, K+), and water balance, as well as the control of blood–brain barrier permeability, scar formation, tissue repair through angiogenesis and neurogenesis, and the modulation of synaptic activity [132,133]. IL-8 transcription in astrocytes is negatively regulated by β-catenin, and positively regulated by the interaction of T cell factors (TCFs), lymphoid enhancing factor (LEF), and activating transcription factor 2 (ATF2) [131].
IL-8 demonstrates strong trophic properties, guiding the movement and survival of neural stem cells and glial progenitor cells. Additionally, it facilitates glia-neuron communication by modulating neuronal excitability, triggering both excitatory and inhibitory activity [134,135]. The release of IL-8 by glial cells can activate CXCR1 and CXCR2 receptors on cholinergic septal neurons. This activation results in the immediate, direct, and reversible modulation of ion channels, leading to a reduction in Ca2+ currents through G-protein activation. Notably, cholinergic septal neurons, a neuronal type particularly vulnerable in patients with Alzheimer’s disease (AD), may be influenced by IL-8, potentially contributing to the cognitive deficits observed in these individuals [134]. Additionally, elevated levels of IL-8 in both plasma and cerebrospinal fluid (CSF) have been associated with increased CSF p-tau levels. Similarly, higher CSF IL-8 levels correlate with elevated CSF Aβ42 levels, while higher CSF sAβPPβ levels are linked to increased plasma IL-8 concentrations [136].
In the context of drug abuse, particularly Methamphetamine (METH) use, the importance of IL-8 and CXCR1 is exemplified. CXCR1 has been associated with the process of neuronal apoptosis induced by METH. The use of METH is linked to oxidative stress, the apoptosis of dopaminergic neurons, and neuroinflammation related to astrocytes [137]. Exposure to METH upregulates the expression of CXCR1 in neurons and amplifies the expression of IL-8 via the nuclear factor-kappa B (NF-κB) pathway in astrocytes. On the other hand, the suppression of CXCR1 expression using siRNA sequences notably mitigated METH-induced neuronal apoptosis and promoted the neuroprotective effect of astrocytes on neurons [137].
Furthermore, the neuroinflammation mediated by glial cells appears to exert an influence on cognitive aging. In healthy older individuals, there is a positive correlation between plasma IL-8 concentrations and glia-related metabolites, such as the total choline in the anterior cingulate cortex and hippocampal myo-inositol, as observed through proton magnetic resonance spectroscopy (1H-MRS) [138]. Moreover, IL-8 might serve as a significant contributor to neuronal loss in Alzheimer’s disease by influencing the release of neurotoxic substances like matrix metalloproteinases (MMPs), and prompting the expression of proteins associated with neuronal cell death, including MMP-2, MMP-9, cyclin D1, and Bim [139].

4. IL-8 and Brain Barrier Integrity

The blood–brain barrier (BBB) and the blood–cerebrospinal fluid–brain barrier (BCSFB) collectively act as crucial interfaces between the cerebrovascular system and the brain parenchyma, thereby restoring homeostasis and enhancing the physiological environment of the CNS [140,141].
The BBB plays a vital role in safeguarding the brain and is frequently compromised during various diseases. It primarily consists of brain endothelial cells securely sealed by intercellular junctional structures, including tight junctions [140,141,142,143,144,145]. These endothelial cells, along with other components, such as glia (astrocytes, oligodendrocytes, microglia), neurons, pericytes, and the basement membrane (BM), collectively form the neurovascular unit, ensuring the proper physiological functioning of the CNS [141,146].
The inflammatory response involves complex interactions of various cell types and signaling molecules. Consequently, peripheral inflammation can trigger a neuroinflammatory response involving the BBB, neurons, astrocytes, and microglia. Additionally, the brain itself can release pro-inflammatory mediators upon stimulation [5].
Chemokines act as signaling molecules for immune and nerve cells. They can induce neuroinflammation to protect the organism from pathogens, helping with phagocytosis of debris and apoptotic cells, and contributing to tissue repair. On the other hand, the overexpression of chemokines can disrupt the integrity of the brain barrier and allow immune cells to infiltrate the brain [7,147].
IL-8 has been shown to induce the recruitment of neutrophils to the brain and regulate their adhesion to endothelial cells. This process can result in a significant influx of neutrophils into the subarachnoid space [33]. Once neutrophils breach the BBB, IL-8 induces their degranulation, leading to the release of chemoattractants for T lymphocytes and priming neutrophils for superoxide production, among other potentially neurotoxic molecules [33,148]. Indeed, patients who have had an ischemic stroke have demonstrated an increase in IL-8 plasma levels [149]. In addition, a correlation was observed between the size of the lesion in acute ischemic stroke patients and the levels of IL-8 in the serum [150]. Moreover, the serum level of IL-8 exhibited a positive correlation with the severity of disability in patients who have had an acute ischemic stroke within the initial 48 h post-stroke, as evaluated using the National Institute of Health Stroke Scale (NIHSS) [151].
IL-8 mRNA has been identified within the choroid plexus (CP) [123], an extensively vascularized tissue residing in the brain’s ventricular system [152]. The CP mainly comprises capillary beds, the pia mater, and numerous epithelial cells resting on a basal lamina. Positioned at the interface between the blood and the CSF, the CP plays an essential role in the production of CSF and the formation of the BCSFB [153,154,155,156].
Moreover, the CP is a significant source of biologically active molecules involved in brain development, stem cell proliferation, differentiation, and brain repair [157]. The CP serves as a gateway for the trafficking of immune cells into the CSF and maintains continuous immune surveillance via CD4+ T cells, macrophages, and dendritic cells. It also regulates immune cell trafficking in response to diseases and trauma [153]. Additionally, the CP synthesizes various growth factors, including insulin-like, fibroblast, and platelet-derived growth factors [155].
Inflammation leads to substantial modifications in both the BBB and the BCSFB, causing the disruption of tight junctions (TJs) and the impairment of barrier functionality [158]. TJs are comprised of various proteins, including transmembrane proteins like Occludin, the Claudin family, and the peripheral membrane-associated Zonula occludens (ZOs) family [159,160]. IL-8 has been shown to down-regulate the mRNA expression of Occludin, Claudin-5, and ZO-1. The expression levels of these proteins decrease with higher concentrations and longer durations of IL-8 exposure, displaying a dose- and time-dependent relationship [159].
The CP is implicated in various neurological disorders, including inflammatory, infectious, traumatic, neoplastic, and systemic diseases, as well as autoimmune diseases [161,162]. Additionally, an increased CP volume has been observed in neurodegenerative disorders [163] and psychiatric conditions [164,165].

5. IL-8 and CSF

The primary role of CP epithelial cells is to secrete CSF into the brain’s ventricles, and the formation of CSF in the CP depends primarily on the transport of Na+, K+, Cl, HCO3, and H2O [162]. The CP mainly contributes to CSF production by allowing free access to the blood compartment through leaky vessels [161]. CSF serves a multitude of functions, including providing mechanical support, acting as a conduit for certain nutrients, eliminating metabolic by-products and waste generated by synaptic activity, and participating in hormonal signaling processes [155,166]. It represents a rich reservoir of various components, including proteins, lipids, hormones, cholesterol, glucose, microRNAs, and numerous other molecules and metabolites, all of which play pivotal roles in modulating a wide spectrum of CNS functions [167].
The identification of the cytokine and chemokine biomarkers within the CSF that correlate with different neuroinflammatory conditions has the potential to serve as a diagnostic tool and offer novel insights into the pathogenesis of these diseases [168,169].
Interestingly, CSF IL-8 levels were found to be elevated in cases of coronavirus disease 2019 (COVID-19) [170,171]. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) can affect multiple organs, including the brain, leading to neuropsychiatric symptoms and cognitive impairments in patients with COVID-19 [172,173]. It is important to note that SARS-CoV-2 can invade the host’s brain tissue through the olfactory tracts, resulting in anosmia or ageusia [172]. In cases of encephalopathy related to SARS-CoV-2, inflammation within the CNS, induced by IL-8, follows the systemic inflammatory cascade. This inflammation may persist and intensify even after immunotherapy [174]. Moreover, the prolonged production of IL-8 could be associated with extended neurological complications in SARS-CoV-2 infections [174].
Furthermore, elevated CSF levels of IL-8 were observed in individuals with schizophrenia [175], schizophrenia spectrum disorders [176,177], bipolar disorder (BD) [178], major depressive disorder (MDD) [175], in adult patients with autism spectrum disorder (ASD) [179], and patients with Parkinson’s disease and dementia (PDD) [180].
In addition, increased levels of CSF IL-8 were observed in patients with Multiple Sclerosis (MS) [181]. Notably, the time between the first anamnestic episode of focal neurological dysfunction and the diagnosis of relapsing-remitting MS was shown to be a key factor linked to an increase in CSF IL-8 levels. Moreover, a higher risk of disease reactivation, an inadequate response to treatments, and clinical disability were observed in correlation with increased CSF IL-8 levels [182].
Conversely, CSF IL-8 levels were found to be significantly lower in individuals who had attempted suicide [183,184], and were also negatively correlated with symptoms of anxiety in suicide attempters [184].

6. CXCL8 Gene and SNPs

The CXCL8 gene is located on chromosome 4q13-21 and comprises four exons and three introns, featuring a unique CAT- and TATA-like structure [113,185,186]. The proximal segment of the CXCL8 promoter includes around 200 nucleotides within the 5′-flanking region of the CXCL8 gene, playing a significant role in regulating the transcription of this gene [187]. Notably, the 5′-flanking region of the IL-8 gene displays distinct sequence dissimilarity compared to other cytokine and acute phase reactant genes [113].
In resting cells, CXCL8 is present in extremely low levels, making it difficult to detect [116,188]. However, the expression of CXCL8 is induced by a range of factors and stressors, with transcription factors NF-κB and activator protein-1 (AP-1) playing key roles in mediating this response. This induction results in a significant increase in CXCL8 expression, typically ranging from 10 to 100-fold [116].
Moreover, genetic polymorphisms, seen in any population, can influence CXCL8 gene expression and may differ between individuals [189,190,191]. For example, the SNP rs4073 (−251A/T), which is located in the promoter region of the CXCL8 gene, influences the transcriptional activity of the CXCL8 gene and has been linked to various diseases [190,191,192]. The A allele of rs4073 is associated with an increased expression of the IL-8 gene and increased IL-8 production. The A/A genotype exhibits the highest values, while the T/T genotype shows the lowest values [191,192,193].
Furthermore, the complex interplay between different SNPs influences CXCL8 gene expression. For instance, in their study, Benakanakere et al., (2016) investigated the effects of stimulating HEK293T cells carrying different genotypes (rs4073 AT, rs2227307 TT, and rs2227306 TC/CC) at the IL-8 locus with the TLR3 agonist poly I:C. They observed that cells with the ATC/TTC haplotype significantly upregulated IL-8 gene expression at both transcriptional and translational levels, leading to enhanced neutrophil transmigration [189].
Table 1 presents four single nucleotide polymorphisms (SNPs) in the Human CXCL8 gene which were reviewed using the SNP database of the National Library of Medicine “https://www.ncbi.nlm.nih.gov/snp/ (accessed on 12 January 2024)”.
CXCL8 gene polymorphisms demonstrate differences in the disease susceptibility between individuals carrying different alleles. For instance, Kang et al., (2015) observed higher frequencies of CXCL8 −251T/A alleles in patients with Alzheimer’s disease (AD), compared to those without AD. However, the significance of these associations was lost after Bonferroni correction [194]. Interestingly, other studies did not find any significant association between patients with AD and the rs4073 polymorphism [195,196,197]. Yet, Infante et al., (2004) discovered a synergistic effect between the rs4073 TT genotype and the IL-1A -889 T allele in patients with AD. Individuals carrying both polymorphisms had about twice the risk of developing AD compared to subjects without these risk genotypes, suggesting a gene–gene interaction [195]. Nonetheless, another study found no interactive effect between the rs4073 and IL-1α -889C/T (rs1800587) polymorphisms in patients with AD [197]. Furthermore, meta-analyses of the rs4073 polymorphism suggested a possible predisposition to AD in individuals of Asian ethnicity [198,199].
In a study by Kamali-Sarvestani et al., (2006), it was noted that there was a significant increase in the presence of the rs4073 TT genotype among patients with Multiple Sclerosis (MS) compared to a group of healthy individuals [200]. Moreover, Dolcetti et al., (2023) discovered a significantly higher concentration of CSF IL-8 in patients with relapsing-remitting MS carrying the rs2227306 T allele and CT/TT genotypes. The study also observed a significant inverse relationship between CSF IL-8 levels and cortical thickness (CT) in individuals carrying the T allele at rs2227306, as assessed through structural MRI measures [201]. In addition, Dolcetti et al., (2023) established a significant positive association between CSF IL-8 levels and patients with a clinical disability in rs2227306 CT/TT assessed with the Expanded Disability Status Scale (EDSS) [201]. Furthermore, the rs2227306 polymorphism was found to be associated with BD I and methamphetamine addiction, but not with BD II, ADHD, MDD, or schizophrenia [202].
Variations in the CXCL8 gene and the resulting changes in IL-8 production could potentially lead to shifts in personality traits that may be associated with an increased risk of suicidal behavior [203]. In their study, Noroozi et al., (2018) determined that the presence of the rs4073 T allele was notably higher in the group of individuals who attempted suicide, in comparison to both the control group and those who completed suicide. Moreover, the haplotype rs4073T/rs2227306C/rs1126647A was significantly less prevalent in the completed suicide group compared to the suicide attempt group. Additionally, the rs4073A/rs2227306T/rs1126647A haplotype was significantly more prevalent in individuals who utilized “hard” suicide methods compared with those who attempted “soft” suicide methods [203].
Janelidze et al., (2015) found that anxiety symptoms were more severe in suicide attempters carrying the rs4073T, and the T allele was more common among females who attempted suicide than in a population-based cohort. Notably, those who attempted suicide and also had rs4073 AA demonstrated a lower median Brief Scale for Anxiety (BSA) score when compared to rs4073 AT and rs4073 TT carriers [184].
Furthermore, Ben Afia et al., (2020) found that the rs1126647 polymorphism within the Tunisian population showed a significant risk for schizophrenia. Notably, females displayed a significant association between the rs1126647 T allele and the T/T genotype, correlating with an elevated risk of paranoid schizophrenia. In males, this predisposition was observed specifically in those carrying the rs1126647 T/T genotype [186]. Moreover, the presence of the rs1126647 T allele and T/T genotype in paranoid schizophrenia was significantly associated with an adult-onset age of 24 years and older. Interestingly, the haplotypes TTT, ACT, and TCT at rs4073/rs2227306/rs1126647, each incorporating the risk allele rs1126647T, were associated with an increased risk for paranoid schizophrenia. However, only the combination of TCT was seen as a risk factor for schizophrenia more generally [186].
In other research, Reyes-Gibby et al., (2013) discovered a notable association between the rs4073 polymorphism and depressed mood, pain, and fatigue in patients with non-small cell lung cancer (NSCLC) in advanced stages (IIIB-IV) of the disease, assessed before cancer treatment. Importantly, individuals carrying rs4073 T/T genotypes were more likely to experience severe depression compared to those with A/T and A/A genotypes. Interestingly, similar rs4073 T/T genotypes among these patients were associated with less severe pain and fatigue compared to carriers of the A/T and A/A genotypes [204].
Additionally, Kim et al., (2013) observed a gene–environment interaction related to the incidence of late-life depression. They identified an association between declining physical health and depression, which was strongest in individuals genetically predisposed to a cytokine-mediated inflammatory response. Notably, they found that rs4073A had a significant modifying effect on the association between physical disorders and the incidence of depression over two years in a Korean population of adults aged 65 and over [205]. However, other studies, including those regarding patients with post-stroke depression [206], depression in breast cancer patients [207], and depression in an elderly Korean population [194], did not find significant associations with the rs4073 polymorphism.

7. Maternal IL-8 during Pregnancy and Implications for Offspring

Chemokines can cross the placental and brain barriers, regulating the communication between neurons and microglia in the CNS [208]. During healthy gestation, IL-8 undergoes tight regulation and reaches higher peripheral concentrations during preterm compared to term labor [209]. Additionally, there is a decrease in IL-8 levels from early to later pregnancy, followed by an increase at postpartum [210]. The natural process of childbirth leads to increased circulating IL-8 levels, accompanied by rises in the numbers of neutrophils and monocytes [1]. This elevated post-birth IL-8 level may result from spillage originating in activated vascular endothelial cells or circulating immune cells [1].
Nelson et al., (2006) reported that IL-8 concentrations in newborn infants, both those at preterm and term, surpassed those found in adults [211]. Moreover, Yektaei-Karin et al., (2007) found that the transmigration of IL-8-induced neutrophils in newborns following normal delivery was significantly higher in cord blood compared to neutrophils from Cesarean section births or adult peripheral blood [1].
IL-8 exhibits dual pro-inflammatory and anti-inflammatory roles based on its concentration, suggesting that higher and lower levels of IL-8 might exert opposing effects [212]. This duality implies that IL-8 could play both damaging and defensive roles in the potential demyelination process [212].
Changes in inflammatory cytokines, influenced by environmental factors affecting both maternal and fetal immune systems, can profoundly impact fetal brain development [213]. The exposure of the developing fetus to elevated levels of maternal cytokines has been associated with structural changes in neuroanatomy [214]. Moreover, elevated levels of IL-8 can have significant adverse effects on the developing brain, influencing both its structure and function. Dysregulated IL-8 appears to play a critical role in connecting perinatal systemic inflammation with atypical white matter development in infants born prematurely [215].
Furthermore, increased IL-8 levels in blood samples taken from the umbilical cord or in the infant shortly after birth are strongly associated with visible white matter injury, cerebral palsy, neurodevelopmental challenges, and cognitive impairments in children born prematurely [215]. Remarkably, even among children born at full term, IL-8 stands out as one of the neonatal cytokines most strongly linked to later diagnoses of autism [215]. Interestingly, Jones et al., (2017) found that mothers with higher mid-gestational IL-8 serum levels had a higher risk of having children with autism spectrum disorders with intellectual disabilities. However, mothers of children with developmental delays or autism spectrum disorders without intellectual disabilities had lower mid-gestational IL-8 levels compared to the general population [216].
The increased secretion of IL-8 in Down syndrome may contribute to the observed reduction in postnatal brain growth seen in individuals with this condition. In infants with Down syndrome, IL-8 levels exceed those observed in both subjects with autism and neurotypical control subjects [211]. Additionally, IL-8 appears to amplify the effect of amyloid beta peptide in stimulating the production of IL-6, IL-1β, TNF-α, and COX-2 in cultured human microglia [211,217], suggesting a potential role in the early development of Alzheimer’s neuropathology in Down syndrome [211].
During the second trimester, mothers of offspring with schizophrenia spectrum disorders displayed significantly higher IL-8 levels compared to mothers in the control group [218]. Moreover, within the group of individuals with schizophrenia, the exposure of the fetus to increased maternal IL-8 levels during the second and third trimesters correlated significantly with enlarged ventricular CSF volumes [214]. Additionally, Ellman et al., (2010) reported significant associations between maternal IL-8 levels and reductions in the volumes of the left entorhinal cortex and right posterior cingulate among individuals with schizophrenia. They also observed volumetric reductions that approached significance in the right caudate, bilateral putamen, and the right superior temporal gyrus [214]. Furthermore, Osborne et al., (2022) found notably elevated IL-8 levels, measured in the early third trimester in pregnant women with a previous history of MDD but without depression symptoms during pregnancy, in comparison to pregnant women without an MDD history [219].
Elevated levels of maternal IL-8 in the first trimester were significantly linked to externalizing symptoms (e.g., aggression, impulsivity) and subsequent conduct problems in the offspring [220]. Given that externalizing symptoms in children are tied to cognitive difficulties, it is possible that fetal exposure to IL-8 increases the risk of developing externalizing symptoms through cognitive impairment [220]. Moreover, higher IL-8 levels during early pregnancy (10–18 weeks) were linked to notable decreases in fine motor skills and problem-solving abilities in children at age two [208].
However, prenatal exposure to IL-8 has also been shown to have opposite effects on neurodevelopmental processes. For instance, maternal IL-8 levels during gestation exhibited a positive correlation with verbal abilities while displaying a negative correlation with a child’s spatial abilities [213]. Moreover, reduced IL-8 levels throughout the second and third trimesters were associated with poorer neurocognitive functioning at age seven [221]. Children exposed to lower prenatal IL-8 levels exhibited lower scores in cognitive performance and motor function. Conversely, higher gestational IL-8 levels were associated with improved performance in the Drawing Task and the Tactile Finger Recognition Task [221].
Interestingly, mothers of a higher socioeconomic status (SES) demonstrated higher IL-8 concentrations, while mothers from lower SES families had lower IL-8 levels. Moreover, the decrease in IL-8 levels during pregnancy was associated with compromised child self-regulation [222]. Additionally, increased maternal socioeconomic disadvantage corresponded to noticeably lower IL-8 concentrations during the third trimester, along with a lower ratio of IL-8 to the anti-inflammatory cytokine IL-10. These associations remained unaffected by maternal medical conditions known to disrupt immune responses [223]. Furthermore, lower maternal serum IL-8 concentrations were linked to the presence of neurologic abnormalities in offspring during early life (at ages of 4 months and 1 year) [223].

8. IL-8 in Depressive and Bipolar Disorders

Both Bipolar Disorder (BD) and Major Depressive Disorder (MDD) are associated with the activation of the immune-inflammatory response system and the compensatory immune-regulatory system [224,225,226]. BD has been linked to an imbalance in the immune system and low-grade inflammation [227]. This suggests that alterations in IL-8 levels could potentially play a significant role in the pathophysiology of BD.
Tang et al., (2021) established a correlation between increased serum IL-8 levels and impaired functional connectivity (FC) in the right precentral gyrus in unmedicated patients with BD II depression using resting-state functional magnetic resonance imaging (rs-fMRI). This finding suggests that inflammation may contribute to brain functional abnormalities in BD [228]. Moreover, Isgren et al., (2015) found higher CSF IL-8 concentrations in patients with euthymic BD compared to control subjects. Additionally, patients taking lithium or/and antipsychotic medications had even higher IL-8 levels compared to those not taking these medications. Furthermore, IL-8 levels were found to increase with age and with a higher CSF/serum albumin ratio [178]. Notably, in a prospective study, Isgren et al., (2017) did not find an association between CSF IL-8 baseline concentrations and clinical outcomes in patients with BD followed for 6–7 years [229].
The specific pattern of IL-8 alterations appears to be complex. Wang et al., (2016) reported higher IL-8 levels in patients with BD I compared to patients with BD II and other specified BDs with short-duration hypomanic episodes (2–3 days) [230]. In another study, elevated levels of IL-8 in peripheral blood were found only during the depressive phase of BD [231]. Interestingly, in the same study, an association was established between lower blood IL-8 levels and a longer illness duration in BD [231].
Despite the association of both BD and MDD with immune system imbalance, IL-8 levels demonstrate different patterns. For instance, serum IL-8 levels were exclusively elevated in BD patients, but not in MDD patients [232]. Additionally, plasma IL-8 levels were directly associated with bipolar depression when compared to MDD [224]. This suggests that IL-8 may serve as a potential biomarker to differentiate between MDD and BD, especially in cases of atypical clinical presentations [232]. Moreover, IL-8 testing may even hold prognostic value in identifying resilience or a risk of depression [233].
Despite the above-described evidence, inconsistencies regarding IL-8 levels in BD exist. For instance, Barbosa et al., (2013) reported decreased plasma levels of IL-8 in patients with BD I compared to controls [234]. Conversely, some studies found no significant differences in IL-8 levels between patients with BD patients and controls [235,236].
Research indicates that immune dysregulation plays a significant role in depression [237], and numerous studies have evaluated the correlation between IL-8 levels and disease progression as well as treatment response. For instance, higher baseline plasma levels of IL-8 in breast cancer survivors were linked to a reduced risk of incident and recurring major depression [233]. Moreover, elevated IL-8 plasma levels demonstrated an association with a lower severity of depressive symptoms in depressed patients, while treatment-induced increases in IL-8 predicted a positive treatment response [238]. Conversely, a decline in serum IL-8 levels was associated with depression [239]. Notably, patients with MDD who positively responded to antidepressant treatment exhibited lower baseline IL-8 levels than those who did not respond [240].
In a study by Zou et al., (2018), patients with MDD showed significantly lower serum levels of IL-8 compared to controls, revealing linear correlations between IL-8 and the severity of depression [241]. They also observed significant linear correlations between IL-8 levels and anxiety levels in patients with comorbid anxiety disorders. Thus, higher IL-8 levels were associated with lower scores on the Hamilton Depression Rating Scale (HAM-D) and the Hamilton Anxiety Rating Scale (HAM-A) [241].
Furthermore, a higher baseline level of IL-8 in plasma was correlated with less pronounced increases in depressed mood and feelings of social disconnection [238]. Interestingly, Cai et al., (2023) suggested that elevated serum IL-8 levels might correspond to improvements in delayed memory and visuospatial/constructional function in patients with MDD [242]. However, it is important to note that this positive association was not universally observed. Baune et al., (2008) found that higher IL-8 levels in healthy elderly individuals were associated with poorer performance on specific neuropsychological tests related to memory, processing speed, and motor function. Notably, IL-8 levels in these individuals were not associated with general cognitive function as assessed by the Mini-Mental State Examination (MMSE) [243].
Intriguingly, the concentrations of IL-8 appear to be influenced by several factors, including sex differences, which were reported in various studies exploring IL-8 findings. For example, an elevated plasma concentration of IL-8 was inversely related to the HAM-D score in females, but this relationship was not detected in males [244]. Additionally, Moriarity et al., (2019) found a correlation between higher initial plasma IL-8 levels and a reduction in depressive symptoms at a follow-up 31 months later, specifically in adolescent males [245].
Moreover, treatment response may also exhibit sex-specificity. For instance, baseline plasma levels of IL-8 and changes in IL-8 related to treatment with electroconvulsive therapy (ECT) were associated with improvements in depression in females, but not males [246]. Similarly, lower baseline plasma levels of IL-8 and subsequent increases in IL-8 were specifically correlated with improved depression in females treated with ketamine [247]. These findings highlight the need for personalized treatment approaches based on individual IL-8 profiles and possible gender-specific responses.
Interestingly, physical activity has also been shown to influence IL-8 levels. For instance, in patients with MDD, serum concentrations of IL-8 significantly increased following vigorous exercise, while no changes occurred after light and moderate exercise. Importantly, depression severity did not seem to impact the acute inflammatory response to exercise [248].
It is noteworthy that research on IL-8 remains multifaceted and ongoing, and there are conflicting findings regarding IL-8 levels in depression. Several studies have reported significantly higher serum IL-8 levels in patients with MDD compared to controls [212,249,250]. Another study found no association between plasma IL-8 levels and depressive symptom severity in physically active individuals after SARS-CoV-2 infection and a follow-up after 3 months [251]. In contrast, Ogłodek (2022) reported an increase in IL-8 serum concentration with depression severity. Notably, the highest increase in IL-8 levels was seen in a group of patients with severe depression that co-occurred with Post-traumatic Stress Disorders (PTSDs) [252]. Additionally, Suneson et al., (2023) found significantly higher plasma IL-8 levels in patients with difficult-to-treat depression compared to controls [253]. Importantly, Szałach et al., (2022) identified that serum IL-8 values exceeding 19.55 pg/mL were associated with a 10.26 likelihood ratio of developing treatment-resistant depression [254].

9. IL-8 and BDNF

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, plays a significant role in neuronal survival and differentiation during development and maintains high expression levels in the adult brain [255,256]. BDNF participates in many functions, including neuronal migration, synaptic structure regulation, and neurotransmitter release, all of which are vital for brain circuits regulating memory, learning, emotions, sleep, and appetite [257]. Moreover, BDNF regulates synaptic transmission and activity-dependent plasticity and is predominantly found in various regions of the brain, including the hippocampus, amygdala, cerebellum, and cerebral cortex, with hippocampal neurons exhibiting the highest BDNF levels [256]. Therefore, given the wide array of functions attributed to BDNF, it is not surprising that it is associated with a multitude of neuropsychiatric conditions [258,259,260].
In neonates without brain injury born between 23- and 41-weeks of gestation, there is a significant gestation-dependent increase in serum BDNF levels (3.7%) and IL-8 levels (8.9%) for every week of gestation [261]. However, BDNF concentrations do not show any association with the first 7 days of life (DOL), while IL-8 levels increase with each DOL by 18.9% [261]. Additionally, BDNF concentrations exhibit a remarkable increase with age, with a progressive and significant ascent from an average of 740 pg/mL in very preterm control infants to nearly 5500 pg/mL in adults [211].
Interestingly, BDNF can directly contribute to anti-inflammatory effects on microglia by regulating cytokine responses, which in turn can impact neurons. BDNF’s priming of microglia may reprogram the inflammatory state, leading to alterations in neuron–microglia interactions [262]. It is worth noting that both human fetal and adult microglial cells produce IL-8 in response to lipopolysaccharide (LPS), along with two key cytokines involved in initiating an inflammatory response, namely IL-1β and TNF-α [33].
Newborns diagnosed with neonatal encephalopathy exhibited increased plasma levels of IL-8 and decreased BDNF levels, in comparison to healthy newborns. Notably, there was an inverse correlation between BDNF levels and the encephalopathy grade, while IL-8 levels were inversely linked to motor outcomes [263]. Additionally, research has found that patients with schizophrenia have significantly lower BDNF levels and higher IL-8 serum levels compared to individuals without schizophrenia [264,265]. Interestingly, a positive correlation was observed between BDNF and IL-8 levels [264,265]. This could suggest that a relative increase in BDNF levels possibly acts as a compensatory response in patients with schizophrenia, although it might not be sufficient to counteract the inflammatory damage caused by increased cytokines like IL-8 [264,265]. Notably, Xiu et al., (2019) discovered a negative correlation between reduced serum BDNF levels and executive function in patients with chronic schizophrenia. They observed an interaction between low BDNF and high IL-8 concentrations, which were positively correlated with executive dysfunction as measured by verbal fluency tests (VFT) and Wisconsin card sorting tests (WCST) in patients with schizophrenia [265].
Moreover, Wang et al., (2016) noted elevated IL-8 plasma levels in individuals with BD, while BDNF levels did not exhibit significant differences compared to a control group [230]. In contrast, another study found no significant difference in IL-8 plasma levels between subthreshold bipolar disorder (SBD) and BD-II at baseline and following a 12-week mood stabilization treatment. However, in the SBD group, the study revealed markedly lower baseline BDNF plasma levels which remained low even after 12 weeks of treatment, despite similar treatment responses between the two groups [266].
Furthermore, Liou et al., (2023) reported a significant difference in IL-8 and BDNF plasma levels between patients with BD and those in a control group. Moreover, patients with BD and comorbid alcohol use disorder (AUD) displayed higher IL-8 levels compared to patients with BD without AUD [267]. Additionally, lower BDNF levels were associated with decreased performance on cognitive assessments, while plasma IL-8 levels in patients with BD demonstrated a significant negative correlation with the number of completed categories in the Wisconsin Card Sorting Test (WCST) [267].

10. IL-8, ROS, and Oxidative Stress

ROS act as signal transduction molecules activating IL-8 and significantly regulating the production of this cytokine [58,268]. While ROS play a crucial role as cellular signaling molecules for normal biological processes, their excessive production can induce harm to various cellular components and functions, potentially disrupting physiological equilibrium [269,270,271].
The etiology of various diseases has been linked to an imbalance between ROS production and the protective antioxidant defenses of cells, particularly in conditions involving inflammation or ischemia-reperfusion, where excessive ROS generation occurs [121,269,272]. In the brain, ROS can trigger cellular damage contributing to cognitive dysfunction and the development of neuropsychiatric conditions [273,274,275].
Oxidative stress occurs when the production of ROS and other oxidants surpasses the body’s antioxidant defenses and ability to neutralize them [276,277]. This is particularly concerning for brain neurons, which rely heavily on oxidative phosphorylation for energy and are vulnerable to oxidative stress [278]. Indeed, even minor disturbances in the redox equilibrium during neural development, when combined with genetic or environmental susceptibilities, can significantly impact neurogenesis, neuronal differentiation, and neural connectivity [276]. ROS generate free radical oxidation products that can interact with cellular metabolites, potentially causing cell death through apoptosis or necrosis [279]. One significant consequence of ROS overproduction and oxidative damage is DNA alteration, potentially leading to permanent mutations and other genomic instabilities [278]. It is important to note that ROS are generated by various sources and are mostly produced in mitochondria as byproducts of cellular metabolism [280,281].
The role of IL-8, ROS, and oxidative stress becomes even more critical when considering the impact of viral infections such as COVID-19. The inflammatory response triggered by SARS-CoV-2 leads to the release of cytokines, chemokines, and ROS, causing the disruption of TJs and compromising the brain barrier’s integrity [282]. Furthermore, coronavirus infection disrupts mitochondrial regulation, leading to a reduction in Adenosine Triphosphate (ATP) synthesis and the activation of NADPH oxidase, thereby contributing to ROS production [283]. Moreover, COVID-19 affects the morphological features and distribution of astrocyte and microglia cells [284]. Notably, the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), is expressed by astrocytes and microglia [285].
Interestingly, Clough et al., (2021) observed an increase in IL-8 gene expression in microglial cells treated with both the SARS-CoV-2 spike protein and heat-inactivated SARS-CoV-2, compared to untreated controls. Moreover, higher IL-8 cytokine expression was seen in microglia treated with heat-inactivated SARS-CoV-2 compared to those treated with the SARS-CoV-2 spike protein [286]. Activated microglia and astrocytes can release ROS and cytokines, contributing to sustained neuroinflammatory responses and neuronal damage [282,287,288].
Furthermore, when stimulated by IL-8, neutrophils migrate to the inflammation site, degranulate and release lysosomal enzymes that not only damage pathogens but also inadvertently harm nearby tissue by oxidizing cellular components like lipid membranes, proteins, and DNA [289]. Furthermore, activated neutrophils contribute to oxidative stress through the production of ROS via NADPH oxidase during the respiratory burst [290]. Prolonged oxidative stress can facilitate the development and persistence of inflammation by activating transcription factors that modify the expression of various genes and proteins, including pro-inflammatory cytokines [291]. Consequently, in severe cases, excessive ROS production by deregulated neutrophils can escalate the local inflammatory response to a systemic level [292].
The oxidative stress and cytokine storm create a vicious cycle, escalating inflammation and ultimately contributing to multi-organ failure in severe COVID-19 cases [286]. Neurons, being particularly vulnerable to such inflammatory and oxidative damage, are increasingly implicated in the development of Long COVID, a post-infection condition characterized by lingering neurological and psychiatric symptoms [283]. Patients with Long COVID often report fatigue, cognitive impairments, sensory dysfunctions, headaches, post-exertional malaise, and experience mood disorders. These symptoms can persist for months or even years, affecting memory, language, processing speed, and executive function [283,293,294,295,296,297,298,299].
Importantly, individuals with pre-existing health comorbidities or neuropsychiatric vulnerabilities are at an increased risk of developing severe COVID-19 and long-term post-COVID neurological and psychiatric consequences [300]. This could be partly explained by the oxidative stress and elevated levels of IL-8 which are found in patients with neuropsychiatric conditions. For example, Wu et al., (2021) observed decreased activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx), along with elevated levels of malondialdehyde (MDA) and IL-8 in individuals with chronic schizophrenia. Moreover, the combined effects of IL-8 with MDA or SOD were found to correlate with executive function in patients with chronic schizophrenia [301]. Similarly, Wei et al., (2020) discovered a connection between oxidative stress parameters and serum BDNF levels in individuals with chronic schizophrenia, revealing lower GPx and SOD activities, reduced BDNF levels, and elevated MDA levels compared to those of controls [276]. Enzymatic antioxidants such as SOD, catalase (CAT), GPx, and non-enzymatic antioxidants like glutathione (GSH) serve as a defense mechanism, reducing ROS activity and maintaining cellular redox balance [281,302,303]. Additionally, MDA, a lipid peroxidation by-product, is commonly used as a marker of oxidative stress [304,305,306].
It was shown that the production and concentration of IL-8 can be suppressed by antioxidants, notably N-acetylcysteine (NAC) [31,57,58,108,109,110,307,308]. NAC possesses both anti-inflammatory and antioxidant properties, facilitating the replenishment of glutathione and the scavenging of free radicals [309,310,311,312]. Glutathione is essential for intracellular and intercellular signaling in the brain, and its depletion contributes to oxidative stress, leading to neuronal metabolic disturbances and consequential alterations in synaptic signaling [313,314,315,316,317,318,319,320]. Adjunctive therapy with NAC has demonstrated positive outcomes in various neuropsychiatric conditions [321,322,323,324,325,326,327,328,329,330].

11. Discussion

Many factors, including genetic predisposition, environmental conditions, and immune responses, contribute to neuropsychiatric conditions. The host’s immune system significantly modulates both physiological and pathological processes, and inflammatory immune responses have been linked to disease severity and treatment efficacy [331,332]. Peripheral immune and inflammatory cells can migrate to the brain through the compromised BBB, and proliferate at sites of inflammation, directly exacerbating the inflammatory response or amplifying it via glial and neuronal activation [333].
Chemokines play a crucial role in the interaction between the immune and nervous systems, being involved in various brain-related processes such as neurodevelopment, neurogenesis, neuromodulation, synaptic transmission, neuroendocrine homeostasis, brain barrier integrity, neuroinflammation, and stress response [19,20,334,335,336].
IL-8 belongs to the CXC chemokine subfamily and is produced early in the inflammatory response. It can remain active for a prolonged period, making it particularly relevant to the long-term inflammatory alterations observed in neuropsychiatric conditions [337,338,339]. IL-8 is secreted by various cell types and is released when exposed to an inflammatory trigger. Furthermore, numerous cell types possess receptors for IL-8, and upon binding, they generate molecules with both local and systemic activity [35,340].
The variety of cellular origins for IL-8 highlights the pleiotropic nature of its functions. While IL-8 is essential for the host’s defense mechanism due to its influencing neutrophil activation and trafficking, its prolonged presence in the bloodstream during inflammatory conditions can result in varying degrees of tissue damage, and contribute to disease-related processes, such as fibrosis, angiogenesis, and tumorigenesis [9,340].
Considering the various roles of IL-8, it is significant that psychological stress can cause an increase in IL-8 secretion, which could potentially worsen disease processes [341]. Experiencing chronic stress during early life can lead to long-lasting immune, endocrine, neural, and inflammatory alterations, contributing to a broad range of neuropsychiatric conditions later in life [342].
The production of IL-8 has been linked to depression, negative affect traits, and perceived stress [289]. Interestingly, individuals with strong social support and larger social networks have lower IL-8 plasma levels, particularly among healthy midlife adults [289]. Additionally, a positive association has been found between IL-8 levels and fewer years of education, indicating a potential link with lower socioeconomic statuses [289]. Suarez et al., (2004) found that both the severity of depressive symptoms, measured by the 21-item Beck Depression Inventory (BDI), and hostility, assessed using the 50-item Cook–Medley Hostility (Ho) scale, independently and synergistically increase IL-8 expression in healthy premenopausal women [343].
Stressful work conditions elevate urinary IL-8, which indicates its potential use as a reliable biomarker of stress [344,345]. For example, Dutheil et al., (2013) reported that emergency physicians on 24 h shifts showed a nearly doubled level of urinary IL-8 compared to controls or those on 14 h shifts. This prolonged immune response persisted for at least three days after the 24 h shifts, despite a day of rest following the 24 h shifts. Notably, older age, stressful work conditions, and long shifts were associated with increased IL-8 levels [344]. Additionally, IL-8 levels correlated positively with exposure to life-and-death emergencies and were exacerbated by sleep deprivation and poor sleep quality during shifts [344]. Furthermore, Fukuda et al., (2008) observed that hospital acute care department nurses with higher professional stress had higher urinary IL-8 levels compared to chronic care departmental nurses [345].
Prolonged and intense exposure to stress can disrupt the body’s energy balance and reduce adaptation mechanisms, resulting in adverse effects on overall well-being [346]. Psychological stress is associated with an elevated production of oxidants, exacerbating oxidative stress within the body [347]. This oxidative burden is particularly pronounced in individuals experiencing chronic stress, as the continuous activation of the HPA axis perpetuates oxidative damage over time [348]. Moreover, Kim et al., (2021) reported that acute psychosocial stress can induce a noticeable elevation in oxidative stress within less than 2 h [349]. Such oxidative stress arises from an imbalance between ROS production and antioxidant defenses and is often exacerbated by various cellular stressors that elevate ROS levels or reduce the body’s ability to neutralize them [350,351].
Given the brain’s high metabolic activity and oxygen consumption, it is especially vulnerable to oxidative stress, which can significantly impair CNS functions and contribute to neuroinflammation and neurodegeneration [352,353,354]. Moreover, the interplay between oxidative stress and inflammatory responses adds complexity to stress-induced pathophysiological alterations. Indeed, oxidative stress and neuroinflammation have the potential to mutually reinforce each other [355]. ROS accumulation within cells can disrupt intracellular signaling, leading to the dysregulation of inflammatory processes [353]. In addition, elevated ROS levels upregulate the production of IL-8 by promoting NF-κB signaling [356]. Thus, ROS and oxidative stress contribute to IL-8 production [60,357,358]. IL-8 exhibits remarkable stability and prolonged biological activity, resisting proteolysis and denaturation, while its mRNA maintains sustained expression in the presence of stimulating agents, underscoring its significant biological impact [290]. Excessive IL-8 correlates with heightened NF-κB translocation and reduced glutathione levels [359]. Additionally, IL-8 facilitates the ROS metabolism and induces ROS production [35,360]. Therefore a potential positive feedback loop may exist between oxidative stress and IL-8, fueling each other and contributing further to cellular damage.
IL-8 plays a crucial role in attracting neutrophils, and a deficiency in CXCL8 can impair the movement of these cells to tissues [361]. For example, children suffering from microcephaly because of Congenital Zika Syndrome showed a significant decrease in serum IL-8 levels, leading to an impairment in leukocyte migration [361]. On the other hand, a rise in the absolute neutrophil count and IL-8 levels in the bloodstream were linked to the severity of COVID-19, wherein plasma IL-8 levels were associated with mortality [362]. Critically ill patients with COVID-19 demonstrated a high neutrophil-to-lymphocyte ratio associated with elevated levels of ROS, which could lead to tissue damage, thrombosis, and the dysfunction of red blood cells, thereby contributing to the severity of COVID-19 [292].
The nervous and immune systems engage in complex communication, which is regulated by multiple mechanisms. This interaction can be disrupted by various triggers such as infections, autoimmune diseases, peripheral and systemic inflammation, traumatic brain injuries, environmental toxins, and stress, potentially leading to neuroinflammation [363,364,365,366,367,368,369,370,371,372,373,374,375].
One important diagnostic tool for measuring this inflammatory response is IL-8. However, the use of this marker needs to be personalized, as diverse populations exhibit variances not only in their susceptibility to and progression of diseases, but also in the levels of inflammatory markers, such as IL-8. For instance, Mayr et al., (2007) reported that healthy young volunteers of African descent had higher average IL-8 levels compared to their Caucasian counterparts [376]. Moreover, plasma IL-8 levels demonstrated an inverse correlation with neutrophil counts in individuals of African descent and in combined groups, but not in those who were Caucasian alone [376]. Additionally, Mayr et al., (2007) reported a lower oxidative burst capacity in stimulated neutrophils among volunteers of African descent [368].
It was also observed among African American women that both stress/distress and poor sleep quality had notable impacts on the production of proinflammatory cytokines during the postpartum period [377]. Specifically, in African American women, but not in White women, evaluations conducted at 7–10 weeks postpartum revealed that poorer sleep quality, heightened parenting stress, increased depressive symptoms, and elevated general perceived stress were all associated with greater LPS-stimulated IL-8 production [377].
The observed variations in IL-8 levels across individuals and different populations can be partially attributed to genetic polymorphisms. The CXCL8 gene, which encodes IL-8, exhibits several functional polymorphisms that may influence IL-8 production [191,378]. For example, rs4073 (−251A/T) has been linked to inflammatory diseases, and the rs4073 A allele has an association with increased IL-8 production [193,379,380]. Additionally, the A/A genotype may reduce the threshold for IL-8 synthesis [380].
Wacharasint et al., (2012) reported that critically ill Caucasian patients with the rs4073 A/A genotype had an increased risk of a PaO(2)/FiO(2) < 200 (the PaO2/FiO2 ratio is the ratio of arterial oxygen partial pressure), and demonstrated greater IL-8 mRNA expression than those with the A/T or T/T genotypes [190]. Additionally, Hildebrand et al., (2007) suggested that rs4073 polymorphism can influence the severity of the inflammatory response following multiple traumas. They found that the rs4073 A/A genotype showed a significantly longer duration of mechanical ventilation after trauma compared to genotype T/T [379]. In contrast, the rs4073 T allele, which is linked to lower IL-8 production, was associated with the severity of microcephaly in children with congenital Zika syndrome [381]. In addition, Zhao et al., (2020) identified the rs4073 T/T genotype as a potential risk factor for sepsis in full-term neonates [382].
Ethnic groups exhibit varying distributions of genetic polymorphisms. For instance, Fujihara et al., (2007) reported that Ovambos and Gambians displayed the lowest rs4073 T allele frequencies at 8% and 10%, respectively, while those who are Japanese had the highest T allele frequency at approximately 80%, with the T/T genotype being 67% [191].
Interestingly, the same genetic polymorphisms can have varying impacts on disease vulnerability across populations. For example, Wang et al., (2013) discovered a significant association between the A/A and A/T genotypes of the rs4073 polymorphism and increased oral cancer risk among Caucasian populations, while no statistically significant association was found among Asian populations [383]. In contrast, Zhang et al., (2019) found a significant association between the rs4073 A allele increasing coronary artery disease (CAD) risk in a Chinese population, but this association was not observed in Caucasians [384]. Zhang et al., (2021) conducted a meta-analysis revealing that CXCL8 rs4073 polymorphisms may affect a predisposition to Alzheimer’s disease in people who are Asian, but not in people who are Caucasian [198]. This suggests that CXCL8 gene polymorphism can have a distinct influence not only on various disease states but also on health outcomes across diverse populations. Such variability indicates the presence of additional factors that may modulate these effects. Indeed, environmental factors can interact with the genome by modifying epigenetic mechanisms that regulate gene expression [385].
Furthermore, it is important to note that there can be an interplay between SNPs of the CXCL8 gene, which may result in a combined effect when inherited together. For instance, the haplotype of CXCL8 rs4073T/rs2227306C/rs1126647T is associated with an increased risk for schizophrenia [186]. Additionally, a significant effect can be seen in gene–gene interactions. For example, Ghazy (2023) discovered a significant correlation between the simultaneous presence of IL-8 rs2227306C and IL-6 rs1800795G alleles in an individual and an increased risk of severe COVID-19 outcomes. Conversely, individuals carrying the IL-8 rs2227306T and IL-6 rs1800795C alleles were found to have a reduced risk of severe COVID-19 [386]. In summary, considering individual and ethnic differences is crucial when interpreting inflammatory markers for an accurate diagnosis and personalized treatment plans.

12. Conclusions

IL-8 emerges as an important mediator in the crosstalk between the body’s defense mechanisms and the nervous system, demonstrating a profound influence on a multitude of psychoneuroimmunological processes. This underscores its potential role in the pathogenesis of various neuropsychiatric conditions. The current data highlight the complex interplay between IL-8, ROS, environmental factors, psychosocial stressors, and genetic backgrounds, underscoring the need for further studies to fully understand these potential influences and their implications for personalized medicine. Polymorphisms in the CXCL8 gene can impact IL-8 production, potentially leading to diverse research findings related to disease susceptibility, progression, and severity across different populations. Therefore, further research on IL-8 is essential to enhance our knowledge of its diverse roles in physiologic and pathologic processes, and to consider individual variations. Its potential use as a biomarker could ultimately lead to more personalized and effective strategies for diagnosing and treating patients with neuropsychiatric conditions.

Author Contributions

Methodology, data curation, writing—original draft preparation, A.S.; review and editing, A.H.; supervision, conceptualization, A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable. The study did not require ethical approval.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article. No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Yektaei-Karin, E.; Moshfegh, A.; Lundahl, J.; Berggren, V.; Hansson, L.O.; Marchini, G. The stress of birth en-hances in vitro spontaneous and IL-8-induced neutrophil chemotaxis in the human newborn. Pediatr. Allergy Immunol. Off. Publ. Eur. Soc. Pediatr. Allergy Immunol. 2007, 18, 643–651. [Google Scholar] [CrossRef] [PubMed]
  2. Süß, P.; Hoffmann, A.; Rothe, T.; Ouyang, Z.; Baum, W.; Staszewski, O.; Schett, G.; Prinz, M.; Krönke, G.; Glass, C.K.; et al. Chronic Peripheral Inflammation Causes a Region-Specific Myeloid Re-sponse in the Central Nervous System. Cell Rep. 2020, 30, 4082–4095.e6. [Google Scholar] [CrossRef] [PubMed]
  3. Shouman, K.; Benarroch, E.E. Peripheral neuroimmune interactions: Selected review and some clinical implications. Clin. Auton. Res. Off. J. Clin. Auton. Res. Soc. 2021, 31, 477–489. [Google Scholar] [CrossRef] [PubMed]
  4. Chavan, S.S.; Pavlov, V.A.; Tracey, K.J. Mechanisms and Therapeutic Relevance of Neuro-immune Communication. Immunity 2017, 46, 927–942. [Google Scholar] [CrossRef] [PubMed]
  5. Halaris, A.; Bechter, K.; Haroon, E.; Leonard, B.E.; Miller, A.; Pariante, C.; Zunszain, P. The Future of Psychoneuroimmunology: Promises and Challenges. In Advances in Psychiatry; Javed, A., Fountoulakis, K.N., Eds.; Springer: Berlin/Heidelberg, Germany, 2019; pp. 235–266. [Google Scholar] [CrossRef]
  6. Halaris, A.; Sohl, E.; Whitham, E.A. Treatment-Resistant Depression Revisited: A Glimmer of Hope. J. Pers. Med. 2021, 11, 155. [Google Scholar] [CrossRef]
  7. Ramesh, G.; MacLean, A.G.; Philipp, M.T. Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain. Mediat. Inflamm. 2013, 2013, 480739. [Google Scholar] [CrossRef]
  8. Salvi, V.; Sozio, F.; Sozzani, S.; Del Prete, A. Role of Atypical Chemokine Receptors in Microglial Activation and Polarization. Front. Aging Neurosci. 2017, 9, 148. [Google Scholar] [CrossRef] [PubMed]
  9. Russo, R.C.; Garcia, C.C.; Teixeira, M.M.; Amaral, F.A. The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev. Clin. Immunol. 2014, 10, 593–619. [Google Scholar] [CrossRef]
  10. Hughes, C.E.; Nibbs, R.J.B. A guide to chemokines and their receptors. FEBS J. 2018, 285, 2944–2971. [Google Scholar] [CrossRef]
  11. Vlachogiannis, P.; Hillered, L.; Enblad, P.; Ronne-Engström, E. Elevated levels of several chemokines in the cerebrospinal fluid of patients with subarachnoid hemorrhage are associated with worse clinical outcome. PLoS ONE 2023, 18, e0282424. [Google Scholar] [CrossRef]
  12. Banisadr, G.; Rostène, W.; Kitabgi, P.; Parsadaniantz, S.M. Chemokines and brain functions. Current drug targets. Inflamm. Allergy 2005, 4, 387–399. [Google Scholar] [CrossRef] [PubMed]
  13. Cartier, L.; Hartley, O.; Dubois-Dauphin, M.; Krause, K.H. Chemokine receptors in the central nervous system: Role in brain inflammation and neurodegenerative diseases. Brain research. Brain Res. Rev. 2005, 48, 16–42. [Google Scholar] [CrossRef] [PubMed]
  14. Réaux-Le Goazigo, A.; Van Steenwinckel, J.; Rostène, W.; Mélik Parsadaniantz, S. Current status of chemokines in the adult CNS. Prog. Neurobiol. 2013, 104, 67–92. [Google Scholar] [CrossRef] [PubMed]
  15. Turner, M.D.; Nedjai, B.; Hurst, T.; Pennington, D.J. Cytokines and chemokines: At the crossroads of cell sig-nalling and inflammatory disease. Biochim. Et Biophys. Acta 2014, 1843, 2563–2582. [Google Scholar] [CrossRef] [PubMed]
  16. Watson AE, S.; Goodkey, K.; Footz, T.; Voronova, A. Regulation of CNS precursor function by neuronal chemokines. Neurosci. Lett. 2020, 715, 134533. [Google Scholar] [CrossRef] [PubMed]
  17. Callewaere, C.; Banisadr, G.; Rostène, W.; Parsadaniantz, S.M. Chemokines and chemokine receptors in the brain: Implication in neuroendocrine regulation. J. Mol. Endocrinol. 2007, 38, 355–363. [Google Scholar] [CrossRef] [PubMed]
  18. Rostène, W.; Kitabgi, P.; Parsadaniantz, S.M. Chemokines: A new class of neuromodulator? Nature reviews. Neuroscience 2007, 8, 895–903. [Google Scholar] [CrossRef] [PubMed]
  19. Rostène, W.; Dansereau, M.A.; Godefroy, D.; Van Steenwinckel, J.; Reaux-Le Goazigo, A.; Mélik-Parsadaniantz, S.; Apartis, E.; Hunot, S.; Beaudet, N.; Sarret, P. Neurochemokines: A menage a trois providing new insights on the functions of chemokines in the central nervous system. J. Neurochem. 2011, 118, 680–694. [Google Scholar] [CrossRef] [PubMed]
  20. Rostène, W.; Guyon, A.; Kular, L.; Godefroy, D.; Barbieri, F.; Bajetto, A.; Banisadr, G.; Callewaere, C.; Conductier, G.; Rovère, C.; et al. Chemokines and chemokine receptors: New actors in neuroendocrine regulations. Front. Neuroendocrinol. 2011, 32, 10–24. [Google Scholar] [CrossRef]
  21. Jaerve, A.; Müller, H.W. Chemokines in CNS injury and repair. Cell Tissue Res. 2012, 349, 229–248. [Google Scholar] [CrossRef]
  22. Steinbach, G.; Bölke, E.; Schulte am Esch, J.; Peiper, M.; Zant, R.; Schwarz, A.; Spiess, B.; van Griensven, M.; Orth, K. Comparison of whole blood interleukin-8 and plasma interleukin-8 as a predictor for sepsis in postoperative patients. Clin. Chim. Acta Int. J. Clin. Chem. 2007, 378, 117–121. [Google Scholar] [CrossRef] [PubMed]
  23. Williams, M.A.; Cave, C.M.; Quaid, G.; Robinson, C.; Daly, T.J.; Witt, D.; Lentsch, A.B.; Solomkin, J.S. Interleukin 8 dimerization as a mechanism for regulation of neutrophil adherence-dependent oxidant production. Shock 2005, 23, 371–376. [Google Scholar] [CrossRef] [PubMed]
  24. Waugh, D.J.; Wilson, C. The interleukin-8 pathway in cancer. Clin. Cancer Res.Off. J. Am. Assoc. Cancer Res. 2008, 14, 6735–6741. [Google Scholar] [CrossRef] [PubMed]
  25. David, J.M.; Dominguez, C.; Hamilton, D.H.; Palena, C. The IL-8/IL-8R Axis: A Double Agent in Tumor Immune Resistance. Vaccines 2016, 4, 22. [Google Scholar] [CrossRef] [PubMed]
  26. Liu, K.; Wu, L.; Yuan, S.; Wu, M.; Xu, Y.; Sun, Q.; Li, S.; Zhao, S.; Hua, T.; Liu, Z.J. Structural basis of CXC chemokine receptor 2 activation and signalling. Nature 2020, 585, 135–140. [Google Scholar] [CrossRef] [PubMed]
  27. Sitaru, S.; Budke, A.; Bertini, R.; Sperandio, M. Therapeutic inhibition of CXCR1/2: Where do we stand? Intern. Emerg. Med. 2023, 18, 1647–1664. [Google Scholar] [CrossRef] [PubMed]
  28. Palomino, D.C.; Marti, L.C. Chemokines and immunity. Einstein 2015, 13, 469–473. [Google Scholar] [CrossRef]
  29. Mukaida, N.; Hishinuma, A.; Zachariae, C.O.; Oppenheim, J.J.; Matsushima, K. Regulation of human interleukin 8 gene expression and binding of several other members of the intercrine family to receptors for interleukin-8. Adv. Exp. Med. Biol. 1991, 305, 31–38. [Google Scholar] [CrossRef] [PubMed]
  30. Baggiolini, M.; Clark-Lewis, I. Interleukin-8, a chemotactic and inflammatory cytokine. FEBS Lett. 1992, 307, 97–101. [Google Scholar] [CrossRef]
  31. Matsumoto, K.; Hashimoto, S.; Gon, Y.; Nakayama, T.; Takizawa, H.; Horie, T. N-acetylcysteine inhibits IL-1 alpha-induced IL-8 secretion by bronchial epithelial cells. Respir. Med. 1998, 92, 512–515. [Google Scholar] [CrossRef]
  32. Malik, A.; Kanneganti, T.D. Function and regulation of IL-1α in inflammatory diseases and cancer. Immunol. Rev. 2018, 281, 124–137. [Google Scholar] [CrossRef] [PubMed]
  33. Ehrlich, L.C.; Hu, S.; Sheng, W.S.; Sutton, R.L.; Rockswold, G.L.; Peterson, P.K.; Chao, C.C. Cytokine regulation of human microglial cell IL-8 production. J. Immunol. 1998, 160, 1944–1948. [Google Scholar] [CrossRef] [PubMed]
  34. Kim, G.Y.; Lee, J.W.; Ryu, H.C.; Wei, J.D.; Seong, C.M.; Kim, J.H. Proinflammatory cytokine IL-1beta stimulates IL-8 synthesis in mast cells via a leukotriene B4 receptor 2-linked pathway, contributing to angiogenesis. J. Immunol. 2010, 184, 3946–3954. [Google Scholar] [CrossRef]
  35. Qazi, B.S.; Tang, K.; Qazi, A. Recent advances in underlying pathologies provide insight into interleukin-8 expression-mediated inflammation and angiogenesis. Int. J. Inflamm. 2011, 2011, 908468. [Google Scholar] [CrossRef]
  36. Standiford, T.J.; Strieter, R.M.; Allen, R.M.; Burdick, M.D.; Kunkel, S.L. IL-7 up-regulates the expression of IL-8 from resting and stimulated human blood monocytes. J. Immunol. 1992, 149, 2035–2039. [Google Scholar] [CrossRef] [PubMed]
  37. Jones, C.E.; Chan, K. Interleukin-17 stimulates the expression of interleukin-8, growth-related oncogene-alpha, and granulocyte-colony-stimulating factor by human airway epithelial cells. Am. J. Respir. Cell Mol. Biol. 2002, 26, 748–753. [Google Scholar] [CrossRef]
  38. Hwang, S.Y.; Kim, J.Y.; Kim, K.W.; Park, M.K.; Moon, Y.; Kim, W.U.; Kim, H.Y. IL-17 induces production of IL-6 and IL-8 in rheumatoid arthritis synovial fibroblasts via NF-kappaB- and PI3-kinase/Akt-dependent pathways. Arthritis Res. Ther. 2004, 6, R120–R128. [Google Scholar] [CrossRef]
  39. Tokura, Y.; Mori, T.; Hino, R. Psoriasis and other Th17-mediated skin diseases. J. UOEH 2010, 32, 317–328. [Google Scholar] [CrossRef]
  40. Kabashima, R.; Sugita, K.; Sawada, Y.; Hino, R.; Nakamura, M.; Tokura, Y. Increased circulating Th17 frequencies and serum IL-22 levels in patients with acute generalized exanthematous pustulosis. J. Eur. Acad. Dermatol. Venereol. JEADV 2011, 25, 485–488. [Google Scholar] [CrossRef]
  41. Dixon, B.R.; Radin, J.N.; Piazuelo, M.B.; Contreras, D.C.; Algood, H.M. IL-17a and IL-22 Induce Expression of Antimicrobials in Gastrointestinal Epithelial Cells and May Contribute to Epithelial Cell Defense against Helicobacter pylori. PLoS ONE 2016, 11, e0148514. [Google Scholar] [CrossRef]
  42. Huang, Q.; Duan, L.; Qian, X.; Fan, J.; Lv, Z.; Zhang, X.; Han, J.; Wu, F.; Guo, M.; Hu, G.; et al. IL-17 Promotes Angiogenic Factors IL-6, IL-8, and Vegf Production via Stat1 in Lung Adenocarcinoma. Sci. Rep. 2016, 6, 36551. [Google Scholar] [CrossRef] [PubMed]
  43. Guo, Y.; Chen, Y.; Liu, L.B.; Chang, K.K.; Li, H.; Li, M.Q.; Shao, J. IL-22 in the endometriotic milieu promotes the proliferation of endometrial stromal cells via stimulating the secretion of CCL2 and IL-8. Int. J. Clin. Exp. Pathol. 2013, 6, 2011–2020. [Google Scholar]
  44. Mukaida, N.; Morita, M.; Ishikawa, Y.; Rice, N.; Okamoto, S.; Kasahara, T.; Matsushima, K. Novel mechanism of glucocorticoid-mediated gene repression. Nuclear factor-kappa B is target for glucocorticoid-mediated interleukin 8 gene repression. J. Biol. Chem. 1994, 269, 13289–13295. [Google Scholar] [CrossRef] [PubMed]
  45. Brat, D.J.; Bellail, A.C.; Van Meir, E.G. The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro-Oncol. 2005, 7, 122–133. [Google Scholar] [CrossRef] [PubMed]
  46. Suwa, E.; Yamaura, K.; Sato, S.; Ueno, K. Increased expression of the histamine H4 receptor following differentiation and mediation of the H4 receptor on interleukin-8 mRNA expression in HaCaT keratinocytes. Exp. Dermatol. 2014, 23, 138–140. [Google Scholar] [CrossRef]
  47. Jeannin, P.; Delneste, Y.; Gosset, P.; Molet, S.; Lassalle, P.; Hamid, Q.; Tsicopoulos, A.; Tonnel, A.B. Histamine induces interleukin-8 secretion by endothelial cells. Blood 1994, 84, 2229–2233. [Google Scholar] [CrossRef]
  48. Bachert, C. Histamine—A major role in allergy? Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 1998, 28 (Suppl. S6), 15–19. [Google Scholar] [CrossRef]
  49. Utgaard, J.O.; Jahnsen, F.L.; Bakka, A.; Brandtzaeg, P.; Haraldsen, G. Rapid secretion of prestored interleukin 8 from Weibel-Palade bodies of microvascular endothelial cells. J. Exp. Med. 1998, 188, 1751–1756. [Google Scholar] [CrossRef]
  50. Li, H.; Guo, D.; Zhang, L.; Feng, X. Glycyrrhizin attenuates histamine-mediated MUC5AC upregulation, inflammatory cytokine production, and aquaporin 5 downregulation through suppressing the NF-κB pathway in human nasal epithelial cells. Chem. -Biol. Interact. 2018, 285, 21–26. [Google Scholar] [CrossRef]
  51. Lin, T.J.; Issekutz, T.B.; Marshall, J.S. Human mast cells transmigrate through human umbilical vein endothelial monolayers and selectively produce IL-8 in response to stromal cell-derived factor-1 alpha. J. Immunol. 2000, 165, 211–220. [Google Scholar] [CrossRef]
  52. Lin, T.J.; Issekutz, T.B.; Marshall, J.S. SDF-1 induces IL-8 production and transendothelial migration of human cord blood-derived mast cells. Int. Arch. Allergy Immunol. 2001, 124, 142–145. [Google Scholar] [CrossRef] [PubMed]
  53. Scupoli, M.T.; Donadelli, M.; Cioffi, F.; Rossi, M.; Perbellini, O.; Malpeli, G.; Corbioli, S.; Vinante, F.; Krampera, M.; Palmieri, M.; et al. Bone marrow stromal cells and the upregulation of interleukin-8 production in human T-cell acute lymphoblastic leukemia through the CXCL12/CXCR4 axis and the NF-kappaB and JNK/AP-1 pathways. Haematologica 2008, 93, 524–532. [Google Scholar] [CrossRef] [PubMed]
  54. Li, K.C.; Huang, Y.H.; Ho, C.Y.; Chu, C.Y.; Cha, S.T.; Tsai, H.H.; Ko, J.Y.; Chang, C.C.; Tan, C.T. The role of IL-8 in the SDF-1α/CXCR4-induced angiogenesis of laryngeal and hypopharyngeal squamous cell carcinoma. Oral Oncol. 2012, 48, 507–515. [Google Scholar] [CrossRef]
  55. Zhou, L.; Zhao, H.; Zhang, C.; Chen, Z.; Li, D.; Qian, G. Study on the mechanism of CXCL12/CXCR4-axis-mediated upregulation of IL-8 and IL-6 on the biological function of acute T lymphocyte leukaemia cells. Cytotechnology 2024, 76, 97–111. [Google Scholar] [CrossRef] [PubMed]
  56. Liebler, J.M.; Kunkel, S.L.; Burdick, M.D.; Standiford, T.J.; Rolfe, M.W.; Strieter, R.M. Production of IL-8 and monocyte chemotactic peptide-1 by peripheral blood monocytes. Disparate responses to phytohemagglutinin and lipopolysaccharide. J. Immunol. 1994, 152, 241–249. [Google Scholar] [CrossRef] [PubMed]
  57. Tanaka, C.; Kamata, H.; Takeshita, H.; Yagisawa, H.; Hirata, H. Redox regulation of lipopolysaccharide (LPS)-induced interleukin-8 (IL-8) gene expression mediated by NF kappa B and AP-1 in human astrocytoma U373 cells. Biochem. Biophys. Res. Commun. 1997, 232, 568–573. [Google Scholar] [CrossRef] [PubMed]
  58. Aydin, M.; Ozkok, E.; Ozturk, O.; Agachan, B.; Yilmaz, H.; Yaylim, I.; Kebabcioglu, S.; Ispir, T. Relationship between interleukin-8 and the oxidant-antioxidant system in end-stage renal failure patients. Exp. Clin. Transplant. Off. J. Middle East Soc. Organ Transplant. 2007, 5, 610–613. [Google Scholar]
  59. Ko, J.W.; Lim, S.Y.; Chung, K.C.; Lim, J.W.; Kim, H. Reactive oxygen species mediate IL-8 expression in Down syndrome candidate region-1-overexpressed cells. Int. J. Biochem. Cell Biol. 2014, 55, 164–170. [Google Scholar] [CrossRef] [PubMed]
  60. Pascoe, C.D.; Roy, N.; Turner-Brannen, E.; Schultz, A.; Vaghasiya, J.; Ravandi, A.; Halayko, A.J.; West, A.R. Oxidized phosphatidylcholines induce multiple functional defects in airway epithelial cells. American journal of physiology. Lung Cell. Mol. Physiol. 2021, 321, L703–L717. [Google Scholar] [CrossRef]
  61. Hyun, J.S.; Satsu, H.; Shimizu, M. Cadmium induces interleukin-8 production via NF-kappaB activation in the human intestinal epithelial cell, Caco-2. Cytokine 2007, 37, 26–34. [Google Scholar] [CrossRef]
  62. Cormet-Boyaka, E.; Jolivette, K.; Bonnegarde-Bernard, A.; Rennolds, J.; Hassan, F.; Mehta, P.; Tridandapani, S.; Webster-Marketon, J.; Boyaka, P.N. An NF-κB-independent and Erk1/2-dependent mechanism controls CXCL8/IL-8 responses of airway epithelial cells to cadmium. Toxicol. Sci. Off. J. Soc. Toxicol. 2012, 125, 418–429. [Google Scholar] [CrossRef] [PubMed]
  63. Phuagkhaopong, S.; Ospondpant, D.; Kasemsuk, T.; Sibmooh, N.; Soodvilai, S.; Power, C.; Vivithanaporn, P. Cadmium-induced IL-6 and IL-8 expression and release from astrocytes are mediated by MAPK and NF-κB pathways. Neurotoxicology 2017, 60, 82–91. [Google Scholar] [CrossRef] [PubMed]
  64. Zhang, Y.; Li, Y.; Zhang, J.; Qi, X.; Cui, Y.; Yin, K.; Lin, H. Cadmium induced inflammation and apoptosis of porcine epididymis via activating RAF1/MEK/ERK and NF-κB pathways. Toxicol. Appl. Pharmacol. 2021, 415, 115449. [Google Scholar] [CrossRef] [PubMed]
  65. Yu, Y.; Chadee, K. Prostaglandin E2 stimulates IL-8 gene expression in human colonic epithelial cells by a posttranscriptional mechanism. J. Immunol. 1998, 161, 3746–3752. [Google Scholar] [CrossRef]
  66. Caristi, S.; Piraino, G.; Cucinotta, M.; Valenti, A.; Loddo, S.; Teti, D. Prostaglandin E2 induces interleukin-8 gene transcription by activating C/EBP homologous protein in human T lymphocytes. J. Biol. Chem. 2005, 280, 14433–14442. [Google Scholar] [CrossRef]
  67. Neuschäfer-Rube, F.; Pathe-Neuschäfer-Rube, A.; Hippenstiel, S.; Kracht, M.; Püschel, G.P. NF-κB-dependent IL-8 induction by prostaglandin E(2) receptors EP(1) and EP(4). Br. J. Pharmacol. 2013, 168, 704–717. [Google Scholar] [CrossRef]
  68. Cho, J.S.; Han, I.H.; Lee, H.R.; Lee, H.M. Prostaglandin E2 Induces IL-6 and IL-8 Production by the EP Receptors/Akt/NF-κB Pathways in Nasal Polyp-Derived Fibroblasts. Allergy Asthma Immunol. Res. 2014, 6, 449–457. [Google Scholar] [CrossRef]
  69. Doukas, J.; Cutler, A.H.; Mordes, J.P. Polyinosinic:polycytidylic acid is a potent activator of endothelial cells. Am. J. Pathol. 1994, 145, 137–147. [Google Scholar]
  70. Takada, K.; Komine-Aizawa, S.; Hirohata, N.; Trinh, Q.D.; Nishina, A.; Kimura, H.; Hayakawa, S. Poly I:C induces collective migration of HaCaT keratinocytes via IL-8. BMC Immunol. 2017, 18, 19. [Google Scholar] [CrossRef]
  71. Hasséus, B.; Jontell, M.; Bergenholtz, G.; Dahlgren, U.I. Langerhans cells from human oral epithelium are more effective at stimulating allogeneic T cells in vitro than Langerhans cells from skin. Clin. Exp. Immunol. 2004, 136, 483–489. [Google Scholar] [CrossRef]
  72. Shapiro, L.; Dinarello, C.A. Osmotic regulation of cytokine synthesis in vitro. Proc. Natl. Acad. Sci. USA 1995, 92, 12230–12234. [Google Scholar] [CrossRef]
  73. Tabary, O.; Muselet, C.; Escotte, S.; Antonicelli, F.; Hubert, D.; Dusser, D.; Jacquot, J. Interleukin-10 inhibits elevated chemokine interleukin-8 and regulated on activation normal T cell expressed and secreted production in cystic fibrosis bronchial epithelial cells by targeting the I(k)B kinase alpha/beta complex. Am. J. Pathol. 2003, 162, 293–302. [Google Scholar] [CrossRef] [PubMed]
  74. Chan, M.M.; Chmura, K.; Chan, E.D. Increased NaCl-induced interleukin-8 production by human bronchial epithelial cells is enhanced by the DeltaF508/W1282X mutation of the cystic fibrosis transmembrane conductance regulator gene. Cytokine 2006, 33, 309–316. [Google Scholar] [CrossRef] [PubMed]
  75. Mazzitelli, I.; Bleichmar, L.; Melucci, C.; Gerber, P.P.; Toscanini, A.; Cuestas, M.L.; Diaz, F.E.; Geffner, J. High Salt Induces a Delayed Activation of Human Neutrophils. Front. Immunol. 2022, 13, 831844. [Google Scholar] [CrossRef]
  76. Ueno, A.; Murakami, K.; Yamanouchi, K.; Watanabe, M.; Kondo, T. Thrombin stimulates production of interleukin-8 in human umbilical vein endothelial cells. Immunology 1996, 88, 76–81. [Google Scholar] [CrossRef] [PubMed]
  77. Zheng, L.; Martins-Green, M. Molecular mechanisms of thrombin-induced interleukin-8 (IL-8/CXCL8) expression in THP-1-derived and primary human macrophages. J. Leukoc. Biol. 2007, 82, 619–629. [Google Scholar] [CrossRef]
  78. Yuliani, F.S.; Chen, J.Y.; Cheng, W.H.; Wen, H.C.; Chen, B.C.; Lin, C.H. Thrombin induces IL-8/CXCL8 expression by DCLK1-dependent RhoA and YAP activation in human lung epithelial cells. J. Biomed. Sci. 2022, 29, 95. [Google Scholar] [CrossRef] [PubMed]
  79. Zhang, Q.Y.; Hammerberg, C.; Baldassare, J.J.; Henderson, P.A.; Burns, D.; Ceska, M.; Voorhees, J.J.; Fisher, G.J. Retinoic acid and phorbol ester synergistically up-regulate IL-8 expression and specifically modulate protein kinase C-epsilon in human skin fibroblasts. J. Immunol. 1992, 149, 1402–1408. [Google Scholar] [CrossRef] [PubMed]
  80. Chang, M.M.; Harper, R.; Hyde, D.M.; Wu, R. A novel mechanism of retinoic acid-enhanced interleukin-8 gene expression in airway epithelium. Am. J. Respir. Cell Mol. Biol. 2000, 22, 502–510. [Google Scholar] [CrossRef]
  81. Dai, X.; Yamasaki, K.; Shirakata, Y.; Sayama, K.; Hashimoto, K. All-trans-retinoic acid induces interleukin-8 via the nuclear factor-kappaB and p38 mitogen-activated protein kinase pathways in normal human keratinocytes. J. Investig. Dermatol. 2004, 123, 1078–1085. [Google Scholar] [CrossRef]
  82. Aman, M.J.; Rudolf, G.; Goldschmitt, J.; Aulitzky, W.E.; Lam, C.; Huber, C.; Peschel, C. Type-I interferons are potent inhibitors of interleukin-8 production in hematopoietic and bone marrow stromal cells. Blood 1993, 82, 2371–2378. [Google Scholar] [CrossRef] [PubMed]
  83. Wang, P.; Wu, P.; Anthes, J.C.; Siegel, M.I.; Egan, R.W.; Billah, M.M. Interleukin-10 inhibits interleukin-8 production in human neutrophils. Blood 1994, 83, 2678–2683. [Google Scholar] [CrossRef] [PubMed]
  84. Jiang, S.; Li, Y.; Lin, T.; Yuan, L.; Li, Y.; Wu, S.; Xia, L.; Shen, H.; Lu, J. IL-35 Inhibits Angiogenesis through VEGF/Ang2/Tie2 Pathway in Rheumatoid Arthritis. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2016, 40, 1105–1116. [Google Scholar] [CrossRef] [PubMed]
  85. Shindo, S.; Hosokawa, Y.; Hosokawa, I.; Shiba, H. Interleukin (IL)-35 Suppresses IL-6 and IL-8 Production in IL-17A-Stimulated Human Periodontal Ligament Cells. Inflammation 2019, 42, 835–840. [Google Scholar] [CrossRef] [PubMed]
  86. Li, M.; Liu, Y.; Fu, Y.; Gong, R.; Xia, H.; Huang, X.; Wu, Y. Interleukin-35 inhibits lipopolysaccharide-induced endothelial cell activation by downregulating inflammation and apoptosis. Exp. Cell Res. 2021, 407, 112784. [Google Scholar] [CrossRef] [PubMed]
  87. Smith, W.B.; Noack, L.; Khew-Goodall, Y.; Isenmann, S.; Vadas, M.A.; Gamble, J.R. Transforming growth factor-beta 1 inhibits the production of IL-8 and the transmigration of neutrophils through activated endothelium. J. Immunol. 1996, 157, 360–368. [Google Scholar] [CrossRef] [PubMed]
  88. Ge, Q.; Moir, L.M.; Black, J.L.; Oliver, B.G.; Burgess, J.K. TGFβ1 induces IL-6 and inhibits IL-8 release in human bronchial epithelial cells: The role of Smad2/3. J. Cell. Physiol. 2010, 225, 846–854. [Google Scholar] [CrossRef] [PubMed]
  89. Singh, R.K.; Varney, M.L. Regulation of interleukin 8 expression in human malignant melanoma cells. Cancer Res. 1998, 58, 1532–1537. [Google Scholar] [PubMed]
  90. Wang, T.; Takikawa, Y.; Sawara, K.; Yoshida, Y.; Suzuki, K. Negative regulation of human astrocytes by interferon (IFN) α in relation to growth inhibition and impaired glucose utilization. Neurochem. Res. 2012, 37, 1898–1905. [Google Scholar] [CrossRef]
  91. Nozell, S.; Laver, T.; Patel, K.; Benveniste, E.N. Mechanism of IFN-beta-mediated inhibition of IL-8 gene expression in astroglioma cells. J. Immunol. 2006, 177, 822–830. [Google Scholar] [CrossRef]
  92. Laver, T.; Nozell, S.E.; Benveniste, E.N. IFN-beta-mediated inhibition of IL-8 expression requires the ISGF3 components Stat1, Stat2, and IRF-9. J. Interferon Cytokine Res. Off. J. Int. Soc. Interferon Cytokine Res. 2008, 28, 13–23. [Google Scholar] [CrossRef] [PubMed]
  93. Tobler, A.; Meier, R.; Seitz, M.; Dewald, B.; Baggiolini, M.; Fey, M.F. Glucocorticoids downregulate gene expression of GM-CSF, NAP-1/IL-8, and IL-6, but not of M-CSF in human fibroblasts. Blood 1992, 79, 45–51. [Google Scholar] [CrossRef] [PubMed]
  94. Bonnans, C.; Vachier, I.; Chavis, C.; Godard, P.; Bousquet, J.; Chanez, P. Lipoxins are potential endogenous antiinflammatory mediators in asthma. Am. J. Respir. Crit. Care Med. 2002, 165, 1531–1535. [Google Scholar] [CrossRef] [PubMed]
  95. Bonnans, C.; Gras, D.; Chavis, C.; Mainprice, B.; Vachier, I.; Godard, P.; Chanez, P. Synthesis and anti-inflammatory effect of lipoxins in human airway epithelial cells. Biomed. Pharmacother. Biomed. Pharmacother. 2007, 61, 261–267. [Google Scholar] [CrossRef] [PubMed]
  96. Ringholz, F.C.; Buchanan, P.J.; Clarke, D.T.; Millar, R.G.; McDermott, M.; Linnane, B.; Harvey, B.J.; McNally, P.; Urbach, V. Reduced 15-lipoxygenase 2 and lipoxin A4/leukotriene B4 ratio in children with cystic fibrosis. Eur. Respir. J. 2014, 44, 394–404. [Google Scholar] [CrossRef] [PubMed]
  97. Tang, X.; Pan, Y.; Zhao, Y. Vitamin D inhibits the expression of interleukin-8 in human periodontal ligament cells stimulated with Porphyromonas gingivalis. Arch. Oral Biol. 2013, 58, 397–407. [Google Scholar] [CrossRef] [PubMed]
  98. Dauletbaev, N.; Herscovitch, K.; Das, M.; Chen, H.; Bernier, J.; Matouk, E.; Bérubé, J.; Rousseau, S.; Lands, L.C. Down-regulation of IL-8 by high-dose vitamin D is specific to hyperinflammatory macrophages and involves mechanisms beyond up-regulation of DUSP1. Br. J. Pharmacol. 2015, 172, 4757–4771. [Google Scholar] [CrossRef] [PubMed]
  99. Hosokawa, Y.; Hosokawa, I.; Shindo, S.; Ozaki, K.; Matsuo, T. Calcitriol Suppressed Inflammatory Reactions in IL-1β-Stimulated Human Periodontal Ligament Cells. Inflammation 2015, 38, 2252–2258. [Google Scholar] [CrossRef] [PubMed]
  100. Tahan, F.; Jazrawi, E.; Moodley, T.; Rovati, G.E.; Adcock, I.M. Montelukast inhibits tumour necrosis factor-alpha-mediated interleukin-8 expression through inhibition of nuclear factor-kappaB p65-associated histone acetyltransferase activity. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2008, 38, 805–811. [Google Scholar] [CrossRef]
  101. Pihlaja, R.; Haaparanta-Solin, M.; Rinne, J.O. The Anti-Inflammatory Effects of Lipoxygenase and Cyclo-Oxygenase Inhibitors in Inflammation-Induced Human Fetal Glia Cells and the Aβ Degradation Capacity of Human Fetal Astrocytes in an Ex vivo Assay. Front. Neurosci. 2017, 11, 299. [Google Scholar] [CrossRef]
  102. Achudhan, D.; Liu, S.C.; Lin, Y.Y.; Huang, C.C.; Tsai, C.H.; Ko, C.Y.; Chiang, I.P.; Kuo, Y.H.; Tang, C.H. Antcin K Inhibits TNF-α, IL-1β and IL-8 Expression in Synovial Fibroblasts and Ameliorates Cartilage Degradation: Implications for the Treatment of Rheumatoid Arthritis. Front. Immunol. 2021, 12, 790925. [Google Scholar] [CrossRef] [PubMed]
  103. Fumagalli, M.; Sangiovanni, E.; Vrhovsek, U.; Piazza, S.; Colombo, E.; Gasperotti, M.; Mattivi, F.; De Fabiani, E.; Dell’Agli, M. Strawberry tannins inhibit IL-8 secretion in a cell model of gastric inflammation. Pharmacol. Res. 2016, 111, 703–712. [Google Scholar] [CrossRef] [PubMed]
  104. Lorenz, P.; Heinrich, M.; Garcia-Käufer, M.; Grunewald, F.; Messerschmidt, S.; Herrick, A.; Gruber, K.; Beckmann, C.; Knoedler, M.; Huber, R.; et al. Constituents from oak bark (Quercus robur L.) inhibit degranulation and allergic mediator release from basophils and mast cells in vitro. J. Ethnopharmacol. 2016, 194, 642–650. [Google Scholar] [CrossRef] [PubMed]
  105. Matsui, S.; Matsumoto, H.; Sonoda, Y.; Ando, K.; Aizu-Yokota, E.; Sato, T.; Kasahara, T. Glycyrrhizin and related compounds down-regulate production of inflammatory chemokines IL-8 and eotaxin 1 in a human lung fibroblast cell line. Int. Immunopharmacol. 2004, 4, 1633–1644. [Google Scholar] [CrossRef] [PubMed]
  106. Takei, H.; Baba, Y.; Hisatsune, A.; Katsuki, H.; Miyata, T.; Yokomizo, K.; Isohama, Y. Glycyrrhizin inhibits interleukin-8 production and nuclear factor-kappaB activity in lung epithelial cells, but not through glucocorticoid receptors. J. Pharmacol. Sci. 2008, 106, 460–468. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, N.; Lv, H.; Shi, B.H.; Hou, X.; Xu, X. Inhibition of IL-6 and IL-8 production in LPS-stimulated human gingival fibroblasts by glycyrrhizin via activating LXRα. Microb. Pathog. 2017, 110, 135–139. [Google Scholar] [CrossRef] [PubMed]
  108. Radomska-Leśniewska, D.M.; Skopińska-Rózewska, E.; Jankowska-Steifer, E.; Sobiecka, M.; Sadowska, A.M.; Hevelke, A.; Malejczyk, J. N-acetylcysteine inhibits IL-8 and MMP-9 release and ICAM-1 expression by bronchoalveolar cells from interstitial lung disease patients. Pharmacol. Rep. PR 2010, 62, 131–138. [Google Scholar] [CrossRef] [PubMed]
  109. Faghfouri, A.H.; Zarezadeh, M.; Tavakoli-Rouzbehani, O.M.; Radkhah, N.; Faghfuri, E.; Kord-Varkaneh, H.; Tan, S.C.; Ostadrahimi, A. The effects of N-acetylcysteine on inflammatory and oxidative stress biomarkers: A systematic review and meta-analysis of controlled clinical trials. Eur. J. Pharmacol. 2020, 884, 173368. [Google Scholar] [CrossRef] [PubMed]
  110. Mardani, N.; Mozafarpoor, S.; Goodarzi, A.; Nikkhah, F. A systematic review of N-acetylcysteine for treatment of acne vulgaris and acne-related associations and consequences: Focus on clinical studies. Dermatol. Ther. 2021, 34, e14915. [Google Scholar] [CrossRef]
  111. Remick, D.G. Interleukin-8. Crit. Care Med. 2005, 33 (Suppl. S12), S466–S467. [Google Scholar] [CrossRef]
  112. Apostolakis, S.; Vogiatzi, K.; Amanatidou, V.; Spandidos, D.A. Interleukin 8 and cardiovascular disease. Cardiovasc. Res. 2009, 84, 353–360. [Google Scholar] [CrossRef] [PubMed]
  113. Matsushima, K.; Yang, D.; Oppenheim, J.J. Interleukin-8: An evolving chemokine. Cytokine 2022, 153, 155828. [Google Scholar] [CrossRef] [PubMed]
  114. Nasser, M.W.; Raghuwanshi, S.K.; Malloy, K.M.; Gangavarapu, P.; Shim, J.Y.; Rajarathnam, K.; Richardson, R.M. CXCR1 and CXCR2 activation and regulation. Role of aspartate 199 of the second extracellular loop of CXCR2 in CXCL8-mediated rapid receptor internalization. J. Biol. Chem. 2007, 282, 6906–6915. [Google Scholar] [CrossRef] [PubMed]
  115. Stillie, R.; Farooq, S.M.; Gordon, J.R.; Stadnyk, A.W. The functional significance behind expressing two IL-8 receptor types on PMN. J. Leukoc. Biol. 2009, 86, 529–543. [Google Scholar] [CrossRef] [PubMed]
  116. Ha, H.; Debnath, B.; Neamati, N. Role of the CXCL8-CXCR1/2 Axis in Cancer and Inflammatory Diseases. Theranostics 2017, 7, 1543–1588. [Google Scholar] [CrossRef] [PubMed]
  117. Nasser, M.W.; Raghuwanshi, S.K.; Grant, D.J.; Jala, V.R.; Rajarathnam, K.; Richardson, R.M. Differential activation and regulation of CXCR1 and CXCR2 by CXCL8 monomer and dimer. J. Immunol. 2009, 183, 3425–3432. [Google Scholar] [CrossRef] [PubMed]
  118. Jones, S.A.; Wolf, M.; Qin, S.; Mackay, C.R.; Baggiolini, M. Different functions for the interleukin 8 receptors (IL-8R) of human neutrophil leukocytes: NADPH oxidase and phospholipase D are activated through IL-8R1 but not IL-8R2. Proc. Natl. Acad. Sci. USA 1996, 93, 6682–6686. [Google Scholar] [CrossRef] [PubMed]
  119. Richardson, R.M.; Pridgen, B.C.; Haribabu, B.; Ali, H.; Snyderman, R. Differential cross-regulation of the human chemokine receptors CXCR1 and CXCR2. Evidence for time-dependent signal generation. J. Biol. Chem. 1998, 273, 23830–23836. [Google Scholar] [CrossRef] [PubMed]
  120. Legler, D.F.; Thelen, M. New insights in chemokine signaling. F1000Research 2018, 7, 95. [Google Scholar] [CrossRef]
  121. Glennon-Alty, L.; Hackett, A.P.; Chapman, E.A.; Wright, H.L. Neutrophils and redox stress in the pathogenesis of autoimmune disease. Free. Radic. Biol. Med. 2018, 125, 25–35. [Google Scholar] [CrossRef]
  122. Ishimoto, N.; Park, J.H.; Kawakami, K.; Tajiri, M.; Mizutani, K.; Akashi, S.; Tame, J.R.H.; Inoue, A.; Park, S.Y. Structural basis of CXC chemokine receptor 1 ligand binding and activation. Nat. Commun. 2023, 14, 4107. [Google Scholar] [CrossRef] [PubMed]
  123. Licinio, J.; Wong, M.L.; Gold, P.W. Neutrophil-activating peptide-1/interleukin-8 mRNA is localized in rat hypothalamus and hippocampus. Neuroreport 1992, 3, 753–756. [Google Scholar] [CrossRef] [PubMed]
  124. Hirsch, G.; Lavoie-Lamoureux, A.; Beauchamp, G.; Lavoie, J.P. Neutrophils are not less sensitive than other blood leukocytes to the genomic effects of glucocorticoids. PLoS ONE 2012, 7, e44606. [Google Scholar] [CrossRef] [PubMed]
  125. Fujio, N.; Masuoka, S.; Shikano, K.; Kusunoki, N.; Nanki, T.; Kawai, S. Apparent Hypothalamic-Pituitary-Adrenal Axis Suppression via Reduction of Interleukin-6 by Glucocorticoid Therapy in Systemic Autoimmune Diseases. PLoS ONE 2016, 11, e0167854. [Google Scholar] [CrossRef]
  126. Hoffman, C.L.; Higham, J.P.; Heistermann, M.; Coe, C.L.; Prendergast, B.J.; Maestripieri, D. Immune function and HPA axis activity in free-ranging rhesus macaques. Physiol. Behav. 2011, 104, 507–514. [Google Scholar] [CrossRef]
  127. Cui, G.B.; An, J.Z.; Zhang, N.; Zhao, M.G.; Liu, S.B.; Yi, J. Elevated interleukin-8 enhances prefrontal synaptic transmission in mice with persistent inflammatory pain. Mol. Pain 2012, 8, 11. [Google Scholar] [CrossRef] [PubMed]
  128. Ghoryani, M.; Faridhosseini, F.; Talaei, A.; Faridhosseini, R.; Tavakkol-Afshari, J.; Dadgar Moghaddam, M.; Azim, P.; Salimi, Z.; Marzouni, H.Z.; Mohammadi, M. Gene expression pattern of CCL2, CCL3, and CXCL8 in patients with bipolar disorder. J. Res. Med. Sci. Off. J. Isfahan Univ. Med. Sci. 2019, 24, 45. [Google Scholar] [CrossRef] [PubMed]
  129. Chang, C.F.; Goods, B.A.; Askenase, M.H.; Beatty, H.E.; Osherov, A.; DeLong, J.H.; Hammond, M.D.; Massey, J.; Landreneau, M.; Love, J.C.; et al. Divergent Functions of Tissue-Resident and Blood-Derived Macrophages in the Hemorrhagic Brain. Stroke 2021, 52, 1798–1808. [Google Scholar] [CrossRef]
  130. Watanabe, Y.; Miura, I.; Ohgami, Y.; Fujiwara, M. Extracellular presence of IL-8 in the astrocyte-rich cultured cerebellar granule cells under acidosis. Life Sci. 1998, 63, 1037–1046. [Google Scholar] [CrossRef]
  131. Robinson, K.F.; Narasipura, S.D.; Wallace, J.; Ritz, E.M.; Al-Harthi, L. Negative regulation of IL-8 in human astrocytes depends on β-catenin while positive regulation is mediated by TCFs/LEF/ATF2 interaction. Cytokine 2020, 136, 155252. [Google Scholar] [CrossRef]
  132. Kim, Y.; Park, J.; Choi, Y.K. The Role of Astrocytes in the Central Nervous System Focused on BK Channel and Heme Oxygenase Metabolites: A Review. Antioxidants 2019, 8, 121. [Google Scholar] [CrossRef] [PubMed]
  133. Siracusa, R.; Fusco, R.; Cuzzocrea, S. Astrocytes: Role and Functions in Brain Pathologies. Front. Pharmacol. 2019, 10, 1114. [Google Scholar] [CrossRef] [PubMed]
  134. Puma, C.; Danik, M.; Quirion, R.; Ramon, F.; Williams, S. The chemokine interleukin-8 acutely reduces Ca(2+) currents in identified cholinergic septal neurons expressing CXCR1 and CXCR2 receptor mRNAs. J. Neurochem. 2001, 78, 960–971. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, J.; Luo, W.; Huang, P.; Peng, L.; Huang, Q. Maternal C-reactive protein and cytokine levels during pregnancy and the risk of selected neuropsychiatric disorders in offspring: A systematic review and meta-analysis. J. Psychiatr. Res. 2018, 105, 86–94. [Google Scholar] [CrossRef] [PubMed]
  136. Bettcher, B.M.; Johnson, S.C.; Fitch, R.; Casaletto, K.B.; Heffernan, K.S.; Asthana, S.; Zetterberg, H.; Blennow, K.; Carlsson, C.M.; Neuhaus, J.; et al. Cerebrospinal Fluid and Plasma Levels of Inflammation Differentially Relate to CNS Markers of Alzheimer’s Disease Pathology and Neuronal Damage. J. Alzheimer’s Dis. JAD 2018, 62, 385–397. [Google Scholar] [CrossRef] [PubMed]
  137. Du, S.H.; Zhang, W.; Yue, X.; Luo, X.Q.; Tan, X.H.; Liu, C.; Qiao, D.F.; Wang, H. Role of CXCR1 and Interleukin-8 in Methamphetamine-Induced Neuronal Apoptosis. Front. Cell. Neurosci. 2018, 12, 230. [Google Scholar] [CrossRef]
  138. Lind, A.; Boraxbekk, C.J.; Petersen, E.T.; Paulson, O.B.; Andersen, O.; Siebner, H.R.; Marsman, A. Do glia provide the link between low-grade systemic inflammation and normal cognitive ageing? A 1 H magnetic resonance spectroscopy study at 7 tesla. J. Neurochem. 2021, 159, 185–196. [Google Scholar] [CrossRef]
  139. Thirumangalakudi, L.; Yin, L.; Rao, H.V.; Grammas, P. IL-8 induces expression of matrix metalloproteinases, cell cycle and pro-apoptotic proteins, and cell death in cultured neurons. J. Alzheimer’s Dis. JAD 2007, 11, 305–311. [Google Scholar] [CrossRef]
  140. Main, B.S.; Minter, M.R. Microbial Immuno-Communication in Neurodegenerative Diseases. Front. Neurosci. 2017, 11, 151. [Google Scholar] [CrossRef]
  141. Zhang, S.; Gan, L.; Cao, F.; Wang, H.; Gong, P.; Ma, C.; Ren, L.; Lin, Y.; Lin, X. The barrier and interface mechanisms of the brain barrier, and brain drug delivery. Brain Res. Bull. 2022, 190, 69–83. [Google Scholar] [CrossRef]
  142. Lippmann, E.S.; Azarin, S.M.; Kay, J.E.; Nessler, R.A.; Wilson, H.K.; Al-Ahmad, A.; Palecek, S.P.; Shusta, E.V. Derivation of blood-brain barrier endothelial cells from human pluripotent stem cells. Nat. Biotechnol. 2012, 30, 783–791. [Google Scholar] [CrossRef] [PubMed]
  143. Kadry, H.; Noorani, B.; Cucullo, L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef] [PubMed]
  144. Khan, M.S.; Mohapatra, S.; Gupta, V.; Ali, A.; Naseef, P.P.; Kurunian, M.S.; Alshadidi AA, F.; Alam, M.S.; Mirza, M.A.; Iqbal, Z. Potential of Lipid-Based Nanocarriers against Two Major Barriers to Drug Delivery-Skin and Blood-Brain Barrier. Membranes 2023, 13, 343. [Google Scholar] [CrossRef]
  145. Yuan, Y.; Sun, J.; Dong, Q.; Cui, M. Blood-brain barrier endothelial cells in neurodegenerative diseases: Sig-nals from the “barrier”. Front. Neurosci. 2023, 17, 1047778. [Google Scholar] [CrossRef]
  146. Xingi, E.; Koutsoudaki, P.N.; Thanou, I.; Phan, M.S.; Margariti, M.; Scheller, A.; Tinevez, J.Y.; Kirchhoff, F.; Tho-maidou, D. LPS-Induced Systemic Inflammation Affects the Dynamic Interactions of Astrocytes and Microglia with the Vasculature of the Mouse Brain Cortex. Cells 2023, 12, 1418. [Google Scholar] [CrossRef]
  147. Wojcieszak, J.; Kuczyńska, K.; Zawilska, J.B. Role of Chemokines in the Development and Progression of Alzheimer’s Disease. J. Mol. Neurosci. MN 2022, 72, 1929–1951. [Google Scholar] [CrossRef]
  148. Taub, D.D.; Anver, M.; Oppenheim, J.J.; Longo, D.L.; Murphy, W.J. T lymphocyte recruitment by interleukin-8 (IL-8). IL-8-induced degranulation of neutrophils releases potent chemoattractants for human T lymphocytes both in vitro and in vivo. J. Clin. Investig. 1996, 97, 1931–1941. [Google Scholar] [CrossRef] [PubMed]
  149. Kostulas, N.; Kivisäkk, P.; Huang, Y.; Matusevicius, D.; Kostulas, V.; Link, H. Ischemic stroke is associated with a systemic increase of blood mononuclear cells expressing interleukin-8 mRNA. Stroke 1998, 29, 462–466. [Google Scholar] [CrossRef]
  150. Domac, F.M.; Misirli, H. The role of neutrophils and interleukin-8 in acute ischemic stroke. Neurosciences 2008, 13, 136–141. [Google Scholar]
  151. Shaheen, H.A.; Daker, L.I.; Abbass, M.M.; Abd El Fattah, A.A. The relationship between the severity of disability and serum IL-8 in acute ischemic stroke patients. Egypt. J. Neurol. Psychiatry Neurosurg. 2018, 54, 26. [Google Scholar] [CrossRef]
  152. Kompaníková, P.; Bryja, V. Regulation of choroid plexus development and its functions. Cell. Mol. Life Sci. CMLS 2022, 79, 304. [Google Scholar] [CrossRef] [PubMed]
  153. Meeker, R.B.; Williams, K.; Killebrew, D.A.; Hudson, L.C. Cell trafficking through the choroid plexus. Cell Adhes. Migr. 2012, 6, 390–396. [Google Scholar] [CrossRef] [PubMed]
  154. Turner, C.A.; Thompson, R.C.; Bunney, W.E.; Schatzberg, A.F.; Barchas, J.D.; Myers, R.M.; Akil, H.; Watson, S.J. Altered choroid plexus gene expression in major depressive disorder. Front. Hum. Neurosci. 2014, 8, 238. [Google Scholar] [CrossRef] [PubMed]
  155. Kaur, C.; Rathnasamy, G.; Ling, E.A. The Choroid Plexus in Healthy and Diseased Brain. J. Neuropathol. Exp. Neurol. 2016, 75, 198–213. [Google Scholar] [CrossRef] [PubMed]
  156. Thompson, D.; Brissette, C.A.; Watt, J.A. The choroid plexus and its role in the pathogenesis of neurological infections. Fluids Barriers CNS 2022, 19, 75. [Google Scholar] [CrossRef] [PubMed]
  157. Lazarevic, I.; Soldati, S.; Mapunda, J.A.; Rudolph, H.; Rosito, M.; de Oliveira, A.C.; Enzmann, G.; Nishihara, H.; Ishikawa, H.; Tenenbaum, T.; et al. The choroid plexus acts as an immune cell reservoir and brain entry site in experimental autoimmune encephalomyelitis. Fluids Barriers CNS 2023, 20, 39. [Google Scholar] [CrossRef]
  158. Tenenbaum, T.; Steinmann, U.; Friedrich, C.; Berger, J.; Schwerk, C.; Schroten, H. Culture models to study leukocyte trafficking across the choroid plexus. Fluids Barriers CNS 2013, 10, 1. [Google Scholar] [CrossRef] [PubMed]
  159. Yu, H.; Huang, X.; Ma, Y.; Gao, M.; Wang, O.; Gao, T.; Shen, Y.; Liu, X. Interleukin-8 regulates endothelial permeability by down-regulation of tight junction but not dependent on integrins induced focal adhesions. Int. J. Biol. Sci. 2013, 9, 966–979. [Google Scholar] [CrossRef]
  160. Luissint, A.C.; Artus, C.; Glacial, F.; Ganeshamoorthy, K.; Couraud, P.O. Tight junctions at the blood brain barrier: Physiological architecture and disease-associated dysregulation. Fluids Barriers CNS 2012, 9, 23. [Google Scholar] [CrossRef]
  161. Wolburg, H.; Paulus, W. Choroid plexus: Biology and pathology. Acta Neuropathol. 2010, 119, 75–88. [Google Scholar] [CrossRef]
  162. Solár, P.; Zamani, A.; Kubíčková, L.; Dubový, P.; Joukal, M. Choroid plexus and the blood-cerebrospinal fluid barrier in disease. Fluids Barriers CNS 2020, 17, 35. [Google Scholar] [CrossRef] [PubMed]
  163. Assogna, M.; Premi, E.; Gazzina, S.; Benussi, A.; Ashton, N.J.; Zetterberg, H.; Blennow, K.; Gasparotti, R.; Padovani, A.; Tadayon, E.; et al. Association of Choroid Plexus Volume With Serum Biomarkers, Clinical Features, and Disease Severity in Patients With Frontotemporal Lobar Degeneration Spectrum. Neurology 2023, 101, e1218–e1230. [Google Scholar] [CrossRef] [PubMed]
  164. Zhou, Y.F.; Huang, J.C.; Zhang, P.; Fan, F.M.; Chen, S.; Fan, H.Z.; Cui, Y.M.; Luo, X.G.; Tan, S.P.; Wang, Z.R.; et al. Choroid Plexus Enlargement and Allostatic Load in Schizophrenia. Schizophr. Bull. 2020, 46, 722–731. [Google Scholar] [CrossRef] [PubMed]
  165. Cao, Y.; Lizano, P.; Deng, G.; Sun, H.; Zhou, X.; Xie, H.; Zhan, Y.; Mu, J.; Long, X.; Xiao, H.; et al. Brain-derived subgroups of bipolar II depression associate with inflammation and choroid plexus morphology. Psychiatry Clin. Neurosci. 2023, 77, 613–621. [Google Scholar] [CrossRef] [PubMed]
  166. Kaur, J.; Fahmy, L.M.; Davoodi-Bojd, E.; Zhang, L.; Ding, G.; Hu, J.; Zhang, Z.; Chopp, M.; Jiang, Q. Waste Clearance in the Brain. Front. Neuroanat. 2021, 15, 665803. [Google Scholar] [CrossRef] [PubMed]
  167. Lun, M.P.; Monuki, E.S.; Lehtinen, M.K. Development and functions of the choroid plexus-cerebrospinal fluid system. Nature reviews. Neuroscience 2015, 16, 445–457. [Google Scholar] [CrossRef] [PubMed]
  168. Kothur, K.; Wienholt, L.; Brilot, F.; Dale, R.C. CSF cytokines/chemokines as biomarkers in neuroinflammatory CNS disorders: A systematic review. Cytokine 2016, 77, 227–237. [Google Scholar] [CrossRef] [PubMed]
  169. Halaris, A. Neuroinflammation and neurotoxicity contribute to neuroprogression in neurological and psychiatric disorders. Future Neurol. 2018, 13, 59–69. [Google Scholar] [CrossRef]
  170. Guartazaca-Guerrero, S.; Rodríguez-Morales, J.; Rizo-Téllez, S.A.; Solleiro-Villavicencio, H.; Hernández-Valencia, A.F.; Carrillo-Ruiz, J.D.; Escobedo, G.; Méndez-García, L.A. High Levels of IL-8 and MCP-1 in Cerebrospinal Fluid of COVID-19 Patients with Cerebrovascular Disease. Exp. Neurobiol. 2021, 30, 256–261. [Google Scholar] [CrossRef] [PubMed]
  171. Devlin, L.; Gombolay, G.Y. Cerebrospinal fluid cytokines in COVID-19: A review and meta-analysis. J. Neurol. 2023, 270, 5155–5161. [Google Scholar] [CrossRef]
  172. Fu, Y.W.; Xu, H.S.; Liu, S.J. COVID-19 and neurodegenerative diseases. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 4535–4544. [Google Scholar] [CrossRef]
  173. Dey, R.; Bishayi, B. Microglial Inflammatory Responses to SARS-CoV-2 Infection: A Comprehensive Review. Cell. Mol. Neurobiol. 2023, 44, 2. [Google Scholar] [CrossRef]
  174. Kudo, T.; Hayashi, Y.; Kunieda, K.; Yoshikura, N.; Kimura, A.; Otsuki, M.; Shimohata, T. Persistent intrathecal interleukin-8 production in a patient with SARS-CoV-2-related encephalopathy presenting aphasia: A case report. BMC Neurol. 2021, 21, 426. [Google Scholar] [CrossRef]
  175. Wang, A.K.; Miller, B.J. Meta-analysis of Cerebrospinal Fluid Cytokine and Tryptophan Catabolite Alterations in Psychiatric Patients: Comparisons Between Schizophrenia, Bipolar Disorder, and Depression. Schizophr. Bull. 2018, 44, 75–83. [Google Scholar] [CrossRef]
  176. Gallego, J.A.; Blanco, E.A.; Husain-Krautter, S.; Madeline Fagen, E.; Moreno-Merino, P.; Del Ojo-Jiménez, J.A.; Ahmed, A.; Rothstein, T.L.; Lencz, T.; Malhotra, A.K. Cytokines in cerebrospinal fluid of patients with schizophrenia spectrum disorders: New data and an updated meta-analysis. Schizophr. Res. 2018, 202, 64–71. [Google Scholar] [CrossRef]
  177. Warren, N.; O’Gorman, C.; Horgan, I.; Weeratunga, M.; Halstead, S.; Moussiopoulou, J.; Campana, M.; Yakimov, V.; Wagner, E.; Siskind, D. Inflammatory cerebrospinal fluid markers in schizophrenia spectrum disorders: A systematic review and meta-analysis of 69 studies with 5710 participants. Schizophr. Res. 2024, 266, 24–31. [Google Scholar] [CrossRef]
  178. Isgren, A.; Jakobsson, J.; Pålsson, E.; Ekman, C.J.; Johansson, A.G.; Sellgren, C.; Blennow, K.; Zetterberg, H.; Landén, M. Increased cerebrospinal fluid interleukin-8 in bipolar disorder patients associated with lithium and antipsychotic treatment. Brain Behav. Immun. 2015, 43, 198–204. [Google Scholar] [CrossRef]
  179. Runge, K.; Fiebich, B.L.; Kuzior, H.; Rausch, J.; Maier, S.J.; Dersch, R.; Nickel, K.; Domschke, K.; Tebartz van Elst, L.; Endres, D. Altered cytokine levels in the cerebrospinal fluid of adult patients with autism spectrum disorder. J. Psychiatr. Res. 2023, 158, 134–142. [Google Scholar] [CrossRef]
  180. Janelidze, S.; Lindqvist, D.; Francardo, V.; Hall, S.; Zetterberg, H.; Blennow, K.; Adler, C.H.; Beach, T.G.; Serrano, G.E.; van Westen, D.; et al. Increased CSF biomarkers of angiogenesis in Parkinson disease. Neurology 2015, 85, 1834–1842. [Google Scholar] [CrossRef]
  181. Matejčíková, Z.; Mareš, J.; Sládková, V.; Svrčinová, T.; Vysloužilová, J.; Zapletalová, J.; Kaňovský, P. Cerebrospinal fluid and serum levels of interleukin-8 in patients with multiple sclerosis and its correlation with Q-albumin. Mult. Scler. Relat. Disord. 2017, 14, 12–15. [Google Scholar] [CrossRef]
  182. Stampanoni Bassi, M.; Iezzi, E.; Landi, D.; Monteleone, F.; Gilio, L.; Simonelli, I.; Musella, A.; Mandolesi, G.; De Vito, F.; Furlan, R.; et al. Delayed treatment of MS is associated with high CSF levels of IL-6 and IL-8 and worse future disease course. J. Neurol. 2018, 265, 2540–2547. [Google Scholar] [CrossRef]
  183. Isung, J.; Aeinehband, S.; Mobarrez, F.; Mårtensson, B.; Nordström, P.; Asberg, M.; Piehl, F.; Jokinen, J. Low vascular endothelial growth factor and interleukin-8 in cerebrospinal fluid of suicide attempters. Transl. Psychiatry 2012, 2, e196. [Google Scholar] [CrossRef]
  184. Janelidze, S.; Suchankova, P.; Ekman, A.; Erhardt, S.; Sellgren, C.; Samuelsson, M.; Westrin, A.; Minthon, L.; Hansson, O.; Träskman-Bendz, L.; et al. Low IL-8 is associated with anxiety in suicidal patients: Genetic variation and decreased protein levels. Acta Psychiatr. Scand. 2015, 131, 269–278. [Google Scholar] [CrossRef]
  185. Zhang, M.; Fang, T.; Wang, K.; Mei, H.; Lv, Z.; Wang, F.; Cai, Z.; Liang, C. Association of polymorphisms in interleukin-8 gene with cancer risk: A meta-analysis of 22 case-control studies. OncoTargets Ther. 2016, 9, 3727–3737. [Google Scholar] [CrossRef]
  186. Ben Afia, A.; Aflouk, Y.; Saoud, H.; Zaafrane, F.; Gaha, L.; Bel Hadj Jrad, B. Inteurleukin-8 gene variations and the susceptibility to schizophrenia. Psychiatry Res. 2020, 293, 113421. [Google Scholar] [CrossRef]
  187. Jundi, K.; Greene, C.M. Transcription of Interleukin-8: How Altered Regulation Can Affect Cystic Fibrosis Lung Disease. Biomolecules 2015, 5, 1386–1398. [Google Scholar] [CrossRef]
  188. Liu, Q.; Li, A.; Tian, Y.; Wu, J.D.; Liu, Y.; Li, T.; Chen, Y.; Han, X.; Wu, K. The CXCL8-CXCR1/2 pathways in cancer. Cytokine Growth Factor Rev. 2016, 31, 61–71. [Google Scholar] [CrossRef]
  189. Benakanakere, M.R.; Finoti, L.S.; Tanaka, U.; Grant, G.R.; Scarel-Caminaga, R.M.; Kinane, D.F. Investigation of the functional role of human Interleukin-8 gene haplotypes by CRISPR/Cas9 mediated genome editing. Sci. Rep. 2016, 6, 31180. [Google Scholar] [CrossRef]
  190. Wacharasint, P.; Nakada, T.A.; Boyd, J.H.; Russell, J.A.; Walley, K.R. AA genotype of IL-8 -251A/T is associated with low PaO(2)/FiO(2) in critically ill patients and with increased IL-8 expression. Respirology 2012, 17, 1253–1260. [Google Scholar] [CrossRef]
  191. Fujihara, J.; Shiwaku, K.; Yasuda, T.; Yuasa, I.; Nishimukai, H.; Iida, R.; Takeshita, H. Variation of interleukin 8 -251 A>T polymorphism in worldwide populations and intra-ethnic differences in Japanese populations. Clin. Chim. Acta Int. J. Clin. Chem. 2007, 377, 79–82. [Google Scholar] [CrossRef] [PubMed]
  192. Wu, Y.; Wang, W.; Li, X.Y.; Qian, L.L.; Dang, S.P.; Tang, X.; Chen, H.J.; Wang, R.X. Strong association between the interleukin-8-251A/T polymorphism and coronary artery disease risk. Medicine 2019, 98, e14715. [Google Scholar] [CrossRef]
  193. Hull, J.; Thomson, A.; Kwiatkowski, D. Association of respiratory syncytial virus bronchiolitis with the interleukin 8 gene region in UK families. Thorax 2000, 55, 1023–1027. [Google Scholar] [CrossRef]
  194. Kang, H.J.; Kim, J.M.; Kim, S.W.; Shin, I.S.; Park, S.W.; Kim, Y.H.; Yoon, J.S. Associations of cytokine genes with Alzheimer’s disease and depression in an elderly Korean population. J. Neurol. Neurosurg. Psychiatry 2015, 86, 1002–1007. [Google Scholar] [CrossRef]
  195. Infante, J.; Sanz, C.; Fernández-Luna, J.L.; Llorca, J.; Berciano, J.; Combarros, O. Gene-gene interaction between interleukin-1A and interleukin-8 increases Alzheimer’s disease risk. J. Neurol. 2004, 251, 482–483. [Google Scholar] [CrossRef]
  196. Vendramini, A.A.; de Lábio, R.W.; Rasmussen, L.T.; Minett, T.; Bertolucci, P.H.; de Arruda Cardoso Smith, M.; Payão, S.L. Interleukin-8 gene polymorphism -251T>A and Alzheimer’s disease. J. Alzheimer’s Dis. JAD 2007, 12, 221–222. [Google Scholar] [CrossRef]
  197. Vendramini, A.A.; de Lábio, R.W.; Rasmussen, L.T.; Dos Reis, N.M.; Minett, T.; Bertolucci, P.H.; de Souza Pinhel, M.A.; Souza, D.R.; Mazzotti, D.R.; de Arruda Cardoso Smith, M.; et al. Interleukin-8-251T > A, Interleukin-1α-889C > T and Apolipoprotein E polymorphisms in Alzheimer’s disease. Genet. Mol. Biol. 2011, 34, 1–5. [Google Scholar] [CrossRef] [PubMed]
  198. Zhang, Q.; Yang, K.; Zhang, Z.; Zhang, R. Associations between Certain Polymorphisms in Proinflammatory Cytokines and Predisposition of Alzheimer’s Disease: A Meta-Analysis. Dement. Geriatr. Cogn. Disord. 2021, 50, 224–230. [Google Scholar] [CrossRef]
  199. Yang, R.; Duan, J.; Luo, F.; Tao, P.; Hu, C. IL-6, IL-8 and IL-10 polymorphisms may impact predisposition of Alzheimer’s disease: A meta-analysis. Acta Neurol. Belg. 2021, 121, 1505–1512. [Google Scholar] [CrossRef]
  200. Kamali-Sarvestani, E.; Nikseresht, A.R.; Aliparasti, M.R.; Vessal, M. IL-8 (-251 A/T) and CXCR2 (+1208 C/T) gene polymorphisms and risk of multiple sclerosis in Iranian patients. Neurosci. Lett. 2006, 404, 159–162. [Google Scholar] [CrossRef]
  201. Dolcetti, E.; Bruno, A.; Azzolini, F.; Gilio, L.; Pavone, L.; Iezzi, E.; Galifi, G.; Gambardella, S.; Ferese, R.; Buttari, F.; et al. Genetic regulation of IL-8 influences disease presentation of multiple sclerosis. Mult. Scler. 2023, 29, 512–520. [Google Scholar] [CrossRef]
  202. Kahaei, M.S.; Ghafouri-Fard, S.; Namvar, A.; Omrani, M.D.; Sayad, A.; Taheri, M. Associations between an intronic variant in IL-8 gene and risk of psychiatric disorders. Ecol. Genet. Genom. 2020, 14, 100050. [Google Scholar] [CrossRef]
  203. Noroozi, R.; Omrani, M.D.; Ayatollahi, S.A.; Sayad, A.; Ata, A.; Fallah, H.; Taheri, M.; Ghafouri-Fard, S. Interleukin (IL)-8 polymorphisms contribute in suicide behavior. Cytokine 2018, 111, 28–32. [Google Scholar] [CrossRef] [PubMed]
  204. Reyes-Gibby, C.C.; Wang, J.; Spitz, M.; Wu, X.; Yennurajalingam, S.; Shete, S. Genetic variations in interleukin-8 and interleukin-10 are associated with pain, depressed mood, and fatigue in lung cancer patients. J. Pain Symptom Manag. 2013, 46, 161–172. [Google Scholar] [CrossRef] [PubMed]
  205. Kim, J.M.; Stewart, R.; Kim, S.W.; Kim, S.Y.; Bae, K.Y.; Kang, H.J.; Jang, J.E.; Shin, I.S.; Yoon, J.S. Physical health and incident late-life depression: Modification by cytokine genes. Neurobiol. Aging 2013, 34, 356.e1–356.e9. [Google Scholar] [CrossRef] [PubMed]
  206. Kim, J.M.; Stewart, R.; Kim, S.W.; Shin, I.S.; Kim, J.T.; Park, M.S.; Park, S.W.; Kim, Y.H.; Cho, K.H.; Yoon, J.S. Associations of cytokine gene polymorphisms with post-stroke depression. World J. Biol. Psychiatry Off. J. World Fed. Soc. Biol. Psychiatry 2012, 13, 579–587. [Google Scholar] [CrossRef] [PubMed]
  207. Kim, J.M.; Stewart, R.; Kim, S.Y.; Kang, H.J.; Jang, J.E.; Kim, S.W.; Shin, I.S.; Park, M.H.; Yoon, J.H.; Park, S.W.; et al. A one year longitudinal study of cytokine genes and depression in breast cancer. J. Affect. Disord. 2013, 148, 57–65. [Google Scholar] [CrossRef] [PubMed]
  208. Kelly, R.S.; Lee-Sarwar, K.; Chen, Y.C.; Laranjo, N.; Fichorova, R.; Chu, S.H.; Prince, N.; Lasky-Su, J.; Weiss, S.T.; Litonjua, A.A. Maternal Inflammatory Biomarkers during Pregnancy and Early Life Neurodevelopment in Offspring: Results from the VDAART Study. Int. J. Mol. Sci. 2022, 23, 15249. [Google Scholar] [CrossRef] [PubMed]
  209. Gillespie, S.L.; Anderson, C.M. Racial discrimination and leukocyte glucocorticoid sensitivity: Implications for birth timing. Soc. Sci. Med. 2018, 216, 114–123. [Google Scholar] [CrossRef] [PubMed]
  210. Christian, L.M.; Porter, K. Longitudinal changes in serum proinflammatory markers across pregnancy and postpartum: Effects of maternal body mass index. Cytokine 2014, 70, 134–140. [Google Scholar] [CrossRef]
  211. Nelson, P.G.; Kuddo, T.; Song, E.Y.; Dambrosia, J.M.; Kohler, S.; Satyanarayana, G.; Vandunk, C.; Grether, J.K.; Nelson, K.B. Selected neurotrophins, neuropeptides, and cytokines: Developmental trajectory and concentrations in neonatal blood of children with autism or Down syndrome. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 2006, 24, 73–80. [Google Scholar] [CrossRef]
  212. Lim, J.; Sohn, H.; Kwon, M.S.; Kim, B. White Matter Alterations Associated with Pro-inflammatory Cytokines in Patients with Major Depressive Disorder. Clin. Psychopharmacol. Neurosci. Off. Sci. J. Korean Coll. Neuropsychopharmacol. 2021, 19, 449–458. [Google Scholar] [CrossRef] [PubMed]
  213. Dozmorov, M.G.; Bilbo, S.D.; Kollins, S.H.; Zucker, N.; Do, E.K.; Schechter, J.C.; Zhang, J.J.; Murphy, S.K.; Hoyo, C.; Fuemmeler, B.F. Associations between maternal cytokine levels during gestation and measures of child cognitive abilities and executive functioning. Brain Behav. Immun. 2018, 70, 390–397. [Google Scholar] [CrossRef]
  214. Ellman, L.M.; Deicken, R.F.; Vinogradov, S.; Kremen, W.S.; Poole, J.H.; Kern, D.M.; Tsai, W.Y.; Schaefer, C.A.; Brown, A.S. Structural brain alterations in schizophrenia following fetal exposure to the inflammatory cytokine interleukin-8. Schizophr. Res. 2010, 121, 46–54. [Google Scholar] [CrossRef] [PubMed]
  215. Sullivan, G.; Galdi, P.; Cabez, M.B.; Borbye-Lorenzen, N.; Stoye, D.Q.; Lamb, G.J.; Evans, M.J.; Quigley, A.J.; Thrippleton, M.J.; Skogstrand, K.; et al. Interleukin-8 dysregulation is implicated in brain dysmaturation following preterm birth. Brain Behav. Immun. 2020, 90, 311–318. [Google Scholar] [CrossRef]
  216. Jones, K.L.; Croen, L.A.; Yoshida, C.K.; Heuer, L.; Hansen, R.; Zerbo, O.; DeLorenze, G.N.; Kharrazi, M.; Yolken, R.; Ashwood, P.; et al. Autism with intellectual disability is associated with increased levels of maternal cytokines and chemokines during gestation. Mol. Psychiatry 2017, 22, 273–279. [Google Scholar] [CrossRef]
  217. Franciosi, S.; Choi, H.B.; Kim, S.U.; McLarnon, J.G. IL-8 enhancement of amyloid-beta (Abeta 1-42)-induced expression and production of pro-inflammatory cytokines and COX-2 in cultured human microglia. J. Neuroimmunol. 2005, 159, 66–74. [Google Scholar] [CrossRef]
  218. Brown, A.S.; Hooton, J.; Schaefer, C.A.; Zhang, H.; Petkova, E.; Babulas, V.; Perrin, M.; Gorman, J.M.; Susser, E.S. Elevated maternal interleukin-8 levels and risk of schizophrenia in adult offspring. Am. J. Psychiatry 2004, 161, 889–895. [Google Scholar] [CrossRef] [PubMed]
  219. Osborne, S.; Biaggi, A.; Hazelgrove, K.; Preez, A.D.; Nikkheslat, N.; Sethna, V.; Zunszain, P.A.; Conroy, S.; Pawlby, S.; Pariante, C.M. Increased maternal inflammation and poorer infant neurobehavioural competencies in women with a history of major depressive disorder from the psychiatry research and motherhood-Depression (PRAM-D) study. Brain Behav. Immun. 2022, 99, 223–230. [Google Scholar] [CrossRef]
  220. Mac Giollabhui, N.; Breen, E.C.; Murphy, S.K.; Maxwell, S.D.; Cohn, B.A.; Krigbaum, N.Y.; Cirillo, P.M.; Perez, C.; Alloy, L.B.; Drabick DA, G.; et al. Maternal inflammation during pregnancy and offspring psychiatric symptoms in childhood: Timing and sex matter. J. Psychiatr. Res. 2019, 111, 96–103. [Google Scholar] [CrossRef]
  221. Ghassabian, A.; Albert, P.S.; Hornig, M.; Yeung, E.; Cherkerzian, S.; Goldstein, R.B.; Buka, S.L.; Goldstein, J.M.; Gilman, S.E. Gestational cytokine concentrations and neurocognitive development at 7 years. Transl. Psychiatry 2018, 8, 64. [Google Scholar] [CrossRef]
  222. Yu, J.; Ghassabian, A.; Chen, Z.; Goldstein, R.B.; Hornig, M.; Buka, S.L.; Goldstein, J.M.; Gilman, S.E. Maternal Immune activity during pregnancy and socioeconomic disparities in children’s self-regulation. Brain Behav. Immun. 2020, 90, 346–352. [Google Scholar] [CrossRef] [PubMed]
  223. Gilman, S.E.; Hornig, M.; Ghassabian, A.; Hahn, J.; Cherkerzian, S.; Albert, P.S.; Buka, S.L.; Goldstein, J.M. Socioeconomic disadvantage, gestational immune activity, and neurodevelopment in early childhood. Proc. Natl. Acad. Sci. USA 2017, 114, 6728–6733. [Google Scholar] [CrossRef] [PubMed]
  224. Brunoni, A.R.; Supasitthumrong, T.; Teixeira, A.L.; Vieira, E.L.; Gattaz, W.F.; Benseñor, I.M.; Lotufo, P.A.; Lafer, B.; Berk, M.; Carvalho, A.F.; et al. Differences in the immune-inflammatory profiles of unipolar and bipolar depression. J. Affect. Disord. 2020, 262, 8–15. [Google Scholar] [CrossRef] [PubMed]
  225. Maes, M.; Nani, J.V.; Noto, C.; Rizzo, L.; Hayashi MA, F.; Brietzke, E. Impairments in Peripheral Blood T Effector and T Regulatory Lymphocytes in Bipolar Disorder Are Associated with Staging of Illness and Anti-cytomegalovirus IgG Levels. Mol. Neurobiol. 2021, 58, 229–242. [Google Scholar] [CrossRef] [PubMed]
  226. Maes, M.; Rachayon, M.; Jirakran, K.; Sodsai, P.; Klinchanhom, S.; Gałecki, P.; Sughondhabirom, A.; Basta-Kaim, A. The Immune Profile of Major Dysmood Disorder: Proof of Concept and Mechanism Using the Precision Nomothetic Psychiatry Approach. Cells 2022, 11, 1183. [Google Scholar] [CrossRef] [PubMed]
  227. Wieck, A.; Grassi-Oliveira, R.; do Prado, C.H.; Viola, T.W.; Petersen, L.E.; Porto, B.; Teixeira, A.L.; Bauer, M.E. Toll-like receptor expression and function in type I bipolar disorder. Brain Behav. Immun. 2016, 54, 110–121. [Google Scholar] [CrossRef] [PubMed]
  228. Tang, G.; Chen, P.; Chen, G.; Zhong, S.; Gong, J.; Zhong, H.; Ye, T.; Chen, F.; Wang, J.; Luo, Z.; et al. Inflammation is correlated with abnormal functional connectivity in unmedicated bipolar depression: An independent component analysis study of resting-state fMRI. Psychol. Med. 2021, 52, 3431–3441. [Google Scholar] [CrossRef] [PubMed]
  229. Isgren, A.; Sellgren, C.; Ekman, C.J.; Holmén-Larsson, J.; Blennow, K.; Zetterberg, H.; Jakobsson, J.; Landén, M. Markers of neuroinflammation and neuronal injury in bipolar disorder: Relation to prospective clinical outcomes. Brain Behav. Immun. 2017, 65, 195–201. [Google Scholar] [CrossRef] [PubMed]
  230. Wang, T.Y.; Lee, S.Y.; Chen, S.L.; Chung, Y.L.; Li, C.L.; Chang, Y.H.; Wang, L.J.; Chen, P.S.; Chen, S.H.; Chu, C.H.; et al. The Differential Levels of Inflammatory Cytokines and BDNF among Bipolar Spectrum Disorders. Int. J. Neuropsychopharmacol. 2016, 19, pyw012. [Google Scholar] [CrossRef]
  231. Misiak, B.; Bartoli, F.; Carrà, G.; Małecka, M.; Samochowiec, J.; Jarosz, K.; Banik, A.; Stańczykiewicz, B. Chemokine alterations in bipolar disorder: A systematic review and meta-analysis. Brain Behav. Immun. 2020, 88, 870–877. [Google Scholar] [CrossRef]
  232. Lu, Y.R.; Rao, Y.B.; Mou, Y.J.; Chen, Y.; Lou, H.F.; Zhang, Y.; Zhang, D.X.; Xie, H.Y.; Hu, L.W.; Fang, P. High concentrations of serum interleukin-6 and interleukin-8 in patients with bipolar disorder. Medicine 2019, 98, e14419. [Google Scholar] [CrossRef]
  233. Irwin, M.R.; Olmstead, R.; Kruse, J.; Breen, E.C.; Haque, R. Association of interleukin-8 and risk of incident and recurrent depression in long-term breast cancer survivors. Brain Behav. Immun. 2022, 105, 131–138. [Google Scholar] [CrossRef] [PubMed]
  234. Barbosa, I.G.; Rocha, N.P.; Bauer, M.E.; de Miranda, A.S.; Huguet, R.B.; Reis, H.J.; Zunszain, P.A.; Horowitz, M.A.; Pariante, C.M.; Teixeira, A.L. Chemokines in bipolar disorder: Trait or state? Eur. Arch. Psychiatry Clin. Neurosci. 2013, 263, 159–165. [Google Scholar] [CrossRef]
  235. Munkholm, K.; Braüner, J.V.; Kessing, L.V.; Vinberg, M. Cytokines in bipolar disorder vs. healthy control subjects: A systematic review and meta-analysis. J. Psychiatr. Res. 2013, 47, 1119–1133. [Google Scholar] [CrossRef]
  236. Modabbernia, A.; Taslimi, S.; Brietzke, E.; Ashrafi, M. Cytokine alterations in bipolar disorder: A meta-analysis of 30 studies. Biol. Psychiatry 2013, 74, 15–25. [Google Scholar] [CrossRef] [PubMed]
  237. Tubbs, J.D.; Ding, J.; Baum, L.; Sham, P.C. Immune dysregulation in depression: Evidence from genome-wide association. Brain Behav. Immun. Health 2020, 7, 100108. [Google Scholar] [CrossRef]
  238. Kruse, J.L.; Boyle, C.C.; Olmstead, R.; Breen, E.C.; Tye, S.J.; Eisenberger, N.I.; Irwin, M.R. Interleukin-8 and depressive responses to an inflammatory challenge: Secondary analysis of a randomized controlled trial. Sci. Rep. 2022, 12, 12627. [Google Scholar] [CrossRef]
  239. Zhu, Z.H.; Song, X.Y.; Man, L.J.; Chen, P.; Tang, Z.; Li, R.H.; Ji, C.F.; Dai, N.B.; Liu, F.; Wang, J.; et al. Comparisons of Serum Interleukin-8 Levels in Major Depressive Patients With Drug-Free Versus SSRIs Versus Healthy Controls. Front. Psychiatry 2022, 13, 858675. [Google Scholar] [CrossRef] [PubMed]
  240. Liu, J.J.; Wei, Y.B.; Strawbridge, R.; Bao, Y.; Chang, S.; Shi, L.; Que, J.; Gadad, B.S.; Trivedi, M.H.; Kelsoe, J.R.; et al. Peripheral cytokine levels and response to antidepressant treatment in depression: A systematic review and meta-analysis. Mol. Psychiatry 2020, 25, 339–350. [Google Scholar] [CrossRef]
  241. Zou, W.; Feng, R.; Yang, Y. Changes in the serum levels of inflammatory cytokines in antidepressant drug-naïve patients with major depression. PLoS ONE 2018, 13, e0197267. [Google Scholar] [CrossRef]
  242. Cai, Y.; Zhu, Z.H.; Li, R.H.; Yin, X.Y.; Chen, R.F.; Man, L.J.; Hou, W.L.; Zhu, H.L.; Wang, J.; Zhang, H.; et al. Association between increased serum interleukin-8 levels and improved cognition in major depressive patients with SSRIs. BMC Psychiatry 2023, 23, 122. [Google Scholar] [CrossRef] [PubMed]
  243. Baune, B.T.; Smith, E.; Reppermund, S.; Air, T.; Samaras, K.; Lux, O.; Brodaty, H.; Sachdev, P.; Trollor, J.N. Inflammatory biomarkers predict depressive, but not anxiety symptoms during aging: The prospective Sydney Memory and Aging Study. Psychoneuroendocrinology 2012, 37, 1521–1530. [Google Scholar] [CrossRef] [PubMed]
  244. Kruse, J.L.; Olmstead, R.; Hellemann, G.; Breen, E.C.; Tye, S.J.; Brooks, J.O.; 3rd Wade, B.; Congdon, E.; Espinoza, R.; Narr, K.L.; et al. Interleukin-8 and lower severity of depression in females, but not males, with treatment-resistant depression. J. Psychiatr. Res. 2021, 140, 350–356. [Google Scholar] [CrossRef] [PubMed]
  245. Moriarity, D.P.; Giollabhui, N.M.; Ellman, L.M.; Klugman, J.; Coe, C.L.; Abramson, L.Y.; Alloy, L.B. Inflammatory Proteins Predict Change in Depressive Symptoms in Male and Female Adolescents. Clin. Psychol. Sci. A J. Assoc. Psychol. Sci. 2019, 7, 754–767. [Google Scholar] [CrossRef] [PubMed]
  246. Kruse, J.L.; Olmstead, R.; Hellemann, G.; Wade, B.; Jiang, J.; Vasavada, M.M.; Brooks Iii, J.O.; Congdon, E.; Espinoza, R.; Narr, K.L.; et al. Inflammation and depression treatment response to electroconvulsive therapy: Sex-specific role of interleukin-8. Brain Behav. Immun. 2020, 89, 59–66. [Google Scholar] [CrossRef]
  247. Kruse, J.L.; Vasavada, M.M.; Olmstead, R.; Hellemann, G.; Wade, B.; Breen, E.C.; Brooks, J.O.; Congdon, E.; Espinoza, R.; Narr, K.L.; et al. Depression treatment response to ketamine: Sex-specific role of interleukin-8, but not other inflammatory markers. Transl. Psychiatry 2021, 11, 167. [Google Scholar] [CrossRef]
  248. Perez, M.L.; Raison, C.L.; Coe, C.L.; Cook, D.B.; Meyer, J.D. Cytokine responses across submaximal exercise intensities in women with major depressive disorder. Brain Behav. Immun. Health 2020, 2, 100046. [Google Scholar] [CrossRef]
  249. Carvalho, L.A.; Bergink, V.; Sumaski, L.; Wijkhuijs, J.; Hoogendijk, W.J.; Birkenhager, T.K.; Drexhage, H.A. Inflammatory activation is associated with a reduced glucocorticoid receptor alpha/beta expression ratio in monocytes of inpatients with melancholic major depressive disorder. Transl. Psychiatry 2014, 4, e344. [Google Scholar] [CrossRef]
  250. Islam, S.; Islam, T.; Nahar, Z.; Shahriar, M.; Islam SM, A.; Bhuiyan, M.A.; Islam, M.R. Altered serum adiponectin and interleukin-8 levels are associated in the pathophysiology of major depressive disorder: A case-control study. PLoS ONE 2022, 17, e0276619. [Google Scholar] [CrossRef]
  251. Matits, L.; Munk, M.; Bizjak, D.A.; Kolassa, I.T.; Karrasch, S.; Vollrath, S.; Jerg, A.; Steinacker, J.M. Inflammation and severity of depressive symptoms in physically active individuals after COVID-19—An exploratory immunopsychological study investigating the effect of inflammation on depressive symptom severity. Brain Behav. Immun. Health 2023, 30, 100614. [Google Scholar] [CrossRef]
  252. Ogłodek, E. Changes in the Serum Levels of Cytokines: IL-1β, IL-4, IL-8 and IL-10 in Depression with and without Posttraumatic Stress Disorder. Brain Sci. 2022, 12, 387. [Google Scholar] [CrossRef] [PubMed]
  253. Suneson, K.; Grudet, C.; Ventorp, F.; Malm, J.; Asp, M.; Westrin, Å.; Lindqvist, D. An inflamed subtype of difficult-to-treat depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2023, 125, 110763. [Google Scholar] [CrossRef] [PubMed]
  254. Szałach, Ł.P.; Cubała, W.J.; Lisowska, K.A. Changes in T-Cell Subpopulations and Cytokine Levels in Patients with Treatment-Resistant Depression-A Preliminary Study. Int. J. Mol. Sci. 2022, 24, 479. [Google Scholar] [CrossRef] [PubMed]
  255. Grade, S.; Weng, Y.C.; Snapyan, M.; Kriz, J.; Malva, J.O.; Saghatelyan, A. Brain-derived neurotrophic factor promotes vasculature-associated migration of neuronal precursors toward the ischemic striatum. PLoS ONE 2013, 8, e55039. [Google Scholar] [CrossRef] [PubMed]
  256. Miranda, M.; Morici, J.F.; Zanoni, M.B.; Bekinschtein, P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front. Cell. Neurosci. 2019, 13, 363. [Google Scholar] [CrossRef] [PubMed]
  257. Halaris, A.; Sharma, A.; Meresh, E.; Pandey, G.; Kang, R.; Sinacore, J. Serum BDNF: A potential biomarker for major depressive disorder and antidepressant response prediction. J. Depress. Anxiety 2015, 4, 1000179. [Google Scholar] [CrossRef]
  258. Joshi, R.; Salton, S.R.J. Neurotrophin Crosstalk in the Etiology and Treatment of Neuropsychiatric and Neu-rodegenerative Disease. Front. Mol. Neurosci. 2022, 15, 932497. [Google Scholar] [CrossRef] [PubMed]
  259. Esvald, E.E.; Tuvikene, J.; Kiir, C.S.; Avarlaid, A.; Tamberg, L.; Sirp, A.; Shubina, A.; Cabrera-Cabrera, F.; Pihlak, A.; Koppel, I.; et al. Revisiting the expression of BDNF and its receptors in mammalian devel-opment. Front. Mol. Neurosci. 2023, 16, 1182499. [Google Scholar] [CrossRef] [PubMed]
  260. Shkundin, A.; Halaris, A. Associations of BDNF/BDNF-AS SNPs with Depression, Schizophrenia, and Bipolar Disorder. J. Pers. Med. 2023, 13, 1395. [Google Scholar] [CrossRef]
  261. Brooks, S.; Friedes, B.D.; Northington, F.; Graham, E.; Tekes, A.; Burton, V.J.; Gerner, G.; Zhu, J.; Chavez-Valdez, R.; Vaidya, D.; et al. Serum brain injury biomarkers are gestationally and post-natally regulated in non-brain injured neonates. Pediatr. Res. 2023, 93, 1943–1954. [Google Scholar] [CrossRef]
  262. Charlton, T.; Prowse, N.; McFee, A.; Heiratifar, N.; Fortin, T.; Paquette, C.; Hayley, S. Brain-derived neu-rotrophic factor (BDNF) has direct anti-inflammatory effects on microglia. Front. Cell. Neurosci. 2023, 17, 1188672. [Google Scholar] [CrossRef] [PubMed]
  263. Dietrick, B.; Molloy, E.; Massaro, A.N.; Strickland, T.; Zhu, J.; Slevin, M.; Donoghue, V.; Sweetman, D.; Kelly, L.; O’Dea, M.; et al. Plasma and Cerebrospinal Fluid Candidate Biomarkers of Neonatal Encephalopathy Severity and Neurodevelopmental Outcomes. J. Pediatr. 2020, 226, 71–79. [Google Scholar] [CrossRef] [PubMed]
  264. Zhang, X.Y.; Tan, Y.L.; Chen, D.C.; Tan, S.P.; Yang, F.D.; Wu, H.E.; Zunta-Soares, G.B.; Huang, X.F.; Kosten, T.R.; Soares, J.C. Interaction of BDNF with cytokines in chronic schizophrenia. Brain Behav. Immun. 2016, 51, 169–175. [Google Scholar] [CrossRef] [PubMed]
  265. Xiu, M.H.; Wang, D.M.; Du, X.D.; Chen, N.; Tan, S.P.; Tan, Y.L.; Yang, F.; Cho, R.Y.; Zhang, X.Y. Interaction of BDNF and cytokines in executive dysfunction in patients with chronic schizophrenia. Psychoneuroendocrinology 2019, 108, 110–117. [Google Scholar] [CrossRef] [PubMed]
  266. Wang, T.Y.; Lee, S.Y.; Chen, S.L.; Chang, Y.H.; Wang, L.J.; Chen, P.S.; Chen, S.H.; Chu, C.H.; Huang, S.Y.; Tzeng, N.S.; et al. Comparing clinical responses and the biomarkers of BDNF and cytokines between subthreshold bipolar disorder and bipolar II disorder. Sci. Rep. 2016, 6, 27431. [Google Scholar] [CrossRef] [PubMed]
  267. Liou, Y.J.; Wang, T.Y.; Lee, S.Y.; Chang, Y.H.; Tsai, T.Y.; Chen, P.S.; Huang, S.Y.; Tzeng, N.S.; Lee, I.H.; Chen, K.C.; et al. Effects of comorbid alcohol use disorder on bipolar disorder: Focusing on neurocognitive function and inflammatory markers. Psychoneuroendocrinology 2023, 152, 106083. [Google Scholar] [CrossRef] [PubMed]
  268. Roebuck, K.A. Oxidant stress regulation of IL-8 and ICAM-1 gene expression: Differential activation and binding of the transcription factors AP-1 and NF-kappaB (Review). Int. J. Mol. Med. 1999, 4, 223–230. [Google Scholar] [CrossRef] [PubMed]
  269. Auten, R.L.; Davis, J.M. Oxygen toxicity and reactive oxygen species: The devil is in the details. Pediatr. Res. 2009, 66, 121–127. [Google Scholar] [CrossRef] [PubMed]
  270. Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Et Biophys. Acta 2016, 1863, 2977–2992. [Google Scholar] [CrossRef]
  271. Zhang, J.; Wang, X.; Vikash, V.; Ye, Q.; Wu, D.; Liu, Y.; Dong, W. ROS and ROS-Mediated Cellular Signaling. Oxidative Med. Cell. Longev. 2016, 2016, 4350965. [Google Scholar] [CrossRef]
  272. Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [PubMed]
  273. Pandya, C.D.; Howell, K.R.; Pillai, A. Antioxidants as potential therapeutics for neuropsychiatric disorders. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2013, 46, 214–223. [Google Scholar] [CrossRef] [PubMed]
  274. Morris, G.; Walder, K.R.; Berk, M.; Marx, W.; Walker, A.J.; Maes, M.; Puri, B.K. The interplay between oxidative stress and bioenergetic failure in neuropsychiatric illnesses: Can we explain it and can we treat it? Mol. Biol. Rep. 2020, 47, 5587–5620. [Google Scholar] [CrossRef]
  275. Büttiker, P.; Weissenberger, S.; Esch, T.; Anders, M.; Raboch, J.; Ptacek, R.; Kream, R.M.; Stefano, G.B. Dysfunctional mitochondrial processes contribute to energy perturbations in the brain and neuropsychiatric symptoms. Front. Pharmacol. 2023, 13, 1095923. [Google Scholar] [CrossRef] [PubMed]
  276. Wei, C.; Sun, Y.; Chen, N.; Chen, S.; Xiu, M.; Zhang, X. Interaction of oxidative stress and BDNF on executive dysfunction in patients with chronic schizophrenia. Psychoneuroendocrinology 2020, 111, 104473. [Google Scholar] [CrossRef] [PubMed]
  277. Nakai, K.; Tsuruta, D. What Are Reactive Oxygen Species, Free Radicals, and Oxidative Stress in Skin Diseases? Int. J. Mol. Sci. 2021, 22, 10799. [Google Scholar] [CrossRef] [PubMed]
  278. Herbet, M.; Korga, A.; Gawrońska-Grzywacz, M.; Izdebska, M.; Piątkowska-Chmiel, I.; Poleszak, E.; Wróbel, A.; Matysiak, W.; Jodłowska-Jędrych, B.; Dudka, J. Chronic Variable Stress Is Responsible for Lipid and DNA Oxidative Disorders and Activation of Oxidative Stress Response Genes in the Brain of Rats. Oxidative Med. Cell. Longev. 2017, 2017, 7313090. [Google Scholar] [CrossRef] [PubMed]
  279. Juszczyk, G.; Mikulska, J.; Kasperek, K.; Pietrzak, D.; Mrozek, W.; Herbet, M. Chronic Stress and Oxidative Stress as Common Factors of the Pathogenesis of Depression and Alzheimer’s Disease: The Role of Antioxidants in Prevention and Treatment. Antioxidants 2021, 10, 1439. [Google Scholar] [CrossRef] [PubMed]
  280. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nature reviews. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  281. Gu, S.; Li, Y.; Jiang, Y.; Huang, J.H.; Wang, F. Glymphatic Dysfunction Induced Oxidative Stress and Neuro-Inflammation in Major Depression Disorders. Antioxidants 2022, 11, 2296. [Google Scholar] [CrossRef]
  282. Almutairi, M.M.; Sivandzade, F.; Albekairi, T.H.; Alqahtani, F.; Cucullo, L. Neuroinflammation and Its Impact on the Pathogenesis of COVID-19. Front. Med. 2021, 8, 745789. [Google Scholar] [CrossRef] [PubMed]
  283. Akanchise, T.; Angelova, A. Potential of Nano-Antioxidants and Nanomedicine for Recovery from Neurological Disorders Linked to Long COVID Syndrome. Antioxidants 2023, 12, 393. [Google Scholar] [CrossRef] [PubMed]
  284. Bayat, A.H.; Azimi, H.; Hassani Moghaddam, M.; Ebrahimi, V.; Fathi, M.; Vakili, K.; Mahmoudiasl, G.R.; Forouzesh, M.; Boroujeni, M.E.; Nariman, Z.; et al. COVID-19 causes neuronal degeneration and reduces neurogenesis in human hippocampus. Apoptosis Int. J. Program. Cell Death 2022, 27, 852–868. [Google Scholar] [CrossRef] [PubMed]
  285. Proust, A.; Queval, C.J.; Harvey, R.; Adams, L.; Bennett, M.; Wilkinson, R.J. Differential effects of SARS-CoV-2 variants on central nervous system cells and blood-brain barrier functions. J. Neuroinflammation 2023, 20, 184. [Google Scholar] [CrossRef] [PubMed]
  286. Clough, E.; Inigo, J.; Chandra, D.; Chaves, L.; Reynolds, J.L.; Aalinkeel, R.; Schwartz, S.A.; Khmaladze, A.; Mahajan, S.D. Mitochondrial Dynamics in SARS-COV2 Spike Protein Treated Human Microglia: Implications for Neuro-COVID. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2021, 16, 770–784. [Google Scholar] [CrossRef] [PubMed]
  287. Barichello, T.; Giridharan, V.V.; Catalão CH, R.; Ritter, C.; Dal-Pizzol, F. Neurochemical effects of sepsis on the brain. Clin. Sci. 2023, 137, 401–414. [Google Scholar] [CrossRef] [PubMed]
  288. Gotelli, E.; Soldano, S.; Hysa, E.; Casabella, A.; Cere, A.; Pizzorni, C.; Paolino, S.; Sulli, A.; Smith, V.; Cutolo, M. Understanding the Immune-Endocrine Effects of Vitamin D in SARS-CoV-2 Infection: A Role in Protecting against Neurodamage. Neuroimmunomodulation 2023, 30, 185–195. [Google Scholar] [CrossRef] [PubMed]
  289. Marsland, A.L.; Sathanoori, R.; Muldoon, M.F.; Manuck, S.B. Stimulated production of interleukin-8 covaries with psychosocial risk factors for inflammatory disease among middle-aged community volunteers. Brain Behav. Immun. 2007, 21, 218–228. [Google Scholar] [CrossRef]
  290. DeForge, L.E.; Preston, A.M.; Takeuchi, E.; Kenney, J.; Boxer, L.A.; Remick, D.G. Regulation of interleukin 8 gene expression by oxidant stress. J. Biol. Chem. 1993, 268, 25568–25576. [Google Scholar] [CrossRef]
  291. Dharshini LC, P.; Rasmi, R.R.; Kathirvelan, C.; Kumar, K.M.; Saradhadevi, K.M.; Sakthivel, K.M. Regulatory Components of Oxidative Stress and Inflammation and Their Complex Interplay in Carcinogenesis. Appl. Biochem. Biotechnol. 2023, 195, 2893–2916. [Google Scholar] [CrossRef]
  292. Laforge, M.; Elbim, C.; Frère, C.; Hémadi, M.; Massaad, C.; Nuss, P.; Benoliel, J.J.; Becker, C. Tissue damage from neutrophil-induced oxidative stress in COVID-19. Nature reviews. Immunology 2020, 20, 515–516. [Google Scholar] [CrossRef] [PubMed]
  293. Bonner-Jackson, A.; Vangal, R.; Li, Y.; Thompson, N.; Chakrabarti, S.; Krishnan, K. Factors associated with cognitive impairment in patients with persisting sequelae of COVID-19. Am. J. Med. 2024; Advance online publication. [Google Scholar] [CrossRef]
  294. Colizzi, M.; Comacchio, C.; De Martino, M.; Peghin, M.; Bontempo, G.; Chiappinotto, S.; Fonda, F.; Isola, M.; Tascini, C.; Balestrieri, M.; et al. COVID-19-induced neuropsychiatric symptoms can persist long after acute infection: A 2-year prospective study of biobehavioral risk factors and psychometric outcomes. Ir. J. Psychol. Med. 2024; 1–8, Advance online publication. [Google Scholar] [CrossRef]
  295. Lee, J.S.; Choi, Y.; Joung, J.Y.; Son, C.G. Clinical and Laboratory Characteristics of Fatigue-dominant Long-COVID subjects: A Cross-Sectional Study. Am. J. Med. 2024; Advance online publication. [Google Scholar] [CrossRef]
  296. Rogn, Å.; Jensen, J.L.; Iversen, P.O.; Singh, P.B. Post-COVID-19 patients suffer from chemosensory, trigeminal, and salivary dysfunctions. Sci. Rep. 2024, 14, 3455. [Google Scholar] [CrossRef] [PubMed]
  297. Titze-de-Almeida, R.; Araújo Lacerda, P.H.; de Oliveira, E.P.; de Oliveira ME, F.; Vianna YS, S.; Costa, A.M.; Pereira Dos Santos, E.; Guérard LM, C.; Ferreira MA, M.; Rodrigues Dos Santos, I.C.; et al. Sleep and memory complaints in long COVID: An insight into clustered psychological phenotypes. PeerJ 2024, 12, e16669. [Google Scholar] [CrossRef]
  298. Toepfner, N.; Brinkmann, F.; Augustin, S.; Stojanov, S.; Behrends, U. Long COVID in pediatrics-epidemiology, diagnosis, and management. Eur. J. Pediatr. 2024, 183, 1543–1553. [Google Scholar] [CrossRef]
  299. Zhao, S.; Martin, E.M.; Reuken, P.A.; Scholcz, A.; Ganse-Dumrath, A.; Srowig, A.; Utech, I.; Kozik, V.; Radscheidt, M.; Brodoehl, S.; et al. Long COVID is associated with severe cognitive slowing: A multicentre cross-sectional study. EClinicalMedicine 2024, 68, 102434. [Google Scholar] [CrossRef]
  300. Yasir, S.; Jin, Y.; Razzaq, F.A.; Caballero-Moreno, A.; Galán-García, L.; Ren, P.; Valdes-Sosa, M.; Rodriguez-Labrada, R.; Bringas-Vega, M.L.; Valdes-Sosa, P.A. The determinants of COVID-induced brain dysfunctions after SARS-CoV-2 infection in hospitalized patients. Front. Neurosci. 2024, 17, 1249282. [Google Scholar] [CrossRef]
  301. Wu, Z.W.; Yu, H.H.; Wang, X.; Guan, H.Y.; Xiu, M.H.; Zhang, X.Y. Interrelationships Between Oxidative Stress, Cytokines, and Psychotic Symptoms and Executive Functions in Patients With Chronic Schizophrenia. Psychosom. Med. 2021, 83, 485–491. [Google Scholar] [CrossRef] [PubMed]
  302. Urso, M.L.; Clarkson, P.M. Oxidative stress, exercise, and antioxidant supplementation. Toxicology 2003, 189, 41–54. [Google Scholar] [CrossRef]
  303. Jelic, M.D.; Mandic, A.D.; Maricic, S.M.; Srdjenovic, B.U. Oxidative stress and its role in cancer. J. Cancer Res. Ther. 2021, 17, 22–28. [Google Scholar] [CrossRef]
  304. Hassan, L.; Bueno, P.; Ferrón-Celma, I.; Ramia, J.M.; Garrote, D.; Muffak, K.; García-Navarro, A.; Mansilla, A.; Villar, J.M.; Ferrón, J.A. Time course of antioxidant enzyme activities in liver transplant recipients. Transplant. Proc. 2005, 37, 3932–3935. [Google Scholar] [CrossRef]
  305. Wang, Q.S.; Zhang, C.L.; Zhao, X.L.; Yu, S.F.; Xie, K.Q. Malondialdehyde and catalase as the serum biomarkers of allyl chloride-induced toxic neuropathy. Toxicology 2006, 227, 36–44. [Google Scholar] [CrossRef] [PubMed]
  306. Pérez-Hernández, O.; González-Reimers, E.; Quintero-Platt, G.; Abreu-González, P.; Vega-Prieto, M.J.; Sánchez-Pérez, M.J.; Martín-González, C.; Martínez-Riera, A.; Santolaria-Fernández, F. Malondialdehyde as a Prognostic Factor in Alcoholic Hepatitis. Alcohol Alcohol. 2017, 52, 305–310. [Google Scholar] [CrossRef] [PubMed]
  307. Spapen, H.; Zhang, H.; Demanet, C.; Vleminckx, W.; Vincent, J.L.; Huyghens, L. Does N-acetyl-L-cysteine influence cytokine response during early human septic shock? Chest 1998, 113, 1616–1624. [Google Scholar] [CrossRef]
  308. Csontos, C.; Rezman, B.; Foldi, V.; Bogar, L.; Drenkovics, L.; Röth, E.; Weber, G.; Lantos, J. Effect of N-acetylcysteine treatment on oxidative stress and inflammation after severe burn. Burn. J. Int. Soc. Burn. Inj. 2012, 38, 428–437. [Google Scholar] [CrossRef]
  309. Atkinson, M.C. The use of N-acetylcysteine in intensive care. Crit. Care Resusc. J. Australas. Acad. Crit. Care Med. 2002, 4, 21–27. [Google Scholar] [CrossRef]
  310. Raghu, G.; Berk, M.; Campochiaro, P.A.; Jaeschke, H.; Marenzi, G.; Richeldi, L.; Wen, F.Q.; Nicoletti, F.; Calverley, P.M.A. The Multifaceted Therapeutic Role of N-Acetylcysteine (NAC) in Disorders Characterized by Oxidative Stress. Curr. Neuropharmacol. 2021, 19, 1202–1224. [Google Scholar] [CrossRef]
  311. Schwalfenberg, G.K. N-Acetylcysteine: A Review of Clinical Usefulness (an Old Drug with New Tricks). J. Nutr. Metab. 2021, 2021, 9949453. [Google Scholar] [CrossRef]
  312. Chen, H.; Ma, N.; Song, X.; Wei, G.; Zhang, H.; Liu, J.; Shen, X.; Zhuge, X.; Chang, G. Protective Effects of N-Acetylcysteine on Lipopolysaccharide-Induced Respiratory Inflammation and Oxidative Stress. Antioxidants 2022, 11, 879. [Google Scholar] [CrossRef] [PubMed]
  313. Jain, A.; Mårtensson, J.; Stole, E.; Auld, P.A.; Meister, A. Glutathione deficiency leads to mitochondrial damage in brain. Proc. Natl. Acad. Sci. USA 1991, 88, 1913–1917. [Google Scholar] [CrossRef]
  314. Heales, S.J.; Davies, S.E.; Bates, T.E.; Clark, J.B. Depletion of brain glutathione is accompanied by impaired mitochondrial function and decreased N-acetyl aspartate concentration. Neurochem. Res. 1995, 20, 31–38. [Google Scholar] [CrossRef]
  315. Drukarch, B.; Schepens, E.; Jongenelen, C.A.; Stoof, J.C.; Langeveld, C.H. Astrocyte-mediated enhancement of neuronal survival is abolished by glutathione deficiency. Brain Res. 1997, 770, 123–130. [Google Scholar] [CrossRef] [PubMed]
  316. Dringen, R. Metabolism and functions of glutathione in brain. Prog. Neurobiol. 2000, 62, 649–671. [Google Scholar] [CrossRef] [PubMed]
  317. Cruz-Aguado, R.; Almaguer-Melian, W.; Díaz, C.M.; Lorigados, L.; Bergado, J. Behavioral and biochemical effects of glutathione depletion in the rat brain. Brain Res. Bull. 2001, 55, 327–333. [Google Scholar] [CrossRef] [PubMed]
  318. Wu, G.; Fang, Y.Z.; Yang, S.; Lupton, J.R.; Turner, N.D. Glutathione metabolism and its implications for health. J. Nutr. 2004, 134, 489–492. [Google Scholar] [CrossRef] [PubMed]
  319. Hong, H.; Lu, Y.; Ji, Z.N.; Liu, G.Q. Up-regulation of P-glycoprotein expression by glutathione depletion-induced oxidative stress in rat brain microvessel endothelial cells. J. Neurochem. 2006, 98, 1465–1473. [Google Scholar] [CrossRef] [PubMed]
  320. González-Fraguela, M.E.; Blanco, L.; Fernández, C.I.; Lorigados, L.; Serrano, T.; Fernández, J.L. Glutathione depletion: Starting point of brain metabolic stress, neuroinflammation and cognitive impairment in rats. Brain Res. Bull. 2018, 137, 120–131. [Google Scholar] [CrossRef] [PubMed]
  321. Berk, M.; Copolov, D.; Dean, O.; Lu, K.; Jeavons, S.; Schapkaitz, I.; Anderson-Hunt, M.; Judd, F.; Katz, F.; Katz, P.; et al. N-acetyl cysteine as a glutathione precursor for schizophrenia—A double-blind, randomized, placebo-controlled trial. Biol. Psychiatry 2008, 64, 361–368. [Google Scholar] [CrossRef] [PubMed]
  322. Duailibi, M.S.; Cordeiro, Q.; Brietzke, E.; Ribeiro, M.; LaRowe, S.; Berk, M.; Trevizol, A.P. N-acetylcysteine in the treatment of craving in substance use disorders: Systematic review and meta-analysis. Am. J. Addict. 2017, 26, 660–666. [Google Scholar] [CrossRef] [PubMed]
  323. Tharoor, H.; Mara, S.; Gopal, S. Role of Novel Dietary Supplement N-acetyl Cysteine in Treating Negative Symptoms in Schizophrenia: A 6-Month Follow-up Study. Indian J. Psychol. Med. 2018, 40, 139–142. [Google Scholar] [CrossRef]
  324. Ooi, S.L.; Green, R.; Pak, S.C. N-Acetylcysteine for the Treatment of Psychiatric Disorders: A Review of Current Evidence. BioMed Res. Int. 2018, 2018, 2469486. [Google Scholar] [CrossRef]
  325. Mullier, E.; Roine, T.; Griffa, A.; Xin, L.; Baumann, P.S.; Klauser, P.; Cleusix, M.; Jenni, R.; Alemàn-Gómez, Y.; Gruetter, R.; et al. N-Acetyl-Cysteine Supplementation Improves Functional Connectivity Within the Cingulate Cortex in Early Psychosis: A Pilot Study. Int. J. Neuropsychopharmacol. 2019, 22, 478–487. [Google Scholar] [CrossRef] [PubMed]
  326. Yolland, C.O.; Hanratty, D.; Neill, E.; Rossell, S.L.; Berk, M.; Dean, O.M.; Castle, D.J.; Tan, E.J.; Phillipou, A.; Harris, A.W.; et al. Meta-analysis of randomised controlled trials with N-acetylcysteine in the treatment of schizophrenia. Aust. N. Zealand J. Psychiatry 2020, 54, 453–466. [Google Scholar] [CrossRef] [PubMed]
  327. Chang, C.T.; Hsieh, P.J.; Lee, H.C.; Lo, C.H.; Tam, K.W.; Loh, E.W. Effectiveness of N-acetylcysteine in Treating Clinical Symptoms of Substance Abuse and Dependence: A Meta-analysis of Randomized Controlled Trials. Clin. Psychopharmacol. Neurosci. Off. Sci. J. Korean Coll. Neuropsychopharmacol. 2021, 19, 282–293. [Google Scholar] [CrossRef] [PubMed]
  328. Lee, T.M.; Lee, K.M.; Lee, C.Y.; Lee, H.C.; Tam, K.W.; Loh, E.W. Effectiveness of N-acetylcysteine in autism spectrum disorders: A meta-analysis of randomized controlled trials. Aust. N. Zealand J. Psychiatry 2021, 55, 196–206. [Google Scholar] [CrossRef] [PubMed]
  329. Martinez-Banaclocha, M. N-Acetyl-Cysteine: Modulating the Cysteine Redox Proteome in Neurodegenerative Diseases. Antioxidants 2022, 11, 416. [Google Scholar] [CrossRef] [PubMed]
  330. Khalatbari Mohseni, G.; Hosseini, S.A.; Majdinasab, N.; Cheraghian, B. Effects of N-acetylcysteine on oxidative stress biomarkers, depression, and anxiety symptoms in patients with multiple sclerosis. Neuropsychopharmacol. Rep. 2023, 43, 382–390. [Google Scholar] [CrossRef] [PubMed]
  331. Radtke, F.A.; Chapman, G.; Hall, J.; Syed, Y.A. Modulating Neuroinflammation to Treat Neuropsychiatric Disorders. BioMed Res. Int. 2017, 2017, 5071786. [Google Scholar] [CrossRef]
  332. Abdoli, A.; Taghipour, A.; Pirestani, M.; Mofazzal Jahromi, M.A.; Roustazadeh, A.; Mir, H.; Ardakani, H.M.; Kenarkoohi, A.; Falahi, S.; Karimi, M. Infections, inflammation, and risk of neuropsychiatric disorders: The neglected role of "co-infection". Heliyon 2020, 6, e05645. [Google Scholar] [CrossRef] [PubMed]
  333. Kempuraj, D.; Thangavel, R.; Natteru, P.A.; Selvakumar, G.P.; Saeed, D.; Zahoor, H.; Zaheer, S.; Iyer, S.S.; Zaheer, A. Neuroinflammation Induces Neurodegeneration. J. Neurol. Neurosurg. Spine 2016, 1, 1003. [Google Scholar]
  334. Stuart, M.J.; Singhal, G.; Baune, B.T. Systematic Review of the Neurobiological Relevance of Chemokines to Psychiatric Disorders. Front. Cell. Neurosci. 2015, 9, 357. [Google Scholar] [CrossRef]
  335. Frydecka, D.; Krzystek-Korpacka, M.; Lubeiro, A.; Stramecki, F.; Stańczykiewicz, B.; Beszłej, J.A.; Piotrowski, P.; Kotowicz, K.; Szewczuk-Bogusławska, M.; Pawlak-Adamska, E.; et al. Profiling inflammatory signatures of schizophrenia: A cross-sectional and meta-analysis study. Brain Behav. Immun. 2018, 71, 28–36. [Google Scholar] [CrossRef] [PubMed]
  336. Kölliker-Frers, R.; Udovin, L.; Otero-Losada, M.; Kobiec, T.; Herrera, M.I.; Palacios, J.; Razzitte, G.; Capani, F. Neuroinflammation: An Integrating Overview of Reactive-Neuroimmune Cell Interactions in Health and Disease. Mediat. Inflamm. 2021, 2021, 9999146. [Google Scholar] [CrossRef] [PubMed]
  337. Schmouder, R.L.; Strieter, R.M.; Wiggins, R.C.; Chensue, S.W.; Kunkel, S.L. In vitro and in vivo interleukin-8 production in human renal cortical epithelia. Kidney Int. 1992, 41, 191–198. [Google Scholar] [CrossRef] [PubMed]
  338. Tsai, S.J. Role of interleukin 8 in depression and other psychiatric disorders. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 106, 110173. [Google Scholar] [CrossRef] [PubMed]
  339. Skibinska, M.; Rajewska-Rager, A.; Dmitrzak-Weglarz, M.; Kapelski, P.; Lepczynska, N.; Kaczmarek, M.; Pawlak, J. Interleukin-8 and tumor necrosis factor-alpha in youth with mood disorders-A longitudinal study. Front. Psychiatry 2022, 13, 964538. [Google Scholar] [CrossRef] [PubMed]
  340. Atta-ur-Rahman Harvey, K.; Siddiqui, R.A. Interleukin-8: An autocrine inflammatory mediator. Curr. Pharm. Des. 1999, 5, 241–253. [Google Scholar]
  341. Weik, U.; Herforth, A.; Kolb-Bachofen, V.; Deinzer, R. Acute stress induces proinflammatory signaling at chronic inflammation sites. Psychosom. Med. 2008, 70, 906–912. [Google Scholar] [CrossRef]
  342. Godoy, L.D.; Rossignoli, M.T.; Delfino-Pereira, P.; Garcia-Cairasco, N.; de Lima Umeoka, E.H. A Comprehensive Overview on Stress Neurobiology: Basic Concepts and Clinical Implications. Front. Behav. Neurosci. 2018, 12, 127. [Google Scholar] [CrossRef] [PubMed]
  343. Suarez, E.C.; Lewis, J.G.; Krishnan, R.R.; Young, K.H. Enhanced expression of cytokines and chemokines by blood monocytes to in vitro lipopolysaccharide stimulation are associated with hostility and severity of depressive symptoms in healthy women. Psychoneuroendocrinology 2004, 29, 1119–1128. [Google Scholar] [CrossRef]
  344. Dutheil, F.; Trousselard, M.; Perrier, C.; Lac, G.; Chamoux, A.; Duclos, M.; Naughton, G.; Mnatzaganian, G.; Schmidt, J. Urinary interleukin-8 is a biomarker of stress in emergency physicians, especially with advancing age--the JOBSTRESS* randomized trial. PLoS ONE 2013, 8, e71658. [Google Scholar] [CrossRef]
  345. Fukuda, H.; Ichinose, T.; Kusama, T.; Sakurai, R.; Anndow, K.; Akiyoshi, N. Stress assessment in acute care department nurses by measuring interleukin-8. Int. Nurs. Rev. 2008, 55, 407–411. [Google Scholar] [CrossRef] [PubMed]
  346. Rahal, A.; Kumar, A.; Singh, V.; Yadav, B.; Tiwari, R.; Chakraborty, S.; Dhama, K. Oxidative stress, prooxidants, and antioxidants: The interplay. BioMed Res. Int. 2014, 2014, 761264. [Google Scholar] [CrossRef] [PubMed]
  347. Wang, L.; Muxin, G.; Nishida, H.; Shirakawa, C.; Sato, S.; Konishi, T. Psychological stress-induced oxidative stress as a model of sub-healthy condition and the effect of TCM. Evid. -Based Complement. Altern. Med. Ecam 2007, 4, 195–202. [Google Scholar] [CrossRef] [PubMed]
  348. Aschbacher, K.; O’Donovan, A.; Wolkowitz, O.M.; Dhabhar, F.S.; Su, Y.; Epel, E. Good stress, bad stress and oxidative stress: Insights from anticipatory cortisol reactivity. Psychoneuroendocrinology 2013, 38, 1698–1708. [Google Scholar] [CrossRef] [PubMed]
  349. Kim, E.; Zhao, Z.; Rzasa, J.R.; Glassman, M.; Bentley, W.E.; Chen, S.; Kelly, D.L.; Payne, G.F. Association of acute psychosocial stress with oxidative stress: Evidence from serum analysis. Redox Biol. 2021, 47, 102138. [Google Scholar] [CrossRef]
  350. Srivastava, K.K.; Kumar, R. Stress, oxidative injury and disease. Indian J. Clin. Biochem. IJCB 2015, 30, 3–10. [Google Scholar] [CrossRef] [PubMed]
  351. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive Oxygen Species in Metabolic and Inflammatory Signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef] [PubMed]
  352. Salim, S. Oxidative Stress and the Central Nervous System. J. Pharmacol. Exp. Ther. 2017, 360, 201–205. [Google Scholar] [CrossRef] [PubMed]
  353. Solleiro-Villavicencio, H.; Rivas-Arancibia, S. Effect of Chronic Oxidative Stress on Neuroinflammatory Response Mediated by CD4+T Cells in Neurodegenerative Diseases. Front. Cell. Neurosci. 2018, 12, 114. [Google Scholar] [CrossRef]
  354. Fabisiak, T.; Patel, M. Crosstalk between neuroinflammation and oxidative stress in epilepsy. Front. Cell Dev. Biol. 2022, 10, 976953. [Google Scholar] [CrossRef]
  355. He, J.; Zhu, G.; Wang, G.; Zhang, F. Oxidative Stress and Neuroinflammation Potentiate Each Other to Promote Progression of Dopamine Neurodegeneration. Oxidative Med. Cell. Longev. 2020, 2020, 6137521. [Google Scholar] [CrossRef] [PubMed]
  356. Sheppard, A.J.; Barfield, A.M.; Barton, S.; Dong, Y. Understanding Reactive Oxygen Species in Bone Regeneration: A Glance at Potential Therapeutics and Bioengineering Applications. Front. Bioeng. Biotechnol. 2022, 10, 836764. [Google Scholar] [CrossRef] [PubMed]
  357. Verhasselt, V.; Goldman, M.; Willems, F. Oxidative stress up-regulates IL-8 and TNF-alpha synthesis by human dendritic cells. Eur. J. Immunol. 1998, 28, 3886–3890. [Google Scholar] [CrossRef]
  358. Ivison, S.M.; Wang, C.; Himmel, M.E.; Sheridan, J.; Delano, J.; Mayer, M.L.; Yao, Y.; Kifayet, A.; Steiner, T.S. Oxidative stress enhances IL-8 and inhibits CCL20 production from intestinal epithelial cells in response to bacterial flagellin. American journal of physiology. Gastrointest. Liver Physiol. 2010, 299, G733–G741. [Google Scholar] [CrossRef] [PubMed]
  359. Sarir, H.; Mortaz, E.; Janse, W.T.; Givi, M.E.; Nijkamp, F.P.; Folkerts, G. IL-8 production by macrophages is synergistically enhanced when cigarette smoke is combined with TNF-alpha. Biochem. Pharmacol. 2010, 79, 698–705. [Google Scholar] [CrossRef] [PubMed]
  360. Miyoshi, T.; Yamashita, K.; Arai, T.; Yamamoto, K.; Mizugishi, K.; Uchiyama, T. The role of endothelial interleukin-8/NADPH oxidase 1 axis in sepsis. Immunology 2010, 131, 331–339. [Google Scholar] [CrossRef] [PubMed]
  361. Bezerra, W.P.; Salmeron AC, A.; Branco AC, C.C.; Morais, I.C.; de Farias Sales, V.S.; Machado PR, L.; Souto, J.T.; de Araújo JM, G.; Guedes PM, D.M.; Sato, M.N.; et al. Low CCL2 and CXCL8 Production and High Prevalence of Allergies in Children with Microcephaly Due to Congenital Zika Syndrome. Viruses 2023, 15, 1832. [Google Scholar] [CrossRef] [PubMed]
  362. Masso-Silva, J.A.; Moshensky, A.; Lam MT, Y.; Odish, M.F.; Patel, A.; Xu, L.; Hansen, E.; Trescott, S.; Nguyen, C.; Kim, R.; et al. Increased Peripheral Blood Neutrophil Activation Phenotypes and Neutrophil Extracellular Trap Formation in Critically Ill Coronavirus Disease 2019 (COVID-19) Patients: A Case Series and Review of the Literature. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2022, 74, 479–489. [Google Scholar] [CrossRef] [PubMed]
  363. Dantzer, R. Neuroimmune Interactions: From the Brain to the Immune System and Vice Versa. Physiol. Rev. 2018, 98, 477–504. [Google Scholar] [CrossRef]
  364. Calcia, M.A.; Bonsall, D.R.; Bloomfield, P.S.; Selvaraj, S.; Barichello, T.; Howes, O.D. Stress and neuroinflammation: A systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology 2016, 233, 1637–1650. [Google Scholar] [CrossRef]
  365. Godinho-Silva, C.; Cardoso, F.; Veiga-Fernandes, H. Neuro-Immune Cell Units: A New Paradigm in Physiology. Annu. Rev. Immunol. 2019, 37, 19–46. [Google Scholar] [CrossRef] [PubMed]
  366. Thomson, C.A.; McColl, A.; Graham, G.J.; Cavanagh, J. Sustained exposure to systemic endotoxin triggers chemokine induction in the brain followed by a rapid influx of leukocytes. J. Neuroinflammation 2020, 17, 94. [Google Scholar] [CrossRef] [PubMed]
  367. Munhoz, C.D.; García-Bueno, B.; Madrigal, J.L.; Lepsch, L.B.; Scavone, C.; Leza, J.C. Stress-induced neuroinflammation: Mechanisms and new pharmacological targets. Braz. J. Med. Biol. Res. Rev. Bras. De Pesqui. Medicas E Biol. 2008, 41, 1037–1046. [Google Scholar] [CrossRef] [PubMed]
  368. Tansey, M.G.; Goldberg, M.S. Neuroinflammation in Parkinson’s disease: Its role in neuronal death and implications for therapeutic intervention. Neurobiol. Dis. 2010, 37, 510–518. [Google Scholar] [CrossRef] [PubMed]
  369. Thibaut, F. Neuroinflammation: New vistas for neuropsychiatric research. Dialogues Clin. Neurosci. 2017, 19, 3–4. [Google Scholar] [CrossRef] [PubMed]
  370. Limphaibool, N.; Iwanowski, P.; Holstad MJ, V.; Kobylarek, D.; Kozubski, W. Infectious Etiologies of Parkinsonism: Pathomechanisms and Clinical Implications. Front. Neurol. 2019, 10, 652. [Google Scholar] [CrossRef] [PubMed]
  371. Aggarwal, V.; Mehndiratta, M.M.; Wasay, M.; Garg, D. Environmental Toxins and Brain: Life on Earth is in Danger. Ann. Indian Acad. Neurol. 2022, 25 (Suppl. S1), S15–S21. [Google Scholar] [CrossRef] [PubMed]
  372. Drieu, A.; Lanquetin, A.; Prunotto, P.; Gulhan, Z.; Pédron, S.; Vegliante, G.; Tolomeo, D.; Serrière, S.; Vercouillie, J.; Galineau, L.; et al. Persistent neuroinflammation and behavioural deficits after single mild traumatic brain injury. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2022, 42, 2216–2229. [Google Scholar] [CrossRef]
  373. Sun, Y.; Koyama, Y.; Shimada, S. Inflammation From Peripheral Organs to the Brain: How Does Systemic Inflammation Cause Neuroinflammation? Front. Aging Neurosci. 2022, 14, 903455. [Google Scholar] [CrossRef]
  374. Tan, S.; Chen, W.; Kong, G.; Wei, L.; Xie, Y. Peripheral inflammation and neurocognitive impairment: Correlations, underlying mechanisms, and therapeutic implications. Front. Aging Neurosci. 2023, 15, 1305790. [Google Scholar] [CrossRef]
  375. Millán Solano, M.V.; Salinas Lara, C.; Sánchez-Garibay, C.; Soto-Rojas, L.O.; Escobedo-Ávila, I.; Tena-Suck, M.L.; Ortíz-Butrón, R.; Choreño-Parra, J.A.; Romero-López, J.P.; Meléndez Camargo, M.E. Effect of Systemic Inflammation in the CNS: A Silent History of Neuronal Damage. Int. J. Mol. Sci. 2023, 24, 11902. [Google Scholar] [CrossRef] [PubMed]
  376. Mayr, F.B.; Spiel, A.O.; Leitner, J.M.; Firbas, C.; Kliegel, T.; Jilma, B. Ethnic differences in plasma levels of interleukin-8 (IL-8) and granulocyte colony stimulating factor (G-CSF). Transl. Res. J. Lab. Clin. Med. 2007, 149, 10–14. [Google Scholar] [CrossRef] [PubMed]
  377. Christian, L.M.; Kowalsky, J.M.; Mitchell, A.M.; Porter, K. Associations of postpartum sleep, stress, and depressive symptoms with LPS-stimulated cytokine production among African American and White women. J. Neuroimmunol. 2018, 316, 98–106. [Google Scholar] [CrossRef] [PubMed]
  378. Chen, C.H.; Ho, C.H.; Hu, S.W.; Tzou, K.Y.; Wang, Y.H.; Wu, C.C. Association between interleukin-8 rs4073 polymorphism and prostate cancer: A meta-analysis. J. Formos. Med. Assoc. Taiwan Yi Zhi 2020, 119, 1201–1210. [Google Scholar] [CrossRef] [PubMed]
  379. Hildebrand, F.; Stuhrmann, M.; van Griensven, M.; Meier, S.; Hasenkamp, S.; Krettek, C.; Pape, H.C. Association of IL-8-251A/T polymorphism with incidence of Acute Respiratory Distress Syndrome (ARDS) and IL-8 synthesis after multiple trauma. Cytokine 2007, 37, 192–199. [Google Scholar] [CrossRef] [PubMed]
  380. Bishu, S.; Koutroumpakis, E.; Mounzer, R.; Stello, K.; Pollock, N.; Evans, A.; Whitcomb, D.C.; Papachristou, G.I. The -251 A/T Polymorphism in the IL8 Promoter is a Risk Factor for Acute Pancreatitis. Pancreas 2018, 47, 87–91. [Google Scholar] [CrossRef]
  381. Santos CN, O.; Magalhães, L.S.; Fonseca AB, L.; Bispo AJ, B.; Porto RL, S.; Alves, J.C.; Dos Santos, C.A.; de Carvalho, J.V.; da Silva, A.M.; Teixeira, M.M.; et al. Association between genetic variants in TREM1, CXCL10, IL4, CXCL8 and TLR7 genes with the occurrence of congenital Zika syndrome and severe microcephaly. Sci. Rep. 2023, 13, 3466. [Google Scholar] [CrossRef] [PubMed]
  382. Zhao, X.F.; Zhu, S.Y.; Hu, H.; He, C.L.; Zhang, Y.; Li, Y.F.; Wu, Y.Q. Association between interleukin-8 rs4073 polymorphisms and susceptibility to neonatal sepsis. Zhongguo Dang Dai Er Ke Za Zhi Chin. J. Contemp. Pediatr. 2020, 22, 323–327. [Google Scholar] [CrossRef]
  383. Wang, Z.; Wang, C.; Zhao, Z.; Liu, F.; Guan, X.; Lin, X.; Zhang, L. Association between -251A>T polymorphism in the interleukin-8 gene and oral cancer risk: A meta-analysis. Gene 2013, 522, 168–176. [Google Scholar] [CrossRef]
  384. Zhang, S.; Gao, Y.; Huang, J. Interleukin-8 Gene -251 A/T (rs4073) Polymorphism and Coronary Artery Disease Risk: A Meta-Analysis. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 1645–1655. [Google Scholar] [CrossRef]
  385. Peedicayil, J. Genome-Environment Interactions and Psychiatric Disorders. Biomedicines 2023, 11, 1209. [Google Scholar] [CrossRef] [PubMed]
  386. Ghazy, A.A. Influence of IL-6 rs1800795 and IL-8 rs2227306 polymorphisms on COVID-19 outcome. J. Infect. Dev. Ctries. 2023, 17, 327–334. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic presentation of IL-8 inducers: IL-1α, IL-1β, IL-7, IL-17, IL-22, tumor necrosis factor-alpha (TNF-α), histamine, stromal cell-derived factor-1 (SDF-1, CXCL12), lipopolysaccharides (LPSs), reactive oxygen species (ROS), cadmium (Cd), phytohemagglutinin (PHA), prostaglandin E2, polyinosinic-polycytidylic acid (poly I:C), concanavalin A (ConA), NaCl, thrombin, and all-trans-retinoic acid (ATRA).
Figure 1. Schematic presentation of IL-8 inducers: IL-1α, IL-1β, IL-7, IL-17, IL-22, tumor necrosis factor-alpha (TNF-α), histamine, stromal cell-derived factor-1 (SDF-1, CXCL12), lipopolysaccharides (LPSs), reactive oxygen species (ROS), cadmium (Cd), phytohemagglutinin (PHA), prostaglandin E2, polyinosinic-polycytidylic acid (poly I:C), concanavalin A (ConA), NaCl, thrombin, and all-trans-retinoic acid (ATRA).
Jpm 14 00488 g001
Figure 2. Schematic presentation of IL-8 reducers: IL-4, IL-10, IL-35, transforming growth factor-beta 1 (TGF-β1), interferon-alpha (IFN-α), interferon-beta (IFN-β), glucocorticoids (GCs), lipoxins, vitamin D, lipoxygenase (LOX) inhibitors, antcin K, tannins, glycyrrhizin (GL), and N-acetylcysteine (NAC).
Figure 2. Schematic presentation of IL-8 reducers: IL-4, IL-10, IL-35, transforming growth factor-beta 1 (TGF-β1), interferon-alpha (IFN-α), interferon-beta (IFN-β), glucocorticoids (GCs), lipoxins, vitamin D, lipoxygenase (LOX) inhibitors, antcin K, tannins, glycyrrhizin (GL), and N-acetylcysteine (NAC).
Jpm 14 00488 g002
Table 1. CXCL8 gene SNPs.
Table 1. CXCL8 gene SNPs.
SNPAllelesGene: ConsequenceGenomic Position
rs4073A > C/A > G/A > TCXCL8: 2KB Upstream Variantchr4:73740307 (GRCh38.p14)
rs1126647A > C/A > TCXCL8: 3 Prime UTR Variantchr4:73743328 (GRCh38.p14)
rs2227306C > TCXCL8: Intron Variantchr4:73741338 (GRCh38.p14)
rs2227307T > C/T > GCXCL8: Intron Variantchr4:73740952 (GRCh38.p14)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shkundin, A.; Halaris, A. IL-8 (CXCL8) Correlations with Psychoneuroimmunological Processes and Neuropsychiatric Conditions. J. Pers. Med. 2024, 14, 488. https://doi.org/10.3390/jpm14050488

AMA Style

Shkundin A, Halaris A. IL-8 (CXCL8) Correlations with Psychoneuroimmunological Processes and Neuropsychiatric Conditions. Journal of Personalized Medicine. 2024; 14(5):488. https://doi.org/10.3390/jpm14050488

Chicago/Turabian Style

Shkundin, Anton, and Angelos Halaris. 2024. "IL-8 (CXCL8) Correlations with Psychoneuroimmunological Processes and Neuropsychiatric Conditions" Journal of Personalized Medicine 14, no. 5: 488. https://doi.org/10.3390/jpm14050488

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop