IMR Press / FBL / Volume 28 / Issue 12 / DOI: 10.31083/j.fbl2812356
Open Access Review
Inflammation and Late-Life Depression: Unraveling the Complex Relationship and Potential Therapeutic Strategies
Show Less
1 School of Medicine, Kunming University of Science and Technology, 650000 Kunming, Yunnan, China
2 Department of Clinical Psychology, The First People's Hospital of Yunnan Province, 650000 Kunming, Yunnan, China
3 The Affiliated Hospital of Kunming University of Science and Technology, 650000 Kunming, Yunnan, China
4 Department of Geriatrics, The First People's Hospital of Yunnan Province, 650000 Kunming, Yunnan, China
*Correspondence: shaoheng90@sina.com (Heng Shao)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(12), 356; https://doi.org/10.31083/j.fbl2812356
Submitted: 7 May 2023 | Revised: 21 August 2023 | Accepted: 7 September 2023 | Published: 28 December 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

The origins of late-life depression are multifaceted and remain challenging to fully understand. While the traditional monoamine neurotransmitter hypothesis provides some insights, it falls short in explaining the disease’s onset and progression, leaving treatments often less than optimal. There is an emergent need to uncover new underlying mechanisms. Among these, the “inflammation hypothesis” has been gaining traction in scientific discussions regarding late-life depression. There is compelling evidence linking inflammation processes to the emergence of this form of depression. This review delves into the nuanced relationship between inflammation and late-life depression, emphasizing the pivotal role and implications of inflammation in its pathogenesis. Changes in Ca2+ homeostasis, cytokine levels, brain-derived neurotrophic factor (BDNF), white cell ratios, and the involvement of the NOD-, LRR-, and Pyrin domain-containing protein 3 (NLRP3) inflammasome have all been suggested as potential biomarkers that tie inflammation to late-life depression. Furthermore, factors such as aging-induced DNA damage, oxidative stress, mitochondrial impairments, disruptions in the hypothalamic-pituitary-adrenal axis, activated microglia and associated neuroinflammation, as well as the gut-brain axis dynamics, could serve as bridges between inflammation and depression. Deepening our understanding of these connections could usher in innovative anti-inflammatory treatments and strategies for late- life depression.

Keywords
late life depression
inflammation
molecular mechanisms
pathway
personalized target and therapy
1. Introduction

Aging is a global trend, with the number and proportion of individuals aged 60 and over increasing [1], expected to rise from 10.0% in 2000 to 21.8% in 2050 [2]. Late-life depression (LLD) is a mental health disorder that primarily impacts older adults, typically those aged 60 years and older. This condition is characterized by ongoing feelings of sadness and hopelessness, coupled with a decreased interest in activities that were previously enjoyed [3]. It is a significant public health issue among the elderly and a leading cause of disability worldwide [4]. The prevalence of LLD varies significantly across the world [5]. A recent meta-analysis reported that the average estimated prevalence of LLD is 31.74%, with developing countries having a higher overall prevalence (40.78%) compared to developed countries (17.05%) [6]. The lifetime prevalence of severe depression in older adults in Western countries is 16.52% [7], while in European countries, it is 29% [8]. In China, the overall prevalence of depressive symptoms in the elderly population is 20.0% [9].

LLD has become a severe public health issue both in China and globally, with high prevalence, increased risk of suicide, and prolonged duration contributing to the growing disease burden [10]. The traditional “monoamine hypothesis” proposes that depression is linked to reduced levels of monoamine neurotransmitters in the brain, including norepinephrine (NE), 5-hydroxytryptamine (5-HT), and dopamine (DA) [11]. However, the etiology and pathogenesis of late-life depression (LLD) are complex, involving brain atrophy, vascular changes, white matter degradation, inflammatory responses, and genetic polymorphisms [12]. Compared to younger depressed patients, older patients with depression often have multiple comorbid physical illnesses and cognitive impairment [13]. The traditional monoamine neurotransmitter hypothesis alone cannot fully explain the pathogenesis and outcomes of LLD.

Currently, pharmacotherapy is the primary treatment for LLD. However, the effectiveness of antidepressants in managing LLD can be limited [14]. The response rate to antidepressants is typically lower in older versus younger depressed patients [15], and older individuals are more prone to relapse [16]. Therefore, exploring novel etiological theories and treatment approaches for LLD is critical.

Inflammatory aging is an inevitable phenomenon during the aging process. It begins with a low-grade “cold” inflammatory phase. Compared to healthy adults, older individuals exhibit only a slight elevation in plasma proinflammatory mediators, which helps maintain homeostasis [17]. However, with advancing age, the body’s homeostatic balance progressively deteriorates, leading to amplified cytokine responses mediated by the chronically activated innate immune system, typically increasing by two- to four-fold [18]. Elevated proinflammatory cytokines significantly modulate neuroplasticity and neurogenesis [19], triggering neuroinflammation [20], and impacting mood and cognition. Studies indicate LLD is closely tied to inflammation, with increased inflammatory responses potentially contributing critically to LLD development [21, 22]. By managing the inflammatory response, it is anticipated that LLD symptoms may be reduced, ultimately enhancing the quality of life for older adults [23]. In the future, further exploration of the relationship between inflammation and LLD is needed to enhance the effectiveness of prevention and treatment strategies for LLD.

2. Biological Basis of Inflammation and LLD
2.1 Inflammatory Pathways and Mediators
2.1.1 Cytokines

In the inflammatory mechanism of LLD, cytokines are among the important regulatory factors. Elevated peripheral cytokine levels are associated with depressive symptoms in the elderly, with the most consistent findings related to IL-6 [24], as well as IL-1β, IL-8, and tumor necrosis factor (TNF-α) [25, 26]. Dhabhar et al. [27] found that pro-inflammatory cytokines are increased in patients with depression, while anti-inflammatory cytokines such as IL-4, IL-10, IL-13, transforming growth factor β, and adiponectin are decreased. Elderly patients are often in a chronic pro-inflammatory state, which increases the activation and initiation of microglial cells, leading to continuous production of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α and a reduction in anti-inflammatory molecules [28]. This results in a rise in pro-inflammatory cytokines and activation of microglial cells, which contribute to brain pathology by increasing blood-brain barrier (BBB) permeability and cytokine production, ultimately leading to the onset of depression [29]. Some studies have found [30] that older individuals with clinical depression do not exhibit elevated levels of inflammation unless they also have other inflammatory conditions such as arthritis. However, Kim et al. [31] conducted a study on LLD and cytokines, providing support for cytokine-mediated inflammatory pathways. The variability in major depressive disorder (MDD) research findings might be attributed to certain studies incorporating depressed patients without systemic inflammation, while others focus on patients with inflammatory depression. Moreover, age disparities among study participants could be a factor, as older populations frequently experience chronic pro-inflammatory states, making them more fitting subjects for examining the inflammatory underpinnings of depression.

2.1.2 Acute Phase Proteins (APPs)

Acute phase proteins (APPs) are a class of proteins synthesized and released during acute inflammation and injury in the body, with important biological functions. They include C-reactive protein (CRP), pre-albumin (PA), and albumin (Alb) [32]. IL-6 is the main regulatory factor for APP synthesis and release, and studies have shown that IL-6 levels are positively correlated with APP levels. IL-6 can also regulate APP synthesis and release through the activation of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway [33]. Among these, CRP has been the most extensively studied in LLD. In a case-control study, Mishra et al. [34] found that CRP levels in patients with late-onset depression were 40% higher than in age-matched non-depressed individuals. Additionally, numerous studies have shown [35, 36, 37, 38] a strong positive correlation between CRP levels and the severity of depression. In a Mendelian randomization study [39], genome-wide association study (GWAS) data revealed shared genetic associations between CRP levels and individual depressive symptoms. Furthermore, several studies have determined that inflammatory markers like CRP, albumin (Alb), and pro-albumin (PA) are positively correlated with the severity of LLD [40, 41]. This association might be attributed to several factors. Firstly, elderly individuals with depression exhibit inflammation-related factors in their systems, which are often paired with the activation of inflammatory responses. Inflammatory factors can directly cause protein breakdown by activating the calpain-calpastin proteolytic system within cells [42]. In addition, inflammatory factors can increase the breakdown of insulin-like growth factor-1 [43], thereby inhibiting protein synthesis in the liver, which is clinically manifested as a decrease in negative APP levels. Second, LLD is often accompanied by physiological metabolic abnormalities, reduced appetite, and impaired digestive function, leading to a decrease in protein and energy intake, lowered serum Alb and PA levels, and eventually resulting in hypoalbuminemia [44]. Further research is necessary to investigate the role of APPs in LLD, as well as the complex relationships between APPs and other relevant factors, such as lifestyle factors in the elderly. Additionally, more consideration should be given to the potential value of APPs as therapeutic targets for the treatment and prevention of LLD, and further exploration of the feasibility of their application is warranted.

2.1.3 White Cell Ratios

The Neutrophil to Lymphocyte Ratio (NLR), Platelet to Lymphocyte Ratio (PLR), and Monocyte to Lymphocyte Ratio (MLR) have been extensively studied in patients with mental disorders. These ratios are considered practical, low-cost, and easily accessible novel inflammatory markers [45, 46]. Numerous studies have demonstrated that NLR, PLR, and MLR are elevated in patients with depression [47, 48, 49, 50]. Some research has suggested that MLR may be a risk factor for the development of depression [51], and PLR parameters might be more predictive than NLR in assessing the prognosis of major depression [52]. However, there is a relative scarcity of research focusing on the white blood cell ratio in the elderly depression population.

In 1964, Walford [53] proposed “The Immunologic Theory of Aging”, further developing and deriving the concept of immune aging. Immunosenescence of neutrophils occurs in a low-grade inflammatory environment, characterized by specific abnormalities in metabolism and function, along with increased apoptosis [54]. Hematopoietic stem cell senescence forms the basis of immunosenescence. Aging hematopoietic stem cells (HSCs) tend to differentiate into myeloid cells, while their ability to support lymphoid cell maturation decreases. This leads to a reduction in the number of T and B cell precursors with age [55]. It has been observed that an age-related B cell population accumulates in the peripheral blood as the body ages [56]. Additionally, the aging of the immune system has a complex relationship with inflammation. This relationship induces neuroinflammation and systemic inflammation through its interaction with the nervous system [57, 58] which in turn affects mood and may trigger depression. As the body ages, most immune cells exhibit characteristics of aging, impacting the number of neutrophils and lymphocytes in the peripheral blood. Future research needs to further explore the underlying mechanisms between blood cell ratios and LLD to evaluate the clinical potential of these biomarkers in the treatment and prevention of LLD.

2.1.4 NLRP3 Inflammasome

The NOD-, LRR-, and Pyrin domain-containing protein 3 (NLRP3) inflammasome is composed of NLRP3, apoptosis-associated speck-like protein containing CARD (ASC), and caspase-1 [59, 60, 61]. This complex plays a vital role in the aging process of various organs, including the thymus [62], kidney [63], and brain [64].

The activation signals for the NLRP3 inflammasome include two primary pathways: first, NF-κB activators [65] and IL-1β [66]; second, multiple risk signals or damage-associated molecular patterns (DAMPs) that have been identified as activators of the NLRP3 inflammasome. These include reactive oxygen species (ROS) [67], cholesterol crystals [68], urate crystals [69] and lipotoxic ceramides [70], all of which are endogenous metabolites that increase with age. Activation of the NLRP3 inflammasome is a response to the accumulation of various DAMPs and can induce systemic chronic inflammation during aging [71]. The mechanism underlying this process is that the NLRP3 inflammasome provides a platform for caspase-1 activation. Activated caspase-1 cleaves gasdermin D (GSDMD) to form pores in the cell membrane, leading to IL-1β and IL-18 leakage and pyroptosis. This causes an inflammatory storm, damages cells, and expands inflammation [72]. It can further lead to blood-brain barrier damage in the elderly [73], induce and aggravate neuroinflammation, and eventually result in depression [74].

Other studies have demonstrated that the activation of NLRP3 inflammasomes present in neurons, astrocytes, and microglia [75, 76] can cause the release of neurotoxic astrocytes and trigger a neurotoxic response. This has significant implications for the onset and progression of depression [77]. In light of the potential role of the NLRP3 inflammasome in geriatric depression, molecules regulating the NLRP3/ASC/caspase-1/GSDMD/IL-1β/IL-18 axis may hold clinical value in the development of antidepressant drugs [78]. Previous research has indicated that the dysregulation of the acetylation switch of the NLRP3 inflammasome is the origin of aging-related chronic inflammation, and NLRP3 deacetylation can prevent and target to reverse this inflammation [79]. In the future, comprehensive clinical studies on the correlation between the NLRP3 inflammasome and LLD in a larger population are needed. Such an approach may lead to a more profound understanding of the pathology of LLD, better stratification of patients, and enhanced improvement of treatment outcomes.

2.1.5 Brain-Derived Neurotrophic Factor (BDNF)

Brain-derived neurotrophic factor (BDNF) is a vital neurotrophic factor that plays a crucial role in brain function and the development of the nervous system [80]. Studies have shown that circulating levels of BDNF decrease with age in humans [81], and in the hippocampus and hypothalamus of male rats, BDNF expression has also been found to diminish with age [82].

Upon binding to high-affinity tropomyosin-associated kinase family (Trk) receptors, mature BDNF is internalized with its receptors and transported through axons to somatic cells. This initiates multiple effects in the nucleus [83], including increasing cell survival and differentiation, complicating dendritic spines [84], regulating synaptic plasticity [85], and rebuilding neural networks [86]. In the central nervous system (CNS), BDNF and downstream pro-survival pathways have been demonstrated to protect neurons from injury and enhance neuronal network reorganization following injury [87]. The dysfunction of synaptic transmission and plasticity is associated with damage to the BDNF/TrkB/CREB pathway, which can affect emotion, behavior, learning, and memory [88]. The decrease of BDNF during aging impacts BDNF-mediated antioxidant capacity, metabolic stress resistance, neurogenesis, and synaptic plasticity of nerve cells [89]. These changes can further influence mood, and a reduction in neurotransmission at hippocampal synapses and BDNF levels has been linked to an increased susceptibility to depression [90, 91]. Research has indicated that decreased BDNF-TrkB signaling during aging promotes microglial activation. Conversely, the upregulation of BDNF signaling inhibits microglial activation through the TrkB-Erk-CREB pathway, thereby reducing the production of inflammation [92]. This finding unveils the potential of BDNF in treating depression in the elderly.

By inhibiting microglial activation and associated inflammatory responses, BDNF may serve as a protective agent, helping to preserve and restore the function of the aging brain. This insight opens new avenues for understanding the complex interplay between neurotrophic factors, aging, and mental health, and may guide future therapeutic strategies for depression and other age-related neurological conditions. Hence, BDNF holds significant importance in the development and treatment of LLD [93, 94] and may act as a potential therapeutic target and biomarker [95]. Continued investigation into BDNF could pave the way for more effective treatment approaches.

2.1.6 Calcium (Ca2+) Homeostasis Imbalance

Intracellular Ca2+ serves as a crucial second messenger that orchestrates a myriad of vital cellular processes, notably influencing aging [96] and neurodegenerative diseases [97]. As neurons age, they experience calcium overload, characterized by heightened Ca2+concentrations [98], augmented Ca2+ storage, and an elevated transfer of Ca2+ from the endoplasmic reticulum (ER) to the mitochondria [99]. Aging or neurodegenerative conditions directly impact proteins responsible for maintaining Ca2+ balance. Notably, mass spectrometry has unveiled an age-correlated decline in the functional cysteine residues of sarco/endoplasmic reticulum ATPase (SERCA) [100], subsequently influencing the regulation of diverse physiological and pathological neuronal activities. Ca2+ conveys its effects through binding to calmodulin (CaM). This binding induces allosteric modifications in CaM, reshaping its interactions with target proteins such as kinases and phosphatases, and subsequently influencing neurotransmitter release, transcription factor regulation, axonal extension, and growth [101]. Research has highlighted that disruptions in ER calcium balance activate cellular stress mechanisms and are intrinsically tied to neurodegeneration and neuronal demise [102]. Aging-induced aberrations in Ca2+ signaling precipitate anomalies in synaptic plasticity and neuronal functionality, further advancing the onset of depression [103]. Further research has found that extracellular Ca2+ or other NLRP3 inflammasome activators can trigger intracellular Ca2+ signaling cascades through the interaction between the calcium-sensing receptor (CASR) and phospholipase C (PLC), thereby affecting NLRP3 [104], ultimately contributing to inflammation and the development of LLD. Investigations aiming to better understand the connection between Ca2+ homeostasis imbalance, inflammation, and LLD could also contribute to identifying potential preventive and therapeutic strategies for LLD through the regulation of Ca2+ homeostasis.

2.1.7 Other Proteins

In addition to the factors mentioned earlier, several protein molecules, such as S100 proteins and neutrophil gelatinase associated lipocalin (NGAL), are believed to play crucial roles in the pathogenesis of LLD. The levels of S100 proteins are closely related to the severity of depressive symptoms, and they can predict the response to antidepressant treatment [105, 106]. Plasma NGAL levels are elevated in elderly individuals with depression and are not influenced by the use of antidepressant medications or the age of onset, making it a potential new inflammatory biomarker for LLD [107, 108, 109]. Heat shock proteins, which regulate the immune system through their anti-inflammatory effects, may contribute to the treatment of LLD [110, 111]. while receptors such as Toll-like receptors (TLRs) and advanced glycation end products (AGEs), might also be involved in the condition’s development. The TLR signaling pathway is a potential common inflammatory pathway and could serve as a potential biomarker for identifying inflammatory subtypes of depression. Based on this, some antidepressant medications may exert anti-inflammatory effects by modulating TLR-dependent and independent inflammatory responses [112, 113]. In addition, changes in the receptor for AGEs (RAGE) signaling pathway may be related to the onset and exacerbation of late-life depressive symptoms [114, 115]. Furthermore, plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of plasminogen activation, has been shown to be involved in the pathogenesis of LLD through its regulation [116, 117, 118].

Factors like heat shock proteins, S100 proteins, TLRs, RAGE, NGAL, and PAI-1 are crucial in the development of LLD. Adjusting these factors could aid in devising more effective treatment strategies and offer novel perspectives and methods for the diagnosis and treatment of LLD (Table 1).

Table 1.Potential inflammatory pathways and mediators linked to late-life depression.
Category Mediators/Pathways
Cytokines Pro-inflammatory: IL-1β, IL-6, TNF-α, IL-8
Anti-inflammatory: IL-4, IL-10, IL-13, transforming growth factor β, adiponectin
Acute phase proteins (Apps) C-reactive protein (CRP), Albumin (Alb), pre-albumin (PA)
Janus kinase (JAK)
Signal transducer and activator of transcription (STAT)
White cell ratios neutrophil-to-lymphocyte ratio (NLR)
platelet-to-lymphocyte ratio (PLR)
monocyte-to-lymphocyte ratio (MLR)
hematopoietic stem cells (HSCs)
NLRP3 inflammasome apoptosis-associated speck-like protein containing(ASC)
NF-κB, damage-associated molecular patterns (DAMPs)
reactive oxygen species (ROS), gasdermin D (GSDMD)
NLRP3/ASC/caspase-1/GSDMD/IL-1β/IL-18 axis
Brain-derived neurotrophic factor (BDNF) kinase family (Trk), central nervous system (CNS)
BDNF/TrkB/CREB pathway, BDNF-TrkB signaling
TrkB-Erk-CREB pathway
Calcium (Ca2+) homeostasis imbalance endoplasmic reticulum (ER)
sarco/endoplasmic reticulum ATPase (SERCA), calmodulin (CaM)
calcium-sensing receptor (CASR), phospholipase C (PLC)
Other proteins S100 proteins, neutrophil gelatinase associated lipocalin (NGAL)
Toll-like receptors (TLRs), advanced glycation end products (AGEs)
plasminogen activator inhibitor-1 (PAI-1)
2.2 Abnormalities in Neurotransmitter Systems

Neuroinflammation refers to an inflammatory reaction that takes place within the central nervous system (including the brain and spinal cord) or the peripheral nervous system. The connection between aging, heightened inflammation, and chronic disease has become well established in recent scientific literature. In the elderly, serum levels of inflammatory molecules such as IL-6, TNF-α, and IL-18 are found to be increased [119, 120]. These inflammatory agents can penetrate the central nervous system either through the circulatory system or by crossing the blood-brain barrier, thereby initiating a neuroinflammatory response [121]. This understanding of the mechanisms underlying neuroinflammation provides valuable insights into the complex interactions between aging and inflammation, potentially guiding future research and therapeutic interventions for age-related neurological conditions.

Neuroinflammation may result in disturbances in neuronal function and neurotransmitter regulation. Previous consensus primarily emphasized the impact on serotoninergic and adrenergic systems. However, some studies propose that neuroinflammation is closely linked to dopamine [122], purinergic [123], and glutamatergic systems [124]. Dopamine is an important neurotransmitter that regulates mood and reward systems. Normal aging, along with age-related proinflammatory processes, is linked to a reduction in the function of dopaminergic molecules and a subsequent impairment of dopamine signaling [125, 126]. Inflammatory cytokines may negatively impact the dopaminergic system by restricting the availability of tetrahydrobiopterin (BH4), thereby reducing dopamine synthesis [127]. Additionally, these cytokines may hinder dopamine release and reuptake mechanisms [128, 129]. The decline in dopamine levels further contributes to the emergence of depressive symptoms. Research has shown that depression-like behaviors can be ameliorated by enhancing dopamine signaling [130]. This understanding of the intricate relationship between inflammation, dopamine, and depression may offer valuable insights for the development of targeted therapeutic strategies to address age-related mood disorders.

The purinergic system consists of various neurotransmitters and receptors, such as adenosine, ATP, P2X, and P2Y receptors. Inflammatory responses and cellular injuries can lead to the release of purine nucleotides, which subsequently activate purine receptors. Purinergic substances are known to stimulate the release of inflammatory mediators and regulate the activity of inflammation-related signaling pathways [131]. Specifically, A2A and P2X7Rs in astrocytes mediate the release of cytokines such as IL-1β and TNF-α, resulting in neuroinflammation [132] and contributing to the onset of depression.Several studies have highlighted that functional changes in the purinergic system, including abnormal adenosine levels, irregular expression of purine receptors, reduced adenosine triphosphate (ATP) release from astrocytes, and activation of P2X7 receptors in the medial prefrontal cortex and hippocampus, are significant factors in the development of depression [133, 134]. Given the role of purinergic signaling in inflammation and depression, research has indicated that purinergic P2X7 receptor antagonists may be vital in treating depression [135]. Targeting purinergic receptors could emerge as a potential strategy for treating depression in the elderly.

Glutamate, the primary excitatory neurotransmitter in the brain [136], plays a complex role in aging and depression. Aging-induced neuroinflammation activates microglial cells, which express metabotropic glutamate receptor 2 (mGluR2) on their surface. The binding of glutamate to microglial cells can trigger the release of inflammatory cytokines and nitric oxide, exerting toxic effects on neural circuits involved in emotion and cognition [137], and ultimately leading to depression. It has been proposed that a reduction in the glutamate to gamma-aminobutyric acid (GABA) ratio may be associated with a decrease in depressive symptoms [138]. The N-methyl-d-aspartate (NMDA) receptor, a member of the glutamate receptor family, plays a role in the transmission of neurotransmitter glutamate and the regulation of synaptic plasticity [139]. Ketamine, a glutamate NMDA receptor antagonist, has demonstrated rapid antidepressant effects [140], and its impact on synaptic plasticity has proven effective in treatment-resistant depression [140, 141, 142]. However, research focusing on the treatment of depression in the elderly remains scarce, and long-term safety data for elderly patients with depression are limited. More empirical research is needed to establish individualized safety dosages and treatment plans, ensuring that these promising avenues for treatment are explored with the necessary caution and rigor. The interplay between purinergic signaling, glutamate pathways, and depression presents a rich field for further investigation, with the potential to yield innovative therapeutic approaches for age-related mental health disorders.

3. Potential Mechanisms Linking Inflammation and LLD
3.1 Aging-DNA Damage

DNA damage is an unavoidable consequence of aging, and inflammation is a response triggered by such damage. DNA damage induces inflammatory responses by activating DNA damage response (DDR) pathways [143, 144]. Factors like age-related DNA damage accumulation, transposon activation [145, 146], cellular senescence [147], and persistent R loop accumulation [148] serve as catalysts. Through the activation of the cGAS-STING axis [149] and ATM [150] mediated NF-κB activation [151], signaling cascades such as NF-κB and IRF3 work together to activate type I interferons [152] and other inflammatory factors. This, in turn, triggers neuroinflammatory responses and contributes to the development of LLD.

A study on rats also suggests that the accumulation of DNA damage during aging may activate DDR [153], triggering neuroinflammatory responses and leading to LLD. Another study found a negative correlation between DNA methylation levels in the brain tissue of older adults and LLD, suggesting that DNA damage and inflammatory responses are related to the onset and development of LLD [154]. Investigating the interaction between DDR and inflammatory responses may help understand the pathogenesis of LLD and provide new treatment ideas.

3.2 Oxidative Stress and Mitochondrial Dysfunction

Oxidative stress and mitochondrial dysfunction may be closely related to the onset and progression of LLD. Evidence supporting this hypothesis includes studies showing abnormal manifestations of oxidative stress and mitochondrial dysfunction in patients with LLD [155, 156, 157]. The relationship between oxidative stress, mitochondrial dysfunction, and LLD may involve the activation of sterile inflammation and damage-associated molecular patterns (DAMPs) pathways [158]. The combined effect of oxidative stress and mitochondrial dysfunction leads to cellular damage, releasing DAMPs [159]. The release of DAMPs further stimulates immune responses, In addition to eliciting a type I interferon response, mitochondrial DNA activates Toll-like receptor 9 and NLRP3 inflammasome, ultimately leading to inflammation [160]. Furthermore, reactive oxygen species (ROS) may be harmful to neurons and synaptic transmission when the body is under high levels of oxidative stress and low antioxidants [161]. Increased oxidative stress may cause further mitochondrial damage, increased apoptosis, and ultimately inflammatory signaling [162, 163, 164], leading to depression-like behavior.

3.3 Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysfunction

The HPA axis is an essential neuroendocrine regulation system that modulates stress responses, helping the body maintain stability. When the HPA axis becomes dysregulated, stress responses become uncontrolled, affecting mental health and promoting the onset and worsening of depression. Recent studies have suggested that individuals with LLD may exhibit hyperactivity and sustained activation of the HPA axis [165, 166]. Excessive activation of the HPA axis leads to an overproduction of cortisol, which negatively impacts neurons and exacerbates LLD symptoms [167]. LLD may be related to the dysregulation of the HPA axis induced by inflammatory pathways. Many researchers have found elevated pro-inflammatory cytokines in patients with LLD, and studies have shown that increased IL-1, IL-6, and TNF-α can activate the HPA axis [168], leading to persistent activation of glucocorticoid receptors (GR), GR dysfunction, and loss of HPA axis negative feedback regulation, ultimately inducing or exacerbating the development of depression. Additionally, cytokines may stimulate indoleamine 2,3-dioxygenase (IDO) to participate in cortisol-mediated negative feedback inhibition of the HPA axis [169]. Recent research also indicates that gut microbiota can activate the HPA axis through microbial antigens, cytokines, and prostaglandins that cross the blood-brain barrier [170, 171], and evidence suggests that various microbial species can affect the production of corticosterone in the ileum, thus influencing HPA axis activity [172]. Consequently, HPA axis dysregulation mediated by inflammatory pathways might be a critical mechanism in LLD. Overactivation of the HPA axis worsens LLD symptoms and interacts with inflammation and gut microbiota, further affecting HPA axis function and creating a detrimental cycle. In-depth investigation of these mechanisms may help develop more effective treatment strategies.

3.4 Microglial Activation and Neuroinflammation

Block ML et al. [173] have shown that individuals with LLD exhibit significantly increased levels of microglial activation and neuroinflammation. Older adults often have vascular diseases and vascular risk factors that can cause chronic ischemic hypoxia in brain tissue, damaging oligodendrocytes, and persistently activating microglia and astrocytes. This results in the release of inflammatory factors such as IL-2, IL-6, IL-1β, TNF-α, and IFN-γ, inducing neuroinflammation [174, 175, 176]. Cytokines and other molecules released during inflammation, such as nitric oxide and free radicals, may produce direct cytotoxic effects, leading to neuronal damage [177]. Moreover, central inflammation may indirectly participate in the pathophysiology of depression through the kynurenine pathway. Inflammatory mediators can activate IDO, shifting tryptophan metabolism from serotonin production to kynurenine production, resulting in a decrease in 5-HT [178, 179], leading to depression. In addition, the imbalance of tryptophan metabolism in the kynurenine pathway may cause neurotransmitter imbalances, affecting neural plasticity and function [180].

On the other hand, neuroinflammation can cause changes in emotion-related monoamine neurotransmitters, HPA axis dysfunction, neuronal apoptosis, and downregulation of BDNF, ultimately leading to the onset of depression [181, 182, 183]. Furthermore, neuroinflammation may lead to blood-brain barrier disruptionand infiltration of inflammatory cells into the central nervous system [184], which may collectively contribute to the development and progression of LLD. A complex interplay exists between microglial activation, neuroinflammation, and LLD. Depression may trigger microglial activation and neuroinflammation, which in turn intensify depressive symptoms and impact neural plasticity and function. The presence of neuroinflammation could further aggravate depressive symptoms, forming a detrimental cycle. Additional research may enhance our understanding of the relationship between microglial activation, neuroinflammation, and LLD.

3.5 Gut-Brain Axis

Age-related changes in the gut microbiota can weaken the function of the intestinal barrier, leading to gut microbial dysbiosis [185, 186], which in turn triggers the release of bacterial products and promotes inflammation, damaging the body’s immune function [187]. The resulting inflammatory factors can increase the permeability of the blood-brain barrier [188], leading to neuroinflammation and subsequent depression. Furthermore, gut microbiota dysbiosis can result in changes in metabolic products such as short-chain fatty acids (SCFAs) and neurotransmitters (GABA, DA, NA, 5-HT) [189, 190], which may contribute to depression. SCFAs can also promote the repair of the intestinal mucosal barrier [191] and the blood-brain barrier [192]. Both SCFAs deficiency and intestinal inflammation can increase the permeability of the intestinal mucosal and blood-brain barriers, leading to neuroinflammation. Recent genomic research, coupled with Mendelian randomization studies, suggests that gut microbiota may play a role in regulating mood and anxiety, potentially via shared genetic pathways. A heightened genetic predisposition to depression has also been linked to this phenomenon [193, 194]. The gut-brain axis and gut microbiota, along with their role in inflammatory responses, could be one of the potential mechanisms underlying LLD. The gut microbiota can impact brain function and mood through several pathways, thus influencing the onset and advancement of LLD (Fig. 1).

Fig. 1.

Metabolic pathways potentially associated with late-life depression. For clarity and ease of reference, the full expansions of all acronyms presented in this figure are listed in alphabetical order as follows. 5-HT: 5-hydroxytryptamine (serotonin); ACTH: adrenocorticotropic hormone; ATM: ataxia telangiectasia mutated; BDNF: brain-derived neurotrophic factor; BBB: blood-brain barrier; cGAS-STING: cyclic gmp-amp synthase and stimulator of interferon genes; CRH: corticotropin-releasing hormone; DA: dopamine; DAMPS: damage-associated molecular patterns; GABA: gamma-aminobutyric acid; GC: glucocorticoid; GR: glucocorticoid receptors; HPA: hypothalamic-pituitary-adrenal; IDO: indoleamine 2,3-dioxygenase; IFN-γ: interferon-gamma; IL-1β: interleukin 1 beta; IL-2: interleukin 2; IL-6: interleukin 6; NA: noradrenaline; NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells; ROS: reactive oxygen species; SCFAS: short-chain fatty acids; TBK1: tank-binding kinase 1; TNF-α: tumor necrosis factor alpha; TRF3: Interferon regulatory factor 3.

4. Therapeutic Implications and Future Directions
4.1 Anti-Inflammatory Treatments and Their Impact on Depressive Symptoms

Treatment-resistant LLD (TRLLD) is a common problem, affecting up to one-third of patients. The inflammatory hypothesis of LLD provides a new direction for treatment [195]. Studies have shown that nonsteroidal anti-inflammatory drugs (NSAIDs) [196, 197, 198, 199], omega-3 fatty acids [200, 201, 202], statins [203, 204], cytokine inhibitors [205], corticosteroids [206, 207], and minocycline [208, 209, 210] have significant antidepressant effects when used as adjunctive therapies. Infliximab monotherapy also demonstrates some antidepressant effects [211]. Ketamine may alleviate depressive symptoms through various mechanisms, such as regulating inflammation-mediated cytokine dysregulation and neurotrophic factors [212, 213, 214]. Exercise and meditation therapies can indirectly reduce inflammation by lowering CRP levels in depressed patients, with good results [215]. Anti-inflammatory diets may potentially serve as an intervention for depression; pro-inflammatory diets are closely related to increased risk of depression diagnosis or symptoms, while anti-inflammatory diets mainly consist of fish, olive oil, and fresh vegetables and fruits [216, 217, 218]. Moreover, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may exert antidepressant, anti-inflammatory, and neuroprotective effects. Borsini’s [219] study provided evidence for LOX and CYP450-derived EPA/DHA bioactive lipid metabolites as molecular targets for human hippocampal neurogenesis and depression, emphasizing the importance of soluble epoxide hydrolase (sEH) inhibitors as potential therapeutic strategies for depression. However, some researchers found no statistically significant differences between vitamin D3 [220] and omega-3 fatty acid [221] treatments for depression compared to control groups. The contradictory results may be due to the heterogeneity of the depression subtypes included in the studies. Anti-inflammatory treatment has been recognized as beneficial in alleviating depressive symptoms, and the evidence supporting the antidepressant effect of such treatment opens the door to more personalized therapeutic plans for patients with depression. However, this approach is not without its challenges and limitations. The use of anti-inflammatory treatment, particularly in conjunction with antidepressants, has been a subject of controversy and debate [222, 223]. Nonsteroidal anti-inflammatory drugs (NSAIDs) have been associated with an increased risk of adverse cardiovascular events [224], and cytokine inhibitors have been linked to a heightened risk of infection [225]. These concerns underscore the complexity of implementing anti-inflammatory strategies in a clinical setting. Currently, there is a notable absence of large-sample, randomized, double-blind clinical studies focusing on anti-inflammatory treatment for geriatric depression. The need for extensive, high-quality research is paramount to assess the efficacy and safety of these therapies. The lack of such studies hampers our understanding of the precise impact of anti-inflammatory treatment on late-life depression and the best methods for administering this treatment.

Inflammation is a multifaceted biological process, involving numerous mediators and pathways. Developing drugs with high selectivity for specific inflammatory pathways or molecular targets presents a formidable challenge. Additionally, elderly patients with depression often suffer from comorbid physical ailments and may be on multiple medications, leading to potential interactions with anti-inflammatory drugs and subsequent safety risks. Therefore, a comprehensive evaluation of the benefits and risks of anti-inflammatory treatment is essential. More large-scale, randomized, double-blind clinical trials are needed to confirm the effectiveness of anti-inflammatory treatment in late-life depression. Such studies would not only contribute to our understanding of which patients are most likely to benefit from this approach but also help to refine treatment regimens, enhancing the likelihood of treatment success. In conclusion, while anti-inflammatory treatment offers promising avenues for depression therapy, especially in the elderly, careful consideration of its complexities and potential risks is vital. Continued research and clinical trials will be instrumental in unlocking its full potential and integrating it safely and effectively into the broader landscape of depression treatment.

4.2 Identifying Patient Subgroups That May Benefit from Anti-Inflammatory Treatment

With the deepening of the understanding of the role of anti-inflammatory treatment in depression, exploring the subgroups of late-life depression patients who may benefit from anti-inflammatory treatment can improve the treatment effect and reduce the risk of treatment. Related studies have confirmed that depressive patients with high inflammatory activity have poor response to antidepressant treatment, while the adjunctive treatment with anti-inflammatory drugs can significantly improve the clinical response rate of treatment-resistant depressive patients. Depressive patients with high baseline hs-CRP levels and treatment resistance have better antidepressant efficacy when combined with anti-inflammatory therapy [226]. Not all inflammatory factors have a direct correlation with antidepressant treatment responses. Some researchers have undertaken a systematic review and identified that inflammatory markers such as IL-6, CRP, and hsCRP show promise as indicators for predicting the effectiveness of treatments for resistant depression. In contrast, markers like IL-1β, IL-10, INF-γ, and TNF-α seem to lack predictive capabilities [227]. Many researchers have delved into ketamine, an antidepressant known for its anti-inflammatory properties, as a potential gauge for treatment outcomes. Some investigations suggest that elevated levels of IL-1β, IL-6, and IL-8 post-ketamine infusion [228, 229] correlate with improved treatment outcomes, though other studies present conflicting findings. Specific findings indicate that existing baseline measurements of IL-1β, IL-6, and IL-8 [230, 231] don’t necessarily predict a response to ketamine treatments. Moreover, patients with chronic inflammatory conditions like rheumatoid arthritis [232], cardiovascular ailments [233], chronic obstructive pulmonary disease (COPD) [234], inflammatory bowel disease [235], and liver cirrhosis [236] that are linked with depression may witness an improvement in depressive symptoms when treated with anti-inflammatory medications. Additionally, fluctuations in blood glucose and cholesterol could be intrinsically linked to responses to treatments like infliximab [237]. Presently, some study outcomes are inconclusive, possibly due to variances in sample sizes, methodologies, or the intricate nature of inflammatory responses in depression. Existing data is insufficient for establishing a consistent threshold for inflammation parameters that can steer these treatments. Yet, overall, inflammatory markers hold the potential not just to anticipate responses to antidepressant treatments, but also to forecast the success of supplementary anti-inflammatory therapies for depression. Future research endeavors should aim at elucidating the patient groups that stand to gain the most from specific anti-inflammatory interventions. The provided review paves the way for deeper inquiries into discerning subsets of elderly individuals with depression who might benefit from treatments targeting inflammation. Such insights could revolutionize precision treatments for late-life depression, bearing significant clinical value in enhancing treatment outcomes and minimizing associated risks.

5. Conclusions

LLD is a common mental illness with complex pathogenesis. Inflammatory responses are one of the potential mechanisms of LLD. Numerous studies have shown that inflammatory responses are significantly increased in patients with LLD, including Ca2+ homeostasis disturbances, cytokines, BDNF, NLR, PLR, MLR, and NLRP3 inflammasomes. These biomarkers are associated with late-life depressivesymptoms. Inflammatory responses affect brain function and emotions through various pathways, such as triggering neuroinflammation that influences neuronal plasticity, regulating neurotransmitter and hormone synthesis and release, ultimately impacting the onset and progression of LLD. Furthermore, oxidative stress and mitochondrial damage, dysbiosis of the gut microbiota, and aging-induced DNA damage-related changes can lead to increased inflammatory responses, further influencing the onset and progression of LLD.

This review delves into the intricate relationship between inflammation and late-life depression, with the goal of shedding light on potential applications in clinical practice:

(1) Identifying New Therapeutic Targets: By unraveling the mechanisms of inflammation in late-life depression, we can forge new therapeutic strategies that go beyond traditional antidepressants. This exploration opens avenues for the creation of more effective and personalized treatment plans. (2) Clinical Assessment of Inflammatory Markers: The detection of inflammatory markers may prove valuable in the clinical assessment and diagnosis of depression in later life. By analyzing these markers in the blood, healthcare providers can obtain objective indicators to gauge the level of inflammation, thereby aiding in informed treatment decisions. (3) Advancement of Personalized Medicine and Precision Psychiatry: The identification of patient subgroups that may benefit from anti-inflammatory treatment allows for more precise targeting of therapies to individual patient needs. This specificity enhances both the targeting and effectiveness of treatment, aligning with the principles of personalized medicine. (4) Holistic Health Management: The link between inflammation and late-life depression underscores the importance of a comprehensive approach to patient care. Beyond the focus on anti-inflammatory treatment, a holistic strategy should encompass maintaining a healthy lifestyle, providing psychological support, and implementing rehabilitation programs. These elements, when integrated, can enhance the overall treatment outcome and improve the quality of life for patients. In conclusion, the complex interplay between inflammation and late-life depression presents both challenges and opportunities. By embracing a multifaceted approach that includes novel therapeutic targets, precise diagnostics, personalized treatment, and holistic care, we can make strides in improving the management and outcomes of depression in the elderly. This review serves as a foundation for future research and clinical innovation, aiming to transform our understanding and treatment of this prevalent and impactful condition.

6. Limitations and Prospects

Although numerous studies have confirmed that inflammation plays a crucial role in the pathogenesis of LLD, the causal relationship between inflammation and LLD remains unclear. Furthermore, existing research faces many challenges and limitations. Firstly, there are inconsistent results and contradictory research findings, possibly due to factors such as sample size, research design, and methodology. Most relevant studies have used small-scale samples and primarily focused on specific populations, such as the elderly and patients with inflammatory diseases. In these cases, both the population size and the generalizability of research results are limited. Moreover, many related studies use cross-sectional designs, which cannot determine the causal relationship between inflammation and LLD. Heterogeneity in research findings may also lead to a lack of universal conclusions. Secondly, although inflammation appears to be related to LLD, the exact biological mechanism remains unclear. Many biomarkers lack sufficient specificity, and there is a shortage of effective biomarkers to assess the extent of the inflammatory response and treatment outcomes, which may also contribute to biases in research results. Lastly, there is currently a lack of personalized treatment plans for patients with LLD. Although some anti-inflammatory drugs have been developed for treating depression, their efficacy is uncertain, and adverse reactions may occur. Most research on inflammation and late-life depression is observational, and more interventional studies are needed to demonstrate the effectiveness and safety of inflammation-modulating treatments for LLD.

Therefore, future research areas need to include larger sample clinical trials, long-term follow-up studies, molecular biology, genomics, proteomics, and Mendelian randomization studies using interdisciplinary research methods. Further exploration of the relationship between inflammation and LLD and the establishment of more accurate prediction models and biomarkers are necessary to better understand the pathophysiological mechanisms of LLD. Various approaches and perspectives should be used to identify patient subgroups that may benefit from anti-inflammatory treatments, as well as to explore more effective treatment strategies and intervention measures.

Abbreviations

LLD, Late-life depression; NE, norepinephrine; 5-HT, 5-hydroxytryptamine; DA, dopamine; TNF-α, tumor necrosis factor; BBB, blood-brain barrier; MDD, major depressive disorder; APPs, Acute phase proteins; CRP, C-reactive protein; JAK, Janus kinase; STAT, Signal transducer and activator of transcription; GWAS, genome-wide association study; Alb, albumin; PA, pre-albumin; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; MLR, monocyte-to-lymphocyte ratio; HSCs, hematopoietic stem cells; NLRP3, NOD-, LRR-, and Pyrin domain-containing protein 3; ASC, apoptosis-associated speck-like protein containing; GSDMD, gasdermin D; BDNF, Brain-derived neurotrophic factor; Trk, kinase family; CNS, central nervous system; ER, endoplasmic reticulum; SERCA, sarco/endoplasmic reticulum ATPase; CaM, calmodulin; CASR, calcium-sensing receptor; PLC, phospholipase C; NGAL, neutrophil gelatinase associated lipocalin; TLRs, Toll-like receptors; AGEs, advanced glycation end products; PAI-1, plasminogen activator inhibitor-1; ATP, Adenosine triphosphate; BH4, tetrahydrobiopterin; mGluR2, metabotropic glutamate receptor 2; NMDA, N-methyl-d-aspartate; GABA, gamma-aminobutyric acid; DDR, DNA damage response; DAMPs, damage-associated molecular patterns; ROS, reactive oxygen species; HPA, Hypothalamic-Pituitary-Adrenal; GR, glucocorticoid receptors; IDO, indoleamine 2,3-dioxygenase; SCFAs, short-chain fatty acids; TRLLD, Treatment-resistant LLD; NSAIDs, nonsteroidal anti-inflammatory drugs; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; sEH, soluble epoxide hydrolase; NF-κB, Nuclear Factor-kappa B; IRF3, Interferon regulatory factor 3; COPD, chronic obstructive pulmonary disease.

Author Contributions

JX, MC, HS, and SW were instrumental in the conceptualization, drafting of the manuscript, supervision, and validation. JX, JY, MZ, WL, SZ, and HC were responsible for literature management and review. SW and HS undertook the tasks of reviewing, editing, and proofreading the manuscript. All authors contributed to editorial changes in the manuscript. All authors have reviewed and approved the final manuscript, ensuring significant participation and agreeing to be accountable for all facets of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We extend our heartfelt gratitude to Jingmei Zhong for her invaluable guidance, meticulous revisions, and diligent proofreading of this manuscript.

Funding

This research has been funded by The National Natural Science Foundation of China (Grant No. 81960259), The Program of Yunnan Clinical Research Center for Geriatric Diseases (Grant No. 2023A1010128), The Yunnan Provincial Clinical Research Center for Geriatric Diseases (Grant No. 2023YJZX-LN20) and The Dong Birong Expert Workstation Program of Yunnan (Grant No. 2023A4010302).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Rudnicka E, Napierała P, Podfigurna A, Męczekalski B, Smolarczyk R, Grymowicz M. The World Health Organization (WHO) approach to healthy ageing. Maturitas. 2020; 139: 6–11.
[2]
Lutz W, Sanderson W, Scherbov S. The coming acceleration of global population ageing. Nature. 2008; 451: 716–719.
[3]
Gundersen E, Bensadon B. Geriatric Depression. Primary Care. 2023; 50: 143–158.
[4]
Simkhada R, Wasti SP, Gc VS, Lee ACK. Prevalence of depressive symptoms and its associated factors in older adults: a cross-sectional study in Kathmandu, Nepal. Aging & Mental Health. 2018; 22: 802–807.
[5]
Andreas S, Schulz H, Volkert J, Dehoust M, Sehner S, Suling A, et al. Prevalence of mental disorders in elderly people: the European MentDis_ICF65+ study. The British Journal of Psychiatry. 2017; 210: 125–131.
[6]
Zenebe Y, Akele B, W/Selassie M, Necho M. Prevalence and determinants of depression among old age: a systematic review and meta-analysis. Annals of General Psychiatry. 2021; 20: 55.
[7]
Volkert J, Schulz H, Härter M, Wlodarczyk O, Andreas S. The prevalence of mental disorders in older people in Western countries - a meta-analysis. Ageing Research Reviews. 2013; 12: 339–353.
[8]
Horackova K, Kopecek M, Machů V, Kagstrom A, Aarsland D, Motlova LB, et al. Prevalence of late-life depression and gap in mental health service use across European regions. European Psychiatry. 2019; 57: 19–25.
[9]
Tang T, Jiang J, Tang X. Prevalence of depressive symptoms among older adults in mainland China: A systematic review and meta-analysis. Journal of Affective Disorders. 2021; 293: 379–390.
[10]
Byers AL, Yaffe K. Depression and risk of developing dementia. Nature Reviews. Neurology. 2011; 7: 323–331.
[11]
Hirschfeld RM. History and evolution of the monoamine hypothesis of depression. The Journal of Clinical Psychiatry. 2000; 61: 4–6.
[12]
Su JA, Chang CC, Yang YH, Chen KJ, Li YP, Lin CY. Risk of incident dementia in late-life depression treated with antidepressants: A nationwide population cohort study. Journal of Psychopharmacology. 2020; 34: 1134–1142.
[13]
Kok RM, Reynolds CF, 3rd. Management of Depression in Older Adults: A Review. JAMA. 2017; 317: 2114–2122.
[14]
Alexopoulos GS. Mechanisms and Treatment of Late-Life Depression. Focus. 2021; 19: 340–354.
[15]
Tedeschini E, Levkovitz Y, Iovieno N, Ameral VE, Nelson JC, Papakostas GI. Efficacy of antidepressants for late-life depression: a meta-analysis and meta-regression of placebo-controlled randomized trials. The Journal of Clinical Psychiatry. 2011; 72: 1660–1668.
[16]
Reynolds CF, 3rd, Dew MA, Pollock BG, Mulsant BH, Frank E, Miller MD, et al. Maintenance treatment of major depression in old age. The New England Journal of Medicine. 2006; 354: 1130–1138.
[17]
Giunta S, Wei Y, Xu K, Xia S. Cold-inflammaging: When a state of homeostatic-imbalance associated with aging precedes the low-grade pro-inflammatory-state (inflammaging): Meaning, evolution, inflammaging phenotypes. Clinical and Experimental Pharmacology & Physiology. 2022; 49: 925–934.
[18]
Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduction and Targeted Therapy. 2023; 8: 239.
[19]
Borsini A, Zunszain PA, Thuret S, Pariante CM. The role of inflammatory cytokines as key modulators of neurogenesis. Trends in Neurosciences. 2015; 38: 145–157.
[20]
Becher B, Spath S, Goverman J. Cytokine networks in neuroinflammation. Nature Reviews. Immunology. 2017; 17: 49–59.
[21]
Morimoto SS, Alexopoulos GS. Immunity, aging, and geriatric depression. The Psychiatric Clinics of North America. 2011; 34: 437–449, ix.
[22]
Alexopoulos GS, Morimoto SS. The inflammation hypothesis in geriatric depression. International Journal of Geriatric Psychiatry. 2011; 26: 1109–1118.
[23]
Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nature Reviews. Immunology. 2016; 16: 22–34.
[24]
Marsland AL, Gianaros PJ, Kuan DCH, Sheu LK, Krajina K, Manuck SB. Brain morphology links systemic inflammation to cognitive function in midlife adults. Brain, Behavior, and Immunity. 2015; 48: 195–204.
[25]
Baune BT, Smith E, Reppermund S, Air T, Samaras K, Lux O, et al. Inflammatory biomarkers predict depressive, but not anxiety symptoms during aging: the prospective Sydney Memory and Aging Study. Psychoneuroendocrinology. 2012; 37: 1521–1530.
[26]
Kim JM, Stewart R, Kim JW, Kang HJ, Bae KY, Kim SW, et al. Changes in pro-inflammatory cytokine levels and late-life depression: A two year population based longitudinal study. Psychoneuroendocrinology. 2018; 90: 85–91.
[27]
Dhabhar FS, Burke HM, Epel ES, Mellon SH, Rosser R, Reus VI, et al. Low serum IL-10 concentrations and loss of regulatory association between IL-6 and IL-10 in adults with major depression. Journal of Psychiatric Research. 2009; 43: 962–969.
[28]
Dilger RN, Johnson RW. Aging, microglial cell priming, and the discordant central inflammatory response to signals from the peripheral immune system. Journal of Leukocyte Biology. 2008; 84: 932–939.
[29]
Hayley S, Hakim AM, Albert PR. Depression, dementia and immune dysregulation. Brain. 2021; 144: 746–760.
[30]
Luning Prak ET, Brooks T, Makhoul W, Beer JC, Zhao L, Girelli T, et al. No increase in inflammation in late-life major depression screened to exclude physical illness. Translational Psychiatry. 2022; 12: 118.
[31]
Kim JM, Stewart R, Kim SW, Kim SY, Bae KY, Kang HJ, et al. Physical health and incident late-life depression: modification by cytokine genes. Neurobiology of Aging. 2013; 34: 356.e1–356.e9.
[32]
Kushner I. The phenomenon of the acute phase response. Annals of the New York Academy of Sciences. 1982; 389: 39–48.
[33]
Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. The New England Journal of Medicine. 1999; 340: 448–454.
[34]
Mishra D, Sardesai U, Razdan R. C-reactive protein level in late-onset depression: A case-control study. Indian Journal of Psychiatry. 2018; 60: 467–471.
[35]
Diniz BS, Reynolds CF, 3rd, Sibille E, Lin CW, Tseng G, Lotrich F, et al. Enhanced Molecular Aging in Late-Life Depression: the Senescent-Associated Secretory Phenotype. The American Journal of Geriatric Psychiatry. 2017; 25: 64–72.
[36]
Rozing MP, Veerhuis R, Westendorp RGJ, Eikelenboom P, Stek M, Marijnissen RM, et al. Inflammation in older subjects with early- and late-onset depression in the NESDO study: a cross-sectional and longitudinal case-only design. Psychoneuroendocrinology. 2019; 99: 20–27.
[37]
Sonsin-Diaz N, Gottesman RF, Fracica E, Walston J, Windham BG, Knopman DS, et al. Chronic Systemic Inflammation Is Associated With Symptoms of Late-Life Depression: The ARIC Study. The American Journal of Geriatric Psychiatry. 2020; 28: 87–98.
[38]
Lamers F, Milaneschi Y, Smit JH, Schoevers RA, Wittenberg G, Penninx BWJH. Longitudinal Association Between Depression and Inflammatory Markers: Results From the Netherlands Study of Depression and Anxiety. Biological Psychiatry. 2019; 85: 829–837.
[39]
Kappelmann N, Arloth J, Georgakis MK, Czamara D, Rost N, Ligthart S, et al. Dissecting the Association Between Inflammation, Metabolic Dysregulation, and Specific Depressive Symptoms: A Genetic Correlation and 2-Sample Mendelian Randomization Study. JAMA Psychiatry. 2021; 78: 161–170.
[40]
Hung KC, Wu CC, Chen HS, Ma WY, Tseng CF, Yang LK, et al. Serum IL-6, albumin and co-morbidities are closely correlated with symptoms of depression in patients on maintenance haemodialysis. Nephrology, Dialysis, Transplantation. 2011; 26: 658–664.
[41]
Pascoe MC, Skoog I, Blomstrand C, Linden T. Albumin and depression in elderly stroke survivors: An observational cohort study. Psychiatry Research. 2015; 230: 658–663.
[42]
Metwally E, Zhao G, Wang Q, Zhang YQ. Ttm50 facilitates calpain activation by anchoring it to calcium stores and increasing its sensitivity to calcium. Cell Research. 2021; 31: 433–449.
[43]
Salminen A, Kaarniranta K, Kauppinen A. Insulin/IGF-1 signaling promotes immunosuppression via the STAT3 pathway: impact on the aging process and age-related diseases. Inflammation Research. 2021; 70: 1043–1061.
[44]
Lobato ZM, Almeida da Silva AC, Lima Ribeiro SM, Biella MM, Santos Silva Siqueira A, Correa de Toledo Ferraz Alves T, et al. Nutritional Status and Adverse Outcomes in Older Depressed Inpatients: A Prospective Study. The Journal of Nutrition, Health & Aging. 2021; 25: 889–894.
[45]
Zahorec R. Ratio of neutrophil to lymphocyte counts–rapid and simple parameter of systemic inflammation and stress in critically ill. Bratislavske Lekarske Listy. 2001; 102: 5–14.
[46]
Özdin S, Böke Ö. Neutrophil/lymphocyte, platelet/lymphocyte and monocyte/lymphocyte ratios in different stages of schizophrenia. Psychiatry Research. 2019; 271: 131–135.
[47]
Bulut NS, Yorguner N, Çarkaxhiu Bulut G. The severity of inflammation in major neuropsychiatric disorders: comparison of neutrophil-lymphocyte and platelet-lymphocyte ratios between schizophrenia, bipolar mania, bipolar depression, major depressive disorder, and obsessive compulsive disorder. Nordic Journal of Psychiatry. 2021; 75: 624–632.
[48]
Demir S, Atli A, Bulut M, İbiloğlu AO, Güneş M, Kaya MC, et al. Neutrophil-lymphocyte ratio in patients with major depressive disorder undergoing no pharmacological therapy. Neuropsychiatric Disease and Treatment. 2015; 11: 2253–2258.
[49]
Hu J, Wang L, Fan K, Ren W, Wang Q, Ruan Y, et al. The Association Between Systemic Inflammatory Markers and Post-Stroke Depression: A Prospective Stroke Cohort. Clinical Interventions in Aging. 2021; 16: 1231–1239.
[50]
Demircan F, Gözel N, Kılınç F, Ulu R, Atmaca M. The Impact of Red Blood Cell Distribution Width and Neutrophil/Lymphocyte Ratio on the Diagnosis of Major Depressive Disorder. Neurology and Therapy. 2016; 5: 27–33.
[51]
Wei Y, Feng J, Ma J, Chen D, Chen J. Neutrophil/lymphocyte, platelet/lymphocyte and monocyte/lymphocyte ratios in patients with affective disorders. Journal of Affective Disorders. 2022; 309: 221–228.
[52]
Kayhan F, Gündüz Ş, Ersoy SA, Kandeğer A, Annagür BB. Relationships of neutrophil-lymphocyte and platelet-lymphocyte ratios with the severity of major depression. Psychiatry Research. 2017; 247: 332–335.
[53]
Walford RL. The immunologic theory of aging. The Gerontologist. 1964; 4: 195–197.
[54]
Dubey M, Nagarkoti S, Awasthi D, Singh AK, Chandra T, Kumaravelu J, et al. Nitric oxide-mediated apoptosis of neutrophils through caspase-8 and caspase-3-dependent mechanism. Cell Death & Disease. 2016; 7: e2348.
[55]
Ambrosi TH, Marecic O, McArdle A, Sinha R, Gulati GS, Tong X, et al. Aged skeletal stem cells generate an inflammatory degenerative niche. Nature. 2021; 597: 256–262.
[56]
Hao Y, O’Neill P, Naradikian MS, Scholz JL, Cancro MP. A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood. 2011; 118: 1294–1304.
[57]
Park J, Baik SH, Mook-Jung I, Irimia D, Cho H. Mimicry of Central-Peripheral Immunity in Alzheimer’s Disease and Discovery of Neurodegenerative Roles in Neutrophil. Frontiers in Immunology. 2019; 10: 2231.
[58]
Nie K, Zhang Y, Gan R, Wang L, Zhao J, Huang Z, et al. Polymorphisms in immune/inflammatory cytokine genes are related to Parkinson’s disease with cognitive impairment in the Han Chinese population. Neuroscience Letters. 2013; 541: 111–115.
[59]
Duncan JA, Bergstralh DT, Wang Y, Willingham SB, Ye Z, Zimmermann AG, et al. Cryopyrin/NALP3 binds ATP/dATP, is an ATPase, and requires ATP binding to mediate inflammatory signaling. Proceedings of the National Academy of Sciences of the United States of America. 2007; 104: 8041–8046.
[60]
Lu A, Magupalli VG, Ruan J, Yin Q, Atianand MK, Vos MR, et al. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell. 2014; 156: 1193–1206.
[61]
Boucher D, Monteleone M, Coll RC, Chen KW, Ross CM, Teo JL, et al. Caspase-1 self-cleavage is an intrinsic mechanism to terminate inflammasome activity. The Journal of Experimental Medicine. 2018; 215: 827–840.
[62]
Youm YH, Kanneganti TD, Vandanmagsar B, Zhu X, Ravussin A, Adijiang A, et al. The Nlrp3 inflammasome promotes age-related thymic demise and immunosenescence. Cell Reports. 2012; 1: 56–68.
[63]
Kaverina N, Schweickart RA, Chan GC, Maggiore JC, Eng DG, Zeng Y, et al. Inhibiting NLRP3 signaling in aging podocytes improves their life- and health-span. Aging. 2023; 15: 6658–6689.
[64]
Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013; 493: 674–678.
[65]
Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. Journal of Immunology. 2009; 183: 787–791.
[66]
Chen F, Jiang G, Liu H, Li Z, Pei Y, Wang H, et al. Melatonin alleviates intervertebral disc degeneration by disrupting the IL-1β/NF-κB-NLRP3 inflammasome positive feedback loop. Bone Research. 2020; 8: 10.
[67]
Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 2008; 320: 674–677.
[68]
Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010; 464: 1357–1361.
[69]
Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006; 440: 237–241.
[70]
Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nature Medicine. 2011; 17: 179–188.
[71]
Youm YH, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin A, et al. Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metabolism. 2013; 18: 519–532.
[72]
He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, et al. Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Research. 2015; 25: 1285–1298.
[73]
Liu WC, Wang X, Zhang X, Chen X, Jin X. Melatonin Supplementation, a Strategy to Prevent Neurological Diseases through Maintaining Integrity of Blood Brain Barrier in Old People. Frontiers in Aging Neuroscience. 2017; 9: 165.
[74]
Zhang Y, Liu L, Liu YZ, Shen XL, Wu TY, Zhang T, et al. NLRP3 Inflammasome Mediates Chronic Mild Stress-Induced Depression in Mice via Neuroinflammation. The International Journal of Neuropsychopharmacology. 2015; 18: pyv006.
[75]
Carranza-Aguilar CJ, Hernández-Mendoza A, Mejias-Aponte C, Rice KC, Morales M, González-Espinosa C, et al. Morphine and Fentanyl Repeated Administration Induces Different Levels of NLRP3-Dependent Pyroptosis in the Dorsal Raphe Nucleus of Male Rats via Cell-Specific Activation of TLR4 and Opioid Receptors. Cellular and Molecular Neurobiology. 2022; 42: 677–694.
[76]
Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019; 575: 669–673.
[77]
Li S, Fang Y, Zhang Y, Song M, Zhang X, Ding X, et al. Microglial NLRP3 inflammasome activates neurotoxic astrocytes in depression-like mice. Cell Reports. 2022; 41: 111532.
[78]
Alcocer-Gómez E, Casas-Barquero N, Williams MR, Romero-Guillena SL, Cañadas-Lozano D, Bullón P, et al. Antidepressants induce autophagy dependent-NLRP3-inflammasome inhibition in Major depressive disorder. Pharmacological Research. 2017; 121: 114–121.
[79]
He M, Chiang HH, Luo H, Zheng Z, Qiao Q, Wang L, et al. An Acetylation Switch of the NLRP3 Inflammasome Regulates Aging-Associated Chronic Inflammation and Insulin Resistance. Cell Metabolism. 2020; 31: 580–591.e5.
[80]
Ionescu-Tucker A, Tong L, Berchtold NC, Cotman CW. Inhibiting BDNF Signaling Upregulates Hippocampal H3K9me3 in a Manner Dependent On In Vitro Aging and Oxidative Stress. Frontiers in Aging. 2022; 3: 796087.
[81]
Erickson KI, Prakash RS, Voss MW, Chaddock L, Heo S, McLaren M, et al. Brain-derived neurotrophic factor is associated with age-related decline in hippocampal volume. The Journal of Neuroscience. 2010; 30: 5368–5375.
[82]
Silhol M, Bonnichon V, Rage F, Tapia-Arancibia L. Age-related changes in brain-derived neurotrophic factor and tyrosine kinase receptor isoforms in the hippocampus and hypothalamus in male rats. Neuroscience. 2005; 132: 613–624.
[83]
Curtis R, Adryan KM, Stark JL, Park JS, Compton DL, Weskamp G, et al. Differential role of the low affinity neurotrophin receptor (p75) in retrograde axonal transport of the neurotrophins. Neuron. 1995; 14: 1201–1211.
[84]
Messaoudi E, Ying SW, Kanhema T, Croll SD, Bramham CR. Brain-derived neurotrophic factor triggers transcription-dependent, late phase long-term potentiation in vivo. The Journal of Neuroscience. 2002; 22: 7453–7461.
[85]
Kang H, Schuman EM. A requirement for local protein synthesis in neurotrophin-induced hippocampal synaptic plasticity. Science. 1996; 273: 1402–1406.
[86]
Lee R, Kermani P, Teng KK, Hempstead BL. Regulation of cell survival by secreted proneurotrophins. Science. 2001; 294: 1945–1948.
[87]
Ghosh A, Carnahan J, Greenberg ME. Requirement for BDNF in activity-dependent survival of cortical neurons. Science. 1994; 263: 1618–1623.
[88]
Tong T, Chen Y, Hao C, Shen J, Chen W, Cheng W, et al. The effects of acupuncture on depression by regulating BDNF-related balance via lateral habenular nucleus BDNF/TrkB/CREB signaling pathway in rats. Behavioural Brain Research. 2023; 451: 114509.
[89]
Mattson MP, Maudsley S, Martin B. BDNF and 5-HT: a dynamic duo in age-related neuronal plasticity and neurodegenerative disorders. Trends in Neurosciences. 2004; 27: 589–594.
[90]
Calabrese F, Molteni R, Cattaneo A, Macchi F, Racagni G, Gennarelli M, et al. Long-Term duloxetine treatment normalizes altered brain-derived neurotrophic factor expression in serotonin transporter knockout rats through the modulation of specific neurotrophin isoforms. Molecular Pharmacology. 2010; 77: 846–853.
[91]
Hong C, Wang Z, Zheng SL, Hu WJ, Wang SN, Zhao Y, et al. Metrnl regulates cognitive dysfunction and hippocampal BDNF levels in D-galactose-induced aging mice. Acta Pharmacologica Sinica. 2023; 44: 741–751.
[92]
Wu SY, Pan BS, Tsai SF, Chiang YT, Huang BM, Mo FE, et al. BDNF reverses aging-related microglial activation. Journal of Neuroinflammation. 2020; 17: 210.
[93]
Dimitriadis M, van den Brink RHS, Comijs HC, Oude Voshaar RC. Prognostic effect of serum BDNF levels in late-life depression: Moderated by childhood trauma and SSRI usage? Psychoneuroendocrinology. 2019; 103: 276–283.
[94]
Gelle T, Samey RA, Plansont B, Bessette B, Jauberteau-Marchan MO, Lalloué F, et al. BDNF and pro-BDNF in serum and exosomes in major depression: Evolution after antidepressant treatment. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2021; 109: 110229.
[95]
Yang XJ, Zhao BC, Li J, Shi C, Song YQ, Gao XZ, et al. Serum NLRP3 Inflammasome and BDNF: Potential Biomarkers Differentiating Reactive and Endogenous Depression. Frontiers in Psychiatry. 2022; 13: 814828.
[96]
Kumar A, Bodhinathan K, Foster TC. Susceptibility to Calcium Dysregulation during Brain Aging. Frontiers in Aging Neuroscience. 2009; 1: 2.
[97]
Bezprozvanny I. Calcium signaling and neurodegenerative diseases. Trends in Molecular Medicine. 2009; 15: 89–100.
[98]
Xiong Y, Cheng Q, Li Y, Han Y, Sun X, Liu L. Vimar/RAP1GDS1 promotes acceleration of brain aging after flies and mice reach middle age. Communications Biology. 2023; 6: 420.
[99]
Calvo-Rodriguez M, Hernando-Perez E, Nuñez L, Villalobos C. Amyloid β Oligomers Increase ER-Mitochondria Ca2+ Cross Talk in Young Hippocampal Neurons and Exacerbate Aging-Induced Intracellular Ca2+ Remodeling. Frontiers in Cellular Neuroscience. 2019; 13: 22.
[100]
Sharov VS, Dremina ES, Galeva NA, Williams TD, Schöneich C. Quantitative mapping of oxidation-sensitive cysteine residues in SERCA in vivo and in vitro by HPLC-electrospray-tandem MS: selective protein oxidation during biological aging. The Biochemical Journal. 2006; 394: 605–615.
[101]
Wayman GA, Kaech S, Grant WF, Davare M, Impey S, Tokumitsu H, et al. Regulation of axonal extension and growth cone motility by calmodulin-dependent protein kinase I. The Journal of Neuroscience. 2004; 24: 3786–3794.
[102]
Williams BL, Lipkin WI. Endoplasmic reticulum stress and neurodegeneration in rats neonatally infected with borna disease virus. Journal of Virology. 2006; 80: 8613–8626.
[103]
Bennett MR. Synaptic P2X7 receptor regenerative-loop hypothesis for depression. The Australian and New Zealand Journal of Psychiatry. 2007; 41: 563–571.
[104]
Xi YH, Li HZ, Zhang WH, Wang LN, Zhang L, Lin Y, et al. The functional expression of calcium-sensing receptor in the differentiated THP-1 cells. Molecular and Cellular Biochemistry. 2010; 342: 233–240.
[105]
Navinés R, Oriolo G, Horrillo I, Cavero M, Aouizerate B, Schaefer M, et al. High S100B Levels Predict Antidepressant Response in Patients With Major Depression Even When Considering Inflammatory and Metabolic Markers. The International Journal of Neuropsychopharmacology. 2022; 25: 468–478.
[106]
Zhang P, Xiong Y, Wang B, Zhou Y, Wang Z, Shi J, et al. Potential value of serum brain-derived neurotrophic factor, vascular endothelial growth factor, and S100B for identifying major depressive disorder in knee osteoarthritis patients. Frontiers in Psychiatry. 2022; 13: 1019367.
[107]
Gouweleeuw L, Naudé PJW, Rots M, DeJongste MJL, Eisel ULM, Schoemaker RG. The role of neutrophil gelatinase associated lipocalin (NGAL) as biological constituent linking depression and cardiovascular disease. Brain, Behavior, and Immunity. 2015; 46: 23–32.
[108]
Naudé PJW, den Boer JA, Comijs HC, Bosker FJ, Zuidersma M, Groenewold NA, et al. Sex-specific associations between Neutrophil Gelatinase-Associated Lipocalin (NGAL) and cognitive domains in late-life depression. Psychoneuroendocrinology. 2014; 48: 169–177.
[109]
Naudé PJW, Eisel ULM, Comijs HC, Groenewold NA, De Deyn PP, Bosker FJ, et al. Neutrophil gelatinase-associated lipocalin: a novel inflammatory marker associated with late-life depression. Journal of Psychosomatic Research. 2013; 75: 444–450.
[110]
Borges TJ, Lang BJ, Lopes RL, Bonorino C. Modulation of Alloimmunity by Heat Shock Proteins. Frontiers in Immunology. 2016; 7: 303.
[111]
Wang H, Ba Y, Han W, Zhang H, Zhu L, Jiang P. Association of heat shock protein polymorphisms with patient susceptibility to coronary artery disease comorbid depression and anxiety in a Chinese population. PeerJ. 2021; 9: e11636.
[112]
Figueroa-Hall LK, Paulus MP, Savitz J. Toll-Like Receptor Signaling in Depression. Psychoneuroendocrinology. 2020; 121: 104843.
[113]
Takenaka Y, Tanaka R, Kitabatake K, Kuramochi K, Aoki S, Tsukimoto M. Profiling Differential Effects of 5 Selective Serotonin Reuptake Inhibitors on TLRs-Dependent and -Independent IL-6 Production in Immune Cells Identifies Fluoxetine as Preferred Anti-Inflammatory Drug Candidate. Frontiers in Pharmacology. 2022; 13: 874375.
[114]
Wu M, Zhao L, Wang Y, Guo Q, An Q, Geng J, et al. Ketamine Regulates the Autophagy Flux and Polarization of Microglia through the HMGB1-RAGE Axis and Exerts Antidepressant Effects in Mice. Journal of Neuropathology and Experimental Neurology. 2022; 81: 931–942.
[115]
Zhang L, Luo L, Xue L, Ran D, Yang F, Tang Q, et al. RAGE signaling pathway is involved in CUS-induced depression-like behaviors by regulating the expression of NR2A and NR2B in rat hippocampus DG. Experimental Neurology. 2023; 361: 114299.
[116]
Jiang H, Li X, Chen S, Lu N, Yue Y, Liang J, et al. Plasminogen Activator Inhibitor-1 in depression: Results from Animal and Clinical Studies. Scientific Reports. 2016; 6: 30464.
[117]
Lee SH, Shin C, Ko YH, Lee MS, Park MH, Pae CU, et al. Plasminogen Activator Inhibitor-1: Potential Inflammatory Marker in Late-life Depression. Clinical Psychopharmacology and Neuroscience. 2023; 21: 147–161.
[118]
Tsai SJ. The P11, tPA/plasminogen system and brain-derived neurotrophic factor: Implications for the pathogenesis of major depression and the therapeutic mechanism of antidepressants. Medical Hypotheses. 2007; 68: 180–183.
[119]
Pedersen M, Bruunsgaard H, Weis N, Hendel HW, Andreassen BU, Eldrup E, et al. Circulating levels of TNF-alpha and IL-6-relation to truncal fat mass and muscle mass in healthy elderly individuals and in patients with type-2 diabetes. Mechanisms of Ageing and Development. 2003; 124: 495–502.
[120]
Ferrucci L, Corsi A, Lauretani F, Bandinelli S, Bartali B, Taub DD, et al. The origins of age-related proinflammatory state. Blood. 2005; 105: 2294–2299.
[121]
Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009; 73: 768–774.
[122]
Liu Z, Qiu AW, Huang Y, Yang Y, Chen JN, Gu TT, et al. IL-17A exacerbates neuroinflammation and neurodegeneration by activating microglia in rodent models of Parkinson’s disease. Brain, Behavior, and Immunity. 2019; 81: 630–645.
[123]
Bao Y, Ledderose C, Seier T, Graf AF, Brix B, Chong E, et al. Mitochondria regulate neutrophil activation by generating ATP for autocrine purinergic signaling. The Journal of Biological Chemistry. 2014; 289: 26794–26803.
[124]
Weidinger A, Milivojev N, Hosmann A, Duvigneau JC, Szabo C, Törö G, et al. Oxoglutarate dehydrogenase complex controls glutamate-mediated neuronal death. Redox Biology. 2023; 62: 102669.
[125]
Kaasinen V, Vilkman H, Hietala J, Någren K, Helenius H, Olsson H, et al. Age-related dopamine D2/D3 receptor loss in extrastriatal regions of the human brain. Neurobiology of Aging. 2000; 21: 683–688.
[126]
Seaman KL, Smith CT, Juarez EJ, Dang LC, Castrellon JJ, Burgess LL, et al. Differential regional decline in dopamine receptor availability across adulthood: Linear and nonlinear effects of age. Human Brain Mapping. 2019; 40: 3125–3138.
[127]
Capuron L, Schroecksnadel S, Féart C, Aubert A, Higueret D, Barberger-Gateau P, et al. Chronic low-grade inflammation in elderly persons is associated with altered tryptophan and tyrosine metabolism: role in neuropsychiatric symptoms. Biological Psychiatry. 2011; 70: 175–182.
[128]
Zhu CB, Lindler KM, Owens AW, Daws LC, Blakely RD, Hewlett WA. Interleukin-1 receptor activation by systemic lipopolysaccharide induces behavioral despair linked to MAPK regulation of CNS serotonin transporters. Neuropsychopharmacology. 2010; 35: 2510–2520.
[129]
Morón JA, Zakharova I, Ferrer JV, Merrill GA, Hope B, Lafer EM, et al. Mitogen-activated protein kinase regulates dopamine transporter surface expression and dopamine transport capacity. The Journal of Neuroscience. 2003; 23: 8480–8488.
[130]
Scheggi S, Melis M, De Felice M, Aroni S, Muntoni AL, Pelliccia T, et al. PPARα modulation of mesolimbic dopamine transmission rescues depression-related behaviors. Neuropharmacology. 2016; 110: 251–259.
[131]
Engel T, Jiménez-Mateos EM, Diaz-Hernandez M. Purinergic Signalling and Inflammation-Related Diseases. Cells. 2022; 11: 3748.
[132]
D’Amico R, Fusco R, Siracusa R, Impellizzeri D, Peritore AF, Gugliandolo E, et al. Inhibition of P2X7 Purinergic Receptor Ameliorates Fibromyalgia Syndrome by Suppressing NLRP3 Pathway. International Journal of Molecular Sciences. 2021; 22: 6471.
[133]
Illes P, Rubini P, Yin H, Tang Y. Impaired ATP Release from Brain Astrocytes May be a Cause of Major Depression. Neuroscience Bulletin. 2020; 36: 1281–1284.
[134]
Xia M, Li Z, Li S, Liang S, Li X, Chen B, et al. Sleep Deprivation Selectively Down-Regulates Astrocytic 5-HT2B Receptors and Triggers Depressive-Like Behaviors via Stimulating P2X7 Receptors in Mice. Neuroscience Bulletin. 2020; 36: 1259–1270.
[135]
Recourt K, de Boer P, van der Ark P, Benes H, van Gerven JMA, Ceusters M, et al. Characterization of the central nervous system penetrant and selective purine P2X7 receptor antagonist JNJ-54175446 in patients with major depressive disorder. Translational Psychiatry. 2023; 13: 266.
[136]
Belardinelli P, König F, Liang C, Premoli I, Desideri D, Müller-Dahlhaus F, et al. TMS-EEG signatures of glutamatergic neurotransmission in human cortex. Scientific Reports. 2021; 11: 8159.
[137]
Taylor DL, Jones F, Kubota ESFCS, Pocock JM. Stimulation of microglial metabotropic glutamate receptor mGlu2 triggers tumor necrosis factor alpha-induced neurotoxicity in concert with microglial-derived Fas ligand. The Journal of Neuroscience. 2005; 25: 2952–2964.
[138]
Narayan GA, Hill KR, Wengler K, He X, Wang J, Yang J, et al. Does the change in glutamate to GABA ratio correlate with change in depression severity? A randomized, double-blind clinical trial. Molecular Psychiatry. 2022; 27: 3833–3841.
[139]
Buller AL, Larson HC, Schneider BE, Beaton JA, Morrisett RA, Monaghan DT. The molecular basis of NMDA receptor subtypes: native receptor diversity is predicted by subunit composition. The Journal of Neuroscience. 1994; 14: 5471–5484.
[140]
Murrough JW, Iosifescu DV, Chang LC, Al Jurdi RK, Green CE, Perez AM, et al. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. The American Journal of Psychiatry. 2013; 170: 1134–1142.
[141]
Dwyer JB, Landeros-Weisenberger A, Johnson JA, Londono Tobon A, Flores JM, Nasir M, et al. Efficacy of Intravenous Ketamine in Adolescent Treatment-Resistant Depression: A Randomized Midazolam-Controlled Trial. The American Journal of Psychiatry. 2021; 178: 352–362.
[142]
Nugent AC, Wills KE, Gilbert JR, Zarate CA, Jr. Synaptic potentiation and rapid antidepressant response to ketamine in treatment-resistant major depression: A replication study. Psychiatry Research. Neuroimaging. 2019; 283: 64–66.
[143]
Jin R, Niu C, Wu F, Zhou S, Han T, Zhang Z, et al. DNA damage contributes to age-associated differences in SARS-CoV-2 infection. Aging Cell. 2022; 21: e13729.
[144]
Yang B, Xie X, Wu Z, Lv D, Hu J, Chen Y, et al. DNA damage-mediated cellular senescence promotes hand-foot syndrome that can be relieved by thymidine prodrug. Genes & Diseases. 2022; 10: 2557–2571.
[145]
De Cecco M, Ito T, Petrashen AP, Elias AE, Skvir NJ, Criscione SW, et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature. 2019; 566: 73–78.
[146]
Simon M, Van Meter M, Ablaeva J, Ke Z, Gonzalez RS, Taguchi T, et al. LINE1 Derepression in Aged Wild-Type and SIRT6-Deficient Mice Drives Inflammation. Cell Metabolism. 2019; 29: 871–885.e5.
[147]
d’Adda di Fagagna F. Living on a break: cellular senescence as a DNA-damage response. Nature Reviews. Cancer. 2008; 8: 512–522.
[148]
Gan W, Guan Z, Liu J, Gui T, Shen K, Manley JL, et al. R-loop-mediated genomic instability is caused by impairment of replication fork progression. Genes & Development. 2011; 25: 2041–2056.
[149]
Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013; 339: 786–791.
[150]
Hinz M, Stilmann M, Arslan SÇ, Khanna KK, Dittmar G, Scheidereit C. A cytoplasmic ATM-TRAF6-cIAP1 module links nuclear DNA damage signaling to ubiquitin-mediated NF-κB activation. Molecular Cell. 2010; 40: 63–74.
[151]
Abe T, Barber GN. Cytosolic-DNA-mediated, STING-dependent proinflammatory gene induction necessitates canonical NF-κB activation through TBK1. Journal of Virology. 2014; 88: 5328–5341.
[152]
Iwanaszko M, Kimmel M. NF-κB and IRF pathways: cross-regulation on target genes promoter level. BMC Genomics. 2015; 16: 307.
[153]
Yousefzadeh MJ, Zhao J, Bukata C, Wade EA, McGowan SJ, Angelini LA, et al. Tissue specificity of senescent cell accumulation during physiologic and accelerated aging of mice. Aging Cell. 2020; 19: e13094.
[154]
Hüls A, Robins C, Conneely KN, De Jager PL, Bennett DA, Epstein MP, et al. Association between DNA methylation levels in brain tissue and late-life depression in community-based participants. Translational Psychiatry. 2020; 10: 262.
[155]
Somani A, Singh AK, Gupta B, Nagarkoti S, Dalal PK, Dikshit M. Oxidative and Nitrosative Stress in Major Depressive Disorder: A Case Control Study. Brain Sciences. 2022; 12: 144.
[156]
Ampo E, Mendes-Silva AP, Goncalves V, Bartley JM, Kuchel GA, Diniz BS. Increased Levels of Circulating Cell-Free mtDNA in the Plasma of Subjects With Late-Life Depression and Frailty: A Preliminary Study. The American Journal of Geriatric Psychiatry. 2022; 30: 332–337.
[157]
Vyas CM, Ogata S, Reynolds CF, 3rd, Mischoulon D, Chang G, Cook NR, et al. Lifestyle and behavioral factors and mitochondrial DNA copy number in a diverse cohort of mid-life and older adults. PLoS ONE. 2020; 15: e0237235.
[158]
Vringer E, Tait SWG. Mitochondria and Inflammation: Cell Death Heats Up. Frontiers in Cell and Developmental Biology. 2019; 7: 100.
[159]
Picca A, Lezza AMS, Leeuwenburgh C, Pesce V, Calvani R, Landi F, et al. Fueling Inflamm-Aging through Mitochondrial Dysfunction: Mechanisms and Molecular Targets. International Journal of Molecular Sciences. 2017; 18: 933.
[160]
Biasizzo M, Kopitar-Jerala N. Interplay Between NLRP3 Inflammasome and Autophagy. Frontiers in Immunology. 2020; 11: 591803.
[161]
Angelova PR, Abramov AY. Role of mitochondrial ROS in the brain: from physiology to neurodegeneration. FEBS Letters. 2018; 592: 692–702.
[162]
Bhatt S, Nagappa AN, Patil CR. Role of oxidative stress in depression. Drug Discovery Today. 2020; 25: 1270–1276.
[163]
Wang CH, Wu SB, Wu YT, Wei YH. Oxidative stress response elicited by mitochondrial dysfunction: implication in the pathophysiology of aging. Experimental Biology and Medicine. 2013; 238: 450–460.
[164]
Atagün Mİ, Canbek ÖA. A systematic review of the literature regarding the relationship between oxidative stress and electroconvulsive therapy. Alpha Psychiatry. 2022, 23: 47–56.
[165]
Rhebergen D, Korten NCM, Penninx BWJH, Stek ML, van der Mast RC, Oude Voshaar R, et al. Hypothalamic-pituitary-adrenal axis activity in older persons with and without a depressive disorder. Psychoneuroendocrinology. 2015; 51: 341–350.
[166]
Penninx BWJH, Beekman ATF, Bandinelli S, Corsi AM, Bremmer M, Hoogendijk WJ, et al. Late-life depressive symptoms are associated with both hyperactivity and hypoactivity of the hypothalamo-pituitary-adrenal axis. The American Journal of Geriatric Psychiatry. 2007; 15: 522–529.
[167]
Stokes PE. The potential role of excessive cortisol induced by HPA hyperfunction in the pathogenesis of depression. European Neuropsychopharmacology. 1995; 5: 77–82.
[168]
Dunn AJ. Cytokine activation of the HPA axis. Annals of the New York Academy of Sciences. 2000; 917: 608–617.
[169]
Myint AM, Kim YK. Network beyond IDO in psychiatric disorders: revisiting neurodegeneration hypothesis. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2014; 48: 304–313.
[170]
Vodička M, Ergang P, Hrnčíř T, Mikulecká A, Kvapilová P, Vagnerová K, et al. Microbiota affects the expression of genes involved in HPA axis regulation and local metabolism of glucocorticoids in chronic psychosocial stress. Brain, Behavior, and Immunity. 2018; 73: 615–624.
[171]
Farzi A, Fröhlich EE, Holzer P. Gut Microbiota and the Neuroendocrine System. Neurotherapeutics. 2018; 15: 5–22.
[172]
Misiak B, Łoniewski I, Marlicz W, Frydecka D, Szulc A, Rudzki L, et al. The HPA axis dysregulation in severe mental illness: Can we shift the blame to gut microbiota? Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2020; 102: 109951.
[173]
Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Progress in Neurobiology. 2005; 76: 77–98.
[174]
Beurel E, Toups M, Nemeroff CB. The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron. 2020; 107: 234–256.
[175]
Marshe VS, Maciukiewicz M, Hauschild AC, Islam F, Qin L, Tiwari AK, et al. Genome-wide analysis suggests the importance of vascular processes and neuroinflammation in late-life antidepressant response. Translational Psychiatry. 2021; 11: 127.
[176]
Benatti C, Blom JMC, Rigillo G, Alboni S, Zizzi F, Torta R, et al. Disease-Induced Neuroinflammation and Depression. CNS & Neurological Disorders Drug Targets. 2016; 15: 414–433.
[177]
Salvador AFM, Kipnis J. Immune response after central nervous system injury. Seminars in Immunology. 2022; 59: 101629.
[178]
Sublette ME, Postolache TT. Neuroinflammation and depression: the role of indoleamine 2,3-dioxygenase (IDO) as a molecular pathway. Psychosomatic Medicine. 2012; 74: 668–672.
[179]
Dantzer R. Role of the Kynurenine Metabolism Pathway in Inflammation-Induced Depression: Preclinical Approaches. Current Topics in Behavioral Neurosciences. 2017; 31: 117–138.
[180]
Miller AH. Conceptual confluence: the kynurenine pathway as a common target for ketamine and the convergence of the inflammation and glutamate hypotheses of depression. Neuropsychopharmacology. 2013; 38: 1607–1608.
[181]
Capuron L, Miller AH. Immune system to brain signaling: neuropsychopharmacological implications. Pharmacology & Therapeutics. 2011; 130: 226–238.
[182]
Miller AH, Haroon E, Raison CL, Felger JC. Cytokine targets in the brain: impact on neurotransmitters and neurocircuits. Depression and Anxiety. 2013; 30: 297–306.
[183]
Kronfol Z, Remick DG. Cytokines and the brain: implications for clinical psychiatry. The American Journal of Psychiatry. 2000; 157: 683–694.
[184]
Serna-Rodríguez MF, Bernal-Vega S, de la Barquera JAOS, Camacho-Morales A, Pérez-Maya AA. The role of damage associated molecular pattern molecules (DAMPs) and permeability of the blood-brain barrier in depression and neuroinflammation. Journal of Neuroimmunology. 2022; 371: 577951.
[185]
O’Toole PW, Jeffery IB. Gut microbiota and aging. Science. 2015; 350: 1214–1215.
[186]
Zapata HJ, Quagliarello VJ. The microbiota and microbiome in aging: potential implications in health and age-related diseases. Journal of the American Geriatrics Society. 2015; 63: 776–781.
[187]
Thevaranjan N, Puchta A, Schulz C, Naidoo A, Szamosi JC, Verschoor CP, et al. Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host & Microbe. 2017; 21: 455–466.e4.
[188]
Dion-Albert L, Cadoret A, Doney E, Kaufmann FN, Dudek KA, Daigle B, et al. Vascular and blood-brain barrier-related changes underlie stress responses and resilience in female mice and depression in human tissue. Nature Communications. 2022; 13: 164.
[189]
Wang Y, Li N, Yang JJ, Zhao DM, Chen B, Zhang GQ, et al. Probiotics and fructo-oligosaccharide intervention modulate the microbiota-gut brain axis to improve autism spectrum reducing also the hyper-serotonergic state and the dopamine metabolism disorder. Pharmacological Research. 2020; 157: 104784.
[190]
Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, et al. Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Frontiers in Cellular and Infection Microbiology. 2018; 8: 13.
[191]
Gasaly N, de Vos P, Hermoso MA. Impact of Bacterial Metabolites on Gut Barrier Function and Host Immunity: A Focus on Bacterial Metabolism and Its Relevance for Intestinal Inflammation. Frontiers in Immunology. 2021; 12: 658354.
[192]
Fock E, Parnova R. Mechanisms of Blood-Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids. Cells. 2023; 12: 657.
[193]
Luo J, Xu Z, Noordam R, van Heemst D, Li-Gao R. Depression and Inflammatory Bowel Disease: A Bidirectional Two-sample Mendelian Randomization Study. Journal of Crohn’s & Colitis. 2022; 16: 633–642.
[194]
Eijsbouts C, Zheng T, Kennedy NA, Bonfiglio F, Anderson CA, Moutsianas L, et al. Genome-wide analysis of 53,400 people with irritable bowel syndrome highlights shared genetic pathways with mood and anxiety disorders. Nature Genetics. 2021; 53: 1543–1552.
[195]
Fava M, Davidson KG. Definition and epidemiology of treatment-resistant depression. The Psychiatric Clinics of North America. 1996; 19: 179–200.
[196]
Mease P, Kuritzky L, Wright WL, Mallick-Searle T, Fountaine R, Yang R, et al. Efficacy and safety of tanezumab, NSAIDs, and placebo in patients with moderate to severe hip or knee osteoarthritis and a history of depression, anxiety, or insomnia: post-hoc analysis of phase 3 trials. Current Medical Research and Opinion. 2022; 38: 1909–1922.
[197]
Berk M, Woods RL, Nelson MR, Shah RC, Reid CM, Storey E, et al. Effect of Aspirin vs Placebo on the Prevention of Depression in Older People: A Randomized Clinical Trial. JAMA Psychiatry. 2020; 77: 1012–1020.
[198]
Iyengar RL, Gandhi S, Aneja A, Thorpe K, Razzouk L, Greenberg J, et al. NSAIDs are associated with lower depression scores in patients with osteoarthritis. The American Journal of Medicine. 2013; 126: 1017.e11–1017.e18.
[199]
Bauer IE, Green C, Colpo GD, Teixeira AL, Selvaraj S, Durkin K, et al. A Double-Blind, Randomized, Placebo-Controlled Study of Aspirin and N-Acetylcysteine as Adjunctive Treatments for Bipolar Depression. The Journal of Clinical Psychiatry. 2018; 80: 18m12200.
[200]
Kiecolt-Glaser JK, Belury MA, Andridge R, Malarkey WB, Glaser R. Omega-3 supplementation lowers inflammation and anxiety in medical students: a randomized controlled trial. Brain, Behavior, and Immunity. 2011; 25: 1725–1734.
[201]
Mischoulon D, Dunlop BW, Kinkead B, Schettler PJ, Lamon-Fava S, Rakofsky JJ, et al. Omega-3 Fatty Acids for Major Depressive Disorder With High Inflammation: A Randomized Dose-Finding Clinical Trial. The Journal of Clinical Psychiatry. 2022; 83: 21m14074.
[202]
Rapaport MH, Nierenberg AA, Schettler PJ, Kinkead B, Cardoos A, Walker R, et al. Inflammation as a predictive biomarker for response to omega-3 fatty acids in major depressive disorder: a proof-of-concept study. Molecular Psychiatry. 2016; 21: 71–79.
[203]
Stafford L, Berk M. The use of statins after a cardiac intervention is associated with reduced risk of subsequent depression: proof of concept for the inflammatory and oxidative hypotheses of depression? The Journal of Clinical Psychiatry. 2011; 72: 1229–1235.
[204]
Kim SW, Bae KY, Kim JM, Shin IS, Hong YJ, Ahn Y, et al. The use of statins for the treatment of depression in patients with acute coronary syndrome. Translational Psychiatry. 2015; 5: e620.
[205]
Tyring S, Gottlieb A, Papp K, Gordon K, Leonardi C, Wang A, et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. Lancet. 2006; 367: 29–35.
[206]
Arana GW, Santos AB, Laraia MT, McLeod-Bryant S, Beale MD, Rames LJ, et al. Dexamethasone for the treatment of depression: a randomized, placebo-controlled, double-blind trial. The American Journal of Psychiatry. 1995; 152: 265–267.
[207]
Bodani M, Sheehan B, Philpot M. The use of dexamethasone in elderly patients with antidepressant-resistant depressive illness. Journal of Psychopharmacology. 1999; 13: 196–197.
[208]
Nettis MA, Lombardo G, Hastings C, Zajkowska Z, Mariani N, Nikkheslat N, et al. The interaction between kynurenine pathway, suicidal ideation and augmentation therapy with minocycline in patients with treatment-resistant depression. Journal of Psychopharmacology. 2023; 37: 531–538.
[209]
Nettis MA, Lombardo G, Hastings C, Zajkowska Z, Mariani N, Nikkheslat N, et al. Augmentation therapy with minocycline in treatment-resistant depression patients with low-grade peripheral inflammation: results from a double-blind randomised clinical trial. Neuropsychopharmacology. 2021; 46: 939–948.
[210]
Husain MI, Chaudhry IB, Husain N, Khoso AB, Rahman RR, Hamirani MM, et al. Minocycline as an adjunct for treatment-resistant depressive symptoms: A pilot randomised placebo-controlled trial. Journal of Psychopharmacology. 2017; 31: 1166–1175.
[211]
McIntyre RS, Subramaniapillai M, Lee Y, Pan Z, Carmona NE, Shekotikhina M, et al. Efficacy of Adjunctive Infliximab vs Placebo in the Treatment of Adults With Bipolar I/II Depression: A Randomized Clinical Trial. JAMA Psychiatry. 2019; 76: 783–790.
[212]
Phillips JL, Norris S, Talbot J, Birmingham M, Hatchard T, Ortiz A, et al. Single, Repeated, and Maintenance Ketamine Infusions for Treatment-Resistant Depression: A Randomized Controlled Trial. The American Journal of Psychiatry. 2019; 176: 401–409.
[213]
Araújo-de-Freitas L, Santos-Lima C, Mendonça-Filho E, Vieira F, França RJAF, Magnavita G, et al. Neurocognitive aspects of ketamine and esketamine on subjects with treatment-resistant depression: A comparative, randomized and double-blind study. Psychiatry Research. 2021; 303: 114058.
[214]
Subramanian S, Haroutounian S, Palanca BJA, Lenze EJ. Ketamine as a therapeutic agent for depression and pain: mechanisms and evidence. Journal of the Neurological Sciences. 2022; 434: 120152.
[215]
Prakhinkit S, Suppapitiporn S, Tanaka H, Suksom D. Effects of Buddhism walking meditation on depression, functional fitness, and endothelium-dependent vasodilation in depressed elderly. Journal of Alternative and Complementary Medicine. 2014; 20: 411–416.
[216]
Lucas M, Chocano-Bedoya P, Schulze MB, Mirzaei F, O’Reilly ÉJ, Okereke OI, et al. Inflammatory dietary pattern and risk of depression among women. Brain, Behavior, and Immunity. 2014; 36: 46–53.
[217]
Vermeulen E, Brouwer IA, Stronks K, Bandinelli S, Ferrucci L, Visser M, et al. Inflammatory dietary patterns and depressive symptoms in Italian older adults. Brain, Behavior, and Immunity. 2018; 67: 290–298.
[218]
Detopoulou P, Panagiotakos DB, Antonopoulou S, Pitsavos C, Stefanadis C. Dietary choline and betaine intakes in relation to concentrations of inflammatory markers in healthy adults: the ATTICA study. The American Journal of Clinical Nutrition. 2008; 87: 424–430.
[219]
Borsini A, Nicolaou A, Camacho-Muñoz D, Kendall AC, Di Benedetto MG, Giacobbe J, et al. Omega-3 polyunsaturated fatty acids protect against inflammation through production of LOX and CYP450 lipid mediators: relevance for major depression and for human hippocampal neurogenesis. Molecular Psychiatry. 2021; 26: 6773–6788.
[220]
Okereke OI, Reynolds CF, 3rd, Mischoulon D, Chang G, Vyas CM, Cook NR, et al. Effect of Long-term Vitamin D3 Supplementation vs Placebo on Risk of Depression or Clinically Relevant Depressive Symptoms and on Change in Mood Scores: A Randomized Clinical Trial. JAMA. 2020; 324: 471–480.
[221]
Okereke OI, Vyas CM, Mischoulon D, Chang G, Cook NR, Weinberg A, et al. Effect of Long-term Supplementation With Marine Omega-3 Fatty Acids vs Placebo on Risk of Depression or Clinically Relevant Depressive Symptoms and on Change in Mood Scores: A Randomized Clinical Trial. JAMA. 2021; 326: 2385–2394.
[222]
Warner-Schmidt JL, Vanover KE, Chen EY, Marshall JJ, Greengard P. Antidepressant effects of selective serotonin reuptake inhibitors (SSRIs) are attenuated by antiinflammatory drugs in mice and humans. Proceedings of the National Academy of Sciences of the United States of America. 2011; 108: 9262–9267.
[223]
Uher R, Tansey KE, Dew T, Maier W, Mors O, Hauser J, et al. An inflammatory biomarker as a differential predictor of outcome of depression treatment with escitalopram and nortriptyline. The American Journal of Psychiatry. 2014; 171: 1278–1286.
[224]
Schjerning Olsen AM, Fosbøl EL, Lindhardsen J, Folke F, Charlot M, Selmer C, et al. Duration of treatment with nonsteroidal anti-inflammatory drugs and impact on risk of death and recurrent myocardial infarction in patients with prior myocardial infarction: a nationwide cohort study. Circulation. 2011; 123: 2226–2235.
[225]
Walters HM, Pan N, Lehman TJA, Adams A, Huang WT, Sitaras L, et al. A prospective study comparing infection risk and disease activity in children with juvenile idiopathic arthritis treated with and without tumor necrosis factor-alpha inhibitors. Clinical Rheumatology. 2015; 34: 457–464.
[226]
Papakostas GI, Shelton RC, Zajecka JM, Bottiglieri T, Roffman J, Cassiello C, et al. Effect of adjunctive L-methylfolate 15 mg among inadequate responders to SSRIs in depressed patients who were stratified by biomarker levels and genotype: results from a randomized clinical trial. The Journal of Clinical Psychiatry. 2014; 75: 855–863.
[227]
Yang C, Wardenaar KJ, Bosker FJ, Li J, Schoevers RA. Inflammatory markers and treatment outcome in treatment resistant depression: A systematic review. Journal of Affective Disorders. 2019; 257: 640–649.
[228]
Zhou Y, Wang C, Lan X, Li H, Chao Z, Ning Y. Plasma inflammatory cytokines and treatment-resistant depression with comorbid pain: improvement by ketamine. Journal of Neuroinflammation. 2021; 18: 200.
[229]
Kruse JL, Vasavada MM, Olmstead R, Hellemann G, Wade B, Breen EC, et al. Depression treatment response to ketamine: sex-specific role of interleukin-8, but not other inflammatory markers. Translational Psychiatry. 2021; 11: 167.
[230]
Kiraly DD, Horn SR, Van Dam NT, Costi S, Schwartz J, Kim-Schulze S, et al. Altered peripheral immune profiles in treatment-resistant depression: response to ketamine and prediction of treatment outcome. Translational Psychiatry. 2017; 7: e1065.
[231]
Kruse JL, Congdon E, Olmstead R, Njau S, Breen EC, Narr KL, et al. Inflammation and Improvement of Depression Following Electroconvulsive Therapy in Treatment-Resistant Depression. The Journal of Clinical Psychiatry. 2018; 79: 17m11597.
[232]
Li YC, Chou YC, Chen HC, Lu CC, Chang DM. Interleukin-6 and interleukin-17 are related to depression in patients with rheumatoid arthritis. International Journal of Rheumatic Diseases. 2019; 22: 980–985.
[233]
Halaris A. Inflammation-Associated Co-morbidity Between Depression and Cardiovascular Disease. Current Topics in Behavioral Neurosciences. 2017; 31: 45–70.
[234]
Du YJ, Yang CJ, Li B, Wu X, Lv YB, Jin HL, et al. Association of pro-inflammatory cytokines, cortisol and depression in patients with chronic obstructive pulmonary disease. Psychoneuroendocrinology. 2014; 46: 141–152.
[235]
Rukavishnikov GV, Neznanov NG, Mazo GE. Microbiota and autoimmune processes as potential therapeutic targets in comorbid depression and inflammatory bowel disease. Zhurnal Nevrologii i Psikhiatrii Imeni S.S. Korsakova. 2021; 121: 134–138. (In Russian)
[236]
Kronsten VT, Tranah TH, Pariante C, Shawcross DL. Gut-derived systemic inflammation as a driver of depression in chronic liver disease. Journal of Hepatology. 2022; 76: 665–680.
[237]
Cui W, Ning Y, Hong W, Wang J, Liu Z, Li MD. Crosstalk Between Inflammation and Glutamate System in Depression: Signaling Pathway and Molecular Biomarkers for Ketamine’s Antidepressant Effect. Molecular Neurobiology. 2019; 56: 3484–3500.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top