Next Article in Journal
Photocatalytic Degradation of Some Typical Antibiotics: Recent Advances and Future Outlooks
Next Article in Special Issue
Mechanisms Underlying Anti-Inflammatory and Anti-Cancer Properties of Stretching—A Review
Previous Article in Journal
Nucleoside Transport and Nucleobase Uptake Null Mutants in Leishmania mexicana for the Routine Expression and Characterization of Purine and Pyrimidine Transporters
Previous Article in Special Issue
Structural Analysis of Mitochondrial Dynamics—From Cardiomyocytes to Osteoblasts: A Critical Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interplay of Environmental, Individual and Genetic Factors in Rheumatoid Arthritis Provocation

1
Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia
2
Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
3
Institute of Environmental Sciences, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
4
Department of Immunology, CHU Toulouse, INSERM U1291, CNRS U5051, University Toulouse IIII, 31000 Toulouse, France
5
Department of Chemistry, University of South Florida, Tampa, FL 33620, USA
6
Mathematical Center, Sobolev Instiute of Mathematics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(15), 8140; https://doi.org/10.3390/ijms23158140
Submission received: 7 June 2022 / Revised: 18 July 2022 / Accepted: 19 July 2022 / Published: 23 July 2022
(This article belongs to the Special Issue Inflammation and the Musculoskeletal System)

Abstract

:
In this review, we explore systemization of knowledge about the triggering effects of non-genetic factors in pathogenic mechanisms that contribute to the development of rheumatoid arthritis (RA). Possible mechanisms involving environmental and individual factors in RA pathogenesis were analyzed, namely, infections, mental stress, sleep deprivation ecology, age, perinatal and gender factors, eating habits, obesity and smoking. The non-genetic factors modulate basic processes in the body with the impact of these factors being non-specific, but these common challenges may be decisive for advancement of the disease in the predisposed body at risk for RA. The provocation of this particular disease is associated with the presence of congenital loci minoris resistentia. The more frequent non-genetic factors form tangles of interdependent relationships and, thereby, several interdependent external factors hit one vulnerable basic process at once, either provoking or reinforcing each other. Understanding the specific mechanisms by which environmental and individual factors impact an individual under RA risk in the preclinical stages can contribute to early disease diagnosis and, if the factor is modifiable, might be useful for the prevention or delay of its development.

1. Introduction

Rheumatoid arthritis (RA) is a recognized model of multifactorial diseases, developing as an inappropriate response to environmental challenges in a genetically predisposed individual. Indeed, less than 30–60% of RA risk is due to the genetic propensity, whereas 40–70% is due to the influence of non-genetic factors [1]. Therefore, the study of the role of non-genetic factors in RA development is of great interest. In addition, in contrast to genetic predisposition, if we have a clear idea of the prognostic significance of these factors, we can manipulate at least some of them in order to prevent or delay RA onset. Even if we cannot eliminate or weaken the effect of any factor, providing clearer understanding of the pathogenic mechanisms of its effect on persons at risk can contribute to the development of approaches to safe therapy by inhibiting undesirable effects in the preclinical RA stages.
In this review, we discuss systematization of the knowledge regarding pathogenic mechanisms that have a triggering effect of non-genetic factors leading to RA development.

2. Methods

PubMed publications were selected using the following keyword pairs: “RA and risk factors” (since 1969–2022years–10,120 results, Figure 1) and “RA and environment” (1949–2022years–2933 results, Figure 1), as well as “RA and family aggregation”, “RA and ethnicity”, “RA and socioeconomic indicators”, “RA and perinatal factors”, “Perinatal programming and immune system”, “RA and gender”, “Immune system and sex hormones”, “RA and age”, “Immune system and age”, “RA and body mass index”, “Obesity and inflammation”, “RA and eating habits”, “RA and coffee”, “Caffeine and immune system”, “Alcohol and immune system”, “RA and alcohol”, “RA and smoking”, “RA and pollutants”, “RA and ecologic factors”, “RA and occupational hazards”, “Immune system and ecologic factors”, “RA and mental stress”, “Mental stress and pathogenesis”, “Mental stress and immune system”, “RA and sleep deprivation”, “Sleep deprivation and immune system”, and “RA and infections”.
After analyzing most of the publications, we selected the most informative of them and tried to create a complete picture of the possible relationships of RA and risk factors.

2.1. Facts of Genetic and Non-Genetic Factor Interplay

A detailed review of the genetic background of RA is not the subject of this article, but it makes sense to mention some of the particularly pertinent known facts.

2.1.1. RA-Associated HLA-DRB1

Allele variants (Shared epitopes, SE) show significance due to increased density of DR4 and DR1 molecule expression on antigen-presenting cells, leading to noticeable presentation of low-affinity peptides—a process of little importance for the development of a T-cell response at a more typical physiological density of DR molecules [2]. Presentation of low-affinity auto-peptides leads to autoreactive T cell activation.
A pronounced SE association and anti-cyclic citrullinated peptide antibodies (ACCP), but not rheumatoid factor (RF) production, is well documented. In a number of experimental models, the stability of SE complexes (DRB1*01and*04) with citrullinated proteins and other arthritogenic antigens was demonstrated to be increased in comparison with antigens not present in RA pathogenesis. And, in addition, the interaction of these complexes with T-lymphocyte receptors (TCR), as well as their influence on T-lymphocyte proliferation and Th1 cytokine response development, was demonstrably increased [3,4,5,6,7,8].
The affinity of TCR binding to HLA molecule–antigen complexes plays an important role in the lymphocyte maturation: too strong or too weak binding leads to a corresponding removal of clones. Despite their inconsistency, the experimental results demonstrated that interaction of these complexes with TCRs is optimal for the development of an immune response to citrullinated peptides and activation of proinflammatory cytokine synthesis.
The list of non-genetic factors triggering HLA gene expression:
  • Expression is due to appearance of citrullinated peptides in the infectious inflammatory sites [9];
  • Smoking leads to the exposure of citrullinated proteins in the respiratory tract [10];
  • Both SE and aryl hydrocarbon receptor (AhR)—a transcription factor mediating xenobiotic effects of many pollutants (including tobacco)—act as signal transduction ligands facilitating differentiation of Th17 cells and osteoclasts; nuclear factor kappaB-mediated synergistic interaction between the SE and AhR pathways was demonstrated in severe arthritis in mice [11].

2.1.2. Non-HLA Gene Polymorphisms

About 100 RA susceptibility loci were identified with SNP accumulation in T-cell and B-cell pathways, nuclear factor kappa B (NFkappaB) and Jak/STAT-signaling cascades, cytokine signaling pathways, proliferation and/or impaired hematopoietic and immune cells [12,13,14,15]. Collectively, RA-associated non-HLA gene polymorphisms are due to the insufficient inhibition of immune cell activity and to an excessive and long proinflammatory reply to external challenges.
The list of non-genetic factors triggering expression of these genes:
4.
Infections due to activation of NFkappaB and Jak/STAT-signaling cascades, cytokine signaling pathways, immune cells proliferation, appearance of citrullinated peptides in the infectious inflammatory site [16,17,18,19,20,21];
5.
Smoking due to citrullinated proteins exposure in lungs [22,23];
6.
Miscarriage, complicated pregnancy, childbirth leading to a Th1-immune reaction flare-up and proinflammatory cytokine expression [24];
7.
Obesity linked to increased proinflammatory cytokine levels [25];
8.
Mental stress and sleep deprivation due to increased proinflammatory cytokine levels [26,27,28].

2.2. Complex Non-Genetic Factors

2.2.1. Family Aggregation

The obvious components of RA family clustering are genetic risk and shared environmental factors [29]. Heritability of ACCP-positive RA is~50% and of ACCP-negative RA is ~20% [29]; therefore, genetic and non-genetic factors might be of relatively equal importance for seropositive RA development, whereas non-genetic factors might be more important for seronegative RA. Study of the contribution of SE, 76 other gene SNPs and non-genetic factors determined to be shared by the family members (smoking, alcohol intake, parity, silica exposure, BMI, fatty fish consumption, socio-economic status) demonstrated: (1) SE together with 76 SNPs explained about 20% of the familial risk [30] and (2) studied non-genetic risk factors did not explain any significant part of the familial risk in both seropositive and negative RA. Therefore, family history of RA remains an important independent risk factor for RA. Many non-genetic factors besides the ones studied might be due to the RA family clustering with an essential cumulative effect.
The list of triggering and protective family associated factors:
  • Genetic;
  • Infections;
  • Microbiome;
  • Lifestyle;
  • Ecology (when living together);
  • Unknown??

2.2.2. Ethnicity

The overall adult RA prevalence is approximately 0.5%; however, considerable variation exists between ethnicities, with a higher prevalence observed in those of European ancestry (0.3–1.1%) than in those of Asian ancestry (0.1–0.5%) [31,32], and even higher prevalence (approximately 5–7%) has been reported in Native American populations [33]. That is due to the known genetic differences between the populations as well as the obvious but not-well-studied differences in non-genetic factors (lifestyle, eating habits, socio-economical differences and so on).
Some examples of the known ethnic genetic specificity [15,34,35,36]:
  • HLA-DRB1 SEs: in Europeans—HLADRB*0401, HLA-DRB*0404, HLA-DRB*0101,
  • in Asians—HLADRB*0405, HLA-DRB*0101, HLA-DRB*0901
  • PTPN22 SNPs confirmed for Europeans, rare in Asians
  • TRAF1/C5 confirmed for Europeans, suggested for Asians
  • STAT4 confirmed both for Europeans and Asians
  • CD40 confirmed for Europeans, no associations for Asians
  • CTLA4 confirmed both for Europeans and Asians
  • PADI4 suggested for Europeans, confirmed for Asians
  • FCRL3 suggested for Europeans, confirmed for Asians
The list of suggested ethnicity-associated triggering and protecting factors:
  • Genetic;
  • Lifestyle;
  • Eating habits;
  • Mentality;
  • Socioeconomic indicators;
  • Climate.

2.2.3. Socioeconomic Indicators

A lower educational level ≤ 8 years (OR = 2.42, 95% CI 1.18–4.93 vs. University degree) and living in poverty contributed (OR = 2.96, 95% CI 1.88–4.65, p < 0.001) to RA development [37,38].
An a priori list of provoking/protective socioeconomic factors:
  • Professional activity—lower status—more professional occupational hazards?
  • Lifestyle
  • Food habits
  • Lower status—fewer opportunities to protect/improve one’s health
  • Less education—less understanding of the importance of regular examinations
  • Living condition quality—overcrowding, uncomfortable housing—infections

2.3. Anthropological Indicators

2.3.1. Perinatal and Early Life Factors

Twin studies demonstrated a high sensitivity of the fetal genome to the effects of various factors of maternal origin.
In monozygotic twins sharing a chorionic shell during the intrauterine development, the pattern of gene methylation differed from that in pairs who developed in two separate chorionic shells. In adulthood, this can lead to significant differences in organs and system functioning in these genetically identical individuals [39]. The impact of perinatal factors on the development of cardiovascular diseases, type II diabetes, and obesity has been demonstrated [40,41,42]. The bulk of publications on this issue was devoted to allergic diseases. A correlation ofTh1/Th2/Th17cytokine levels, pro-inflammatory cytokine/chemokine indexes, and IgE levels in peripheral blood of pregnant women at 34 weeks of gestation and in umbilical cord blood of their newborns was demonstrated, persisting one year after delivery [43]. A relationship of cytokine levels in mother and her child and a number of disorders of fetal immune system maturation might be due to epigenetic modeling during prenatal development and abnormal gene expression [44,45]. Such programming of the fetal immune system under conditions of a complicated pregnancy might occur in families with a history of RA. Infections during the first year of life were associated with increased risk of seronegative RA (OR = 2.6). Maternal smoking during pregnancy changes the pattern of newborn gene methylation; the abnormally methylated gene clusters included significant impact on RA pathways related to cell cycle, angiogenesis, T cell regulation and other white blood cell related pathways, which increased the risk for RA development [46,47]. Peculiarities of prenatal development and pregnancy abnormalities were not necessarily closely tied to future RA development and even might be protective. For example, low birth weight (OR = 0.7), being small for the gestational age (OR = 0.5) and preterm birth (OR = 0.6) were shown to have a borderline protective effect [48].
List of hypothetical links of perinatal factors and RA risk in predisposed individuals:
  • Intrauterine fetal infections ⇒ immune system programming
  • Maternal infection during pregnancy ⇒ programming of fetal immune system
  • Complicated pregnancy and childbirth ⇒ fetal immune system programming
  • Microbiome—maternal origin, breast feeding/bottle feeding
  • Maternal smoking
  • Early life infection ⇒ programming the immature immune system and impact formation of the microbiome

2.3.2. Gender Associated Factors

Sexual dimorphism in expression of human rheumatic diseases involves immunomodulatory effects of post-puberty levels of sex steroid hormones [49]. Due to the presence of hormone receptors on immune cells [50], sex hormones might influence different aspects of immune system functioning and potentially affect the risk, activity and progression of RA [51]. Sex hormones undergo complex dynamics during pregnancy, childbirth, the postpartum period, and the onset of menopause [52,53,54,55].
These multidirectional changes in sex hormones are superimposed on the effects of adrenocorticotropic hormone and cortisol with well-known impacts on the immune system and inflammation. Moreover, the levels of corticosteroid production are different in normal and complicated pregnancies [55]. Low maternal cortisol may influence the fetal hypothalamic–pituitary–adrenal axis (HPA) and disease patterns later in life following a complicated pregnancy [56]. A negative association between maternal cortisol and infant birth weight was demonstrated [57].
We tried to link sex-related events and associated hormonal fluctuations with the impact on the development of RA (Supplementary Table S1).
Repeated normal pregnancies, childbirth, postpartum breastfeeding with normal feedback in the network of sex hormones and glucocorticoids ⇒ bursts of production of these hormones with a protective effect ⇒ reduced RA risk;
Normal pregnancy with hidden feedback impairments in the sex hormone network and glucocorticoids ⇒ RA onset within 1 year after delivery;
Adverse pregnancy as a clinical manifestation of feedback impairments in the network of sex hormones and glucocorticoids ⇒ RA triggering
Menopause ⇒ decrease in hormone levels and their protective effects ⇒ RA risk

2.3.3. Age

RA affects any age group, with the peak occurring during the sixth decade of life [58,59]. The triggering role of age might be due to the decline in host immunity with promotion of immune reactivity to self-antigens, weakened antimicrobial immunity, predisposition for tissue inflammation and osteoarthritis due to chronic microtraumas of joint tissues [60,61].
Impact of age on the immune system and joint tissues [61]:
  • Weakened antimicrobial immunity;
  • Susceptibility to respiratory infections;
  • Reactivation of chronic viral infections;
  • Predisposition for tissue inflammation;
  • Osteoarthritis due to chronic microtraumas of joint tissues.

2.3.4. Body Mass Index

Obesity is recognized as a chronic low-grade systemic inflammatory state, NFκappaB and NLRP3 inflammasome signaling pathways and proinflammatory cytokine transcription being the key events [25].
Obesity effects on the immune system [62] due to RA triggering: effector/memory T-cell population increase, impoverishment of TCR diversity, M2 to M1 macrophage shift in adipose, increase in the TH1 cell population and decrease in the Treg cell numbers in adipose, NF-kB cascade activation in PBMCs, increased production of MIF, IL-6, TNF-a, MMP-9 mRNA expression in PBMCs, inhibition of phagocytic activity of PBMCs and increased infection susceptibility [62]. Chronic joint tissue microtraumas is additive.
Link of BMI and RA:
  • Overweight ⇒ chronic low grade, systemic inflammatory state
  • Microbiome structure peculiarities
  • Overweight ⇔ stress ⇔ sleep deprivation
  • Overweight ⇒ infections
  • Overweight ⇒ chronic joint tissue microtraumas

2.3.5. Eating Habits

Diets containing fatty fish (marine omega-3 fatty acid, OA3FA) are beneficial for RA prevention and reduce the need for nonsteroidal anti-inflammatory drugs [63]. RA development is associated with lower levels of OA3FA, especially in ACCP-positive persons at risk. Three mechanisms were described: (I) OA3FA inhibits proinflammatory eicosanoid production (prostaglandinE2 and leukotriene B4), which in turn inhibits NFkappaB activation and proinflammatory interleukin production, ultimately resulting in autoreactive B cells and synoviocyte activation and maturation; (II) OA3FA promotes cell surface receptor expression (vascular cell adhesion molecule (VCAM)-1, and PPARγ in monocytes) or repression (CCL5,HLA-DQ/DR), leading to reduction of Th17 differentiation, enhancement of Foxp3+CD4+T cells regulatory functions, promotion of M2 polarization and (III) interaction between OA3FA and the SE is suspected as an inverse association between OA3FA concentrations, and SE+RF (OR = 0.26), or SE+ACCP (OR = 0.44) was reported in RA risk cohorts. The other promising approach for protection might be the Mediterranean diet with low saturated fat content, contributing to a decrease in RA activity [64]. Though the evidence is insufficient to unconditionally include this diet in the recommendations for RA patients, at least there is reason for more in-depth research, since it was demonstrated that this diet may lower RA severity due to antioxidant and anti-inflammatory properties [65,66].
The earliest preclinical events in RA development were proven to start at the barrier tissue mucosal membranes, including the gut. Although the data on RA association with microbiome features are extremely contradictory, mainly due to small sample sizes, a priori it can be assumed that microbiome–local immunity and barrier cell interactions play a role in the earliest RA stages. So, dietary manipulation of the microbiome might be effective in suppressing undesirable events that take place in early RA stages, when the process has not yet gone awry [67,68,69,70].
On the other hand, a Western diet with higher intake of red and processed meats, sweets, and refined grain associated with elevated inflammatory markers might increase RA risk [71].

2.4. Salt and RA

Of interest is the number of publications discussing the problem linking RA to sodium chloride consumption. This problem has not been sufficiently studied with regard to RA, probably due to significant difficulties in technical methods. However, the in vitro effects of excess sodium chloride concentrations on immune cells suggest a potential trigger role in the pathogenesis of RA.
Indeed, salt increased migration of macrophage-like RAW264.7 cells in a dose-dependent fashion with no migratory response noted in isotonic or hypotonic media controls, or other osmo-active agents [72], and high NaCl concentrations promoted IFNβ production and signaling in human and mouse macrophages and inhibited M2 macrophage activation [73,74]. Dendritic cells treated with high NaCl concentrations produced increased levels of interleukin-1β and promoted T cell production of cytokines IL-17A and interferon gamma (IFN-γ) [75].Induction of Th17 response due to the activation of glucocorticoid kinase 1 (SGK1), a serine/threonine kinase, governing Na(+) transport and salt (NaCl) homeostasis in the cells, on the one hand, is critical for regulating IL-23R expression and, on the other hand, for stabilizing the TH17 cell phenotype and controlling the balance between regulatory Treg and Th17 cells [76]. Therefore, salt promotes the suppression of Treg proliferation and function as well [76,77]. So, in vitro high salt concentrations demonstrated a perceptible effect, including triggering Th-17 responses, which is relevant in RA pathogenesis.
Next, experiments in rodent models demonstrated more severe clinical and histological arthritis in the high-salt diet and collagen-induced arthritic mice, together with higher numbers of Th17 cells among splenocytes and increased expression of synovial and intestinal IL-17, compared to control collagen-induced arthritis (CIA) mice fed a normal salt diet [78]. Sehnert et al. studied the impact of a low-salt vs. a normal and a high-salt diet on the CIA and K/BxN serum transfer-induced arthritis (STIA) [79]. In both mouse models, a low-salt diet significantly decreased arthritis severity, with less inflammatory joint infiltrates and cartilage breakdown. Moreover, IL-1 receptor blocking (in STIA) reduced complement-fixing anti-CII IgG2a levels and decreased anti-CII IgG2a/IgG1 ratios (as a more Th2-like response). In addition, reduced IL-17 and monocyte chemoattractant protein-1 levels (in CIA) were demonstrated.
The results of studies on the effect of a salt regimen in humans are less convincing. On the one hand, a low-salt (6 g/d for months) vs. a high-salt (12 g/d) diet in healthy individuals led to decrease in the blood monocyte number and reduced production of proinflammatory cytokines (IL-6 and IL-23), along with an enhanced ability to produce anti-inflammatory cytokine IL-10 [80]. On the other hand, no impact of a salt regimen on Treg/Th17 lymphocyte levels and in vitro Th17 cell differentiation was revealed in both healthy individuals and RA patients [81].
To further focus on RA, a comparison of synovial fluid between RA patients and OA patients revealed that Na+ and IL-17 were more abundant in RA synovial fluid, indicating a possible link of salt intake and rheumatoid inflammation [78]. Urinary Na/K ratio positively and significantly correlated with DAS28-ESR [82]. High daily sodium intake (estimated from foods plus added salt) in 18,555 individuals, including 392 self-reported rheumatoid arthritis individuals, showed a significant association with rheumatoid arthritis (OR = 1.5) [83]. In a nested case-control study, including 386 individuals who had stated their dietary median of 7.7 years before the onset of symptoms of RA and 1886 matched controls, revealed a link of high sodium intake with a more than doubled increased RA risk, but only among smokers [OR = 2.26) [84]. Jiang et al. revealed that high sodium chloride consumption enhances the effects of smoking in ACCP RA development [85].
Dynamic testing of RA in patients who were on a low-salt diet for 3 weeks (with analysis of urinary sodium excretion for confirmation adherence to the dietary regimen) and then returned to a normal salt diet revealed a trend toward a reduction in the Th17 cell frequencies and a countertrend for Treg and return to the previous levels after 2 weeks of following the normal salt regime with no significant apoptosis or altered proliferation [86]. It should be noted that publications on the problem of the link between RA and the salt regime are currently scarce. Apparently, given some encouraging findings, further research is needed for confirmation.

2.4.1. Coffee

The results of the RA link with coffee consumption are ambiguous. On the one hand an increased risk was demonstrated (>4 cups) for RF-positive and ACCP-positive RA [87,88,89,90]. On the other hand, 1–8 cups per day appeared to have a protective effect [91]. At that level, multiple anti-inflammatory effects of caffeine are quite consistent with the protective effects of coffee: anti-inflammatory cytokines increased and pro-inflammatory cytokines decreased production, neutrophils and monocytes chemotaxis were inhibited, and B-cell antibody production decreased [92]. In addition, some coffee components possess antioxidant properties [93,94].

2.4.2. Alcohol

On the one hand, moderate alcohol consumption reduces RA risk [66], but on the other hand, patients who had stopped drinking due to their illness or a desire to improve their health had worse physical functioning and higher levels in pain-related variables [95,96].
Studies of the mechanisms of alcohol consumption are quite consistent with the protective effect on RA development. Healthy premenopausal women having one drink/day had a significant serum increase in estradiol [97], with its remarkable anti-inflammatory effects. Alcohol consumption decreases systemic inflammation and inflammatory arthritis [98], diminishes response to immunogens and suppresses pro-inflammatory cytokine synthesis [99]. Alcohol addition to drinking water inhibits clinical signs of arthritis and joint destruction in mice and upregulation of testosterone production due to the decrease in NF-kB activation, cytokine/chemokine production and leukocyte chemotaxis [98].

2.4.3. Smoking

The association of smoking and RA is also well-known. In a Swedish cohort of 277,777 male construction workers, chronic smoking was associated with increased RA risk (RR = 2.1) [100]. The pathophysiological mechanisms involve increased oxidative stress, apoptosis, development of a systemic proinflammatory state, autoantibody production, and interplay with genetic factors (Supplementary Table S2).

2.5. Burden of Society

2.5.1. Residence

The incidence of RA in Taiwan cities is higher than in rural areas [101]. In the Polish cohorts, the physical condition of the sick urban citizens was more severe compared to the villagers [102]. Swedish studies did not reveal significant differences in RA incidence in sparsely populated areas and cities [103]. Conflicting results may be due to the ethnogenetic characteristics of cohorts, environmental differences, size of cities (in a Swedish study, a settlement with 25,000 inhabitants was considered a large city). The reasons for the heavier course of RA in cities may be exposure to traffic pollution, overcrowding, and more intensive contacts due to increased incidence of infections, the more intense rhythm of city life and provocation of mental stress.

2.5.2. Mental Stress

Eustress is defined as favorable stress that invokes development of a balanced protective adaptive response (including Th1→Th2 shift of immune responses) to stressors in order to mobilize the body’s mental and physical reserves to resolve the situation [104]. This attests at least to the absence of any direct involvement of stress in provoking RA, that being a Th1-mediated disease. On the other hand, stress-induced shifts in the immune system may have serious consequences for susceptibility to infections [105,106]. A number of studies demonstrated the association of stress with acute respiratory rhinovirus infections, respiratory syncytial virus, Coronavirus 229E type, parvoviruses and herpesvirus infection reactivation [107,108]. All these viruses are known to be RA triggers [109,110,111]. However, in our opinion, only repetitive episodes of trivial infections are significant for provoking and maintaining RA activity. It is unlikely that a single infectious episode provoked by a normal anti-stress adaptive response in turn might provoke RA onset, unless that episode is the last straw in a series of immune system provocations of the genetically predisposed individual.
With excessive or chronic stress, or if there is a deviation in stress susceptibility, decompensated stress (distress) develops, and this is precisely the situation that triggers somatic disease development.
Analysis in the GWAS Catalog [112] revealed 14 matches of RA-and-depression-associated SNPs (Figure 2).
The comparison of RA and distress mechanisms indicates the possibility of mutual potentiation of these conditions (Supplementary Table S3) [113]. That is (1) typical for RA excessive NFkappaB pathway signaling and proinflammatory cytokine hyperproduction leading to the abolition or weakening of the Th1→Th2 shift in acute stress and promoting distress; (2) in RA, the features of HPA functioning with reduced production of glucocorticoids by the adrenal glands and, in some patients, a decrease in glucocorticoid receptor expression with the development of steroid resistance were demonstrated, leading to distress development via abolition or weakening of the Th1→Th2 immune shift in acute stress; (3) sympathetic/parasympathetic tone imbalance, detected in the preclinical RA stage, may contribute to distress development and (4) a reduced or even completely absent immune system cell response to catecholamines in RA may cancel or weaken theTh1→Th2 immune shift. Therefore, a detailed analysis reveals a certain synergy of triggering mechanisms of distress and RA. Minimal and unobtrusive stress for most individuals, for those with risk for RA, might provoke a prolonged pro-inflammatory cytokine production. Thus, the mental stress influence on RA development depends on the intensity of the stress, its duration, and individual characteristics of the HPA axis, which are obviously determined by genetic and epigenetic factors.
Hypothetical link of mental stress and RA:
  • Residence (urban/rural)
  • Psychological discomfort at work and at home—occupational hazards
  • ⇓⇓⇓
  • Distress ⇒ infections ⇒ RA
  • NFkappaB signaling pathway and proinflammatory cytokine production ⇒ RA

2.5.3. Sleep Deprivation

Regulation of the wake–sleep cycle is controlled by multiple neurochemical and molecular biological cascades, in which, in addition to the neuromodulatory system (in particular low molecular weight neurotransmitters and neuropeptides), other factors are involved, including HPA, the NFkappaB signaling system, cytokines, in regulating protein synthesis, protecting the brain tissue from oxidative or glutamatergic stress [114]. In connection with the problems discussed in this review, it is important to note that wake–sleep cycle fluctuations in HPA hormone levels (in particular, cortisol and cytokines) occur not only in the central nervous system but at the periphery as well. While the endocrine system was long believed to obey circadian rhythms and to be involved in sleep regulation, recently it has become evident that various immune system components also have a circadian rhythmicity. Therefore, both brain and peripheral cytokines are included in wake–sleep regulation, the most pro-inflammatory cytokines likely being somnogenic, whereas most anti-inflammatory cytokines are not. Peak activities of the pituitary hormones, prolactin and growth hormone production as well as another circadian pineal gland hormone—melatonin—and decrease in cortisol production occur overnight due to nocturnal sleep with the subsequent morning decrease in activity of nocturnal hormones and peak activity of cortisol after awakening [115,116]. All of these hormones are known to regulate immune system activity. Enhanced nocturnal prolactin, GH and melatonin concentrations as well as low cortisol levels and the following morning changes of the hormone activity are synergistically due to a Th1 shift at night and return to a Th1/Th2 balance of immune reactions in the morning [115,117]. These regularities were demonstrated not just for cytokine production, but in particular for the circadian rhythms of phagocytes and NK activities [114,117]. Normal circadian balance fluctuations might have a beneficial effect on the anti-infectious immune reactions. In humans the primary response to viral antigens following vaccination was enhanced by sleep [118,119,120]. A variety of disturbances in sleep duration and quality are due to an imbalance in the complex interactions of HPA hormones, melatonin and their receptors [116,117,118,119,120,121,122]. In particular, recurrent short sleep sessions were associated with a flatter diurnal cortisol pattern [121]. The disturbed circadian hormone modulation of the immune system leads to alterations in inflammatory gene expression [123] and upregulation of transcriptional pathways (e.g., NFkappaB) responsible for the inflammatory response [26], even in individuals in the absence of other health problems. So, sleep deprivation is associated with increased levels of the inflammatory markers C-RP [27,28]. Sleep deprivation may be due to the alteration of immune cell functions [114]: decrease in cell numbers of NK and other lymphocyte subsets, decrease in NK lytic activity, and decreased phagocytosis. Sleep deprivation might slow the catabolism of IgG. So, sleep disorders are fraught with a decrease in the effectiveness of anti-infective immunity. The proinflammatory shifts and disturbances in the HPA hormone activity caused by sleep deprivation bring these states closer to induction of mental stress and obesity. This triad is a fairly frequent combination [116,124,125]. Another important aspect is that short sleep duration and poor sleep efficiency in both mid and late pregnancy were associated with higher levels of IL-6 [126], and so, it might be a risk factor for adverse pregnancy outcomes [127]. In turn, besides the peculiarities of diurnal fluctuations of HPA hormones and melatonin, sleep disorders may be due to lifestyle factors and the aging process [128,129]. Therefore, sleep deprivation itself, as well as in connection with mental stress, obesity and with triggered adverse pregnancy outcomes, might play a role in provoking RA and its activity.
Sleep deprivation probability in persons under RA risk seems to be rather high. Various forms of clinically significant sleep disturbance were found in over 60% of RA patients [130,131]. Poor sleep quality correlated with greater pain severity, joint disability and RA activity [130,132,133,134]. High sleep deprivation incidence in RA is likely linked to the features of circadian ACTH, cortisol and prolactin, as well as melatonin level fluctuations [135,136,137,138], due to the demonstrated defect of HPA functioning.
Link of sleep deprivation and RA:
Sleep deprivation ⇔ Mental stress ⇔ Obesity ⇒ Proinflammatory shift ⇒ RA Sleep deprivation ⇔ Mental stress ⇒Increased susceptibility to infections ⇒ RA
Sleep deprivation ⇒ Increased risk of adverse pregnancy ⇒RA

2.6. Infections

Microorganisms and viruses are undoubtedly major RA triggers. Possible mechanisms triggering host autoimmune responses by pathogens are well known: molecular mimicry, epitope spreading, polyclonal lymphocyte activation, bystander activation and viral persistence [139,140,141,142]. All of these processes were demonstrated in RA [80]. The fact is that these mechanisms are beneficial phenomena, contributing to the immune system’s ability to attack multiple pathogens.
Therefore, the problem is not in these processes as such but is due to: (1) immune system disability to cope quickly enough with an infectious challenge due to RA-associated gene SNPs and abnormal downregulation of genes of some innate and adaptive immune system factors [110,143,144,145,146,147]; (2) imperfect control of the anti-infectious response, namely, inadequate lymphocyte activity modulation (PTPN22, CTLA-4,BTLA and other RA-associated gene SNPs), as well as an enrichment of RA-associated gene SNPs in NFkappaB and JAK/STAT signaling cascades due to the excessive proinflammatory response [14,148,149,150] and (3) the impaired sanitation of the infectious inflammatory focus from pathogenic and self-modified molecules due to pro-oxidant and antioxidant factor imbalance and inadequate activity of several enzymes involved in remodeling of the extracellular matrix [151,152,153]. Our long-term observations of RA patients and their first-degree relatives demonstrated that the persons under RA risk suffer from frequent and prolonged trivial infections. The peak of infections was observed within two years before RA onset and decreased in three years after RA onset. Nevertheless, infections continued playing a role in maintaining RA activity in the advanced stage of the disease. Increased incidence of excessive bacterial colonization in feces, urine, skin and nasopharynx samples of advanced RA patients without signs of infection indicates that the effectiveness of anti-infection resistance is relative, and a delicate balance may be disturbed. Both the whole set of infections carried over a year, and certain infections (purulent upper respiratory tract infections, acute and chronic tonsillitis exacerbations, skin infections and episodes of HSV infection reactivation) were demonstrated to actually be involved in RA triggering and persistence of RA activity [110,111,154].
Hypothetical link of infections and RA:
RA-associated HLA alleles ⇒ susceptibility to certain trivial infections ⇒ increased infection incidence and duration [155,156,157,158,159,160,161].
Technogenic burden (exotoxins, occupational hazards, mental stress, overcrowding) ⇒ infections;
Imbalanced anti-infection resistance ⇒ increased susceptibility to trivial infections ⇒ unbalanced anti-infection response (deficiency of some factors of innate immunity, prolonged pro-inflammatory cytokine production) ⇒ infectious pathogen persistence ⇒ lymphocyte inhibitory signaling systems deficiency⇒ impossibility of timely folding of the immune response;
Certain infections ⇒ posttranslational protein modifications in inflammatory sites ⇒ violation of readjustment inflammatory focus (imbalance of pro-oxidant and antioxidant factors, GST, MMP-3) ⇒ lymphocyte inhibitory signaling system deficiency ⇒ persistent RF, ACCP, anti-carbamylated protein antibodies (anti-CarP) production.

Occupational Hazards and Eco-Toxicants

The more severe RA in urban settings may be due to the more technogenic atmospheric emissions. RA incidence was inversely proportional to the distance of the residence from high-emission motorways [162,163]. Increased RA risk linked with silica, carbon monoxide, ozone, vapor, gas, dust and fume exposure, and cosmetic-associated mineral oil was demonstrated in a bulk of studies [164,165,166,167,168,169].
Another bulk of experiments revealed the impact of various ecotoxicants on basic intracellular processes, contributing to RA development (Supplementary Table S4). In particular, ecotoxicant-provoked oxidative stress might be important for moving a person at risk from one preclinical stage to another and to RA onset [170,171]. There are at least two mechanisms: (1) generated ROS stimulate activation NFkappaB signaling [172,173,174] and (2) oxidative stress can provoke protein carbamylation and the appearance of anti-Carp antibodies, intensively studied as RA prognostic markers [175,176].
The individual dispersive efficacy of ecotoxicant degradation mechanisms—a so-called “syndrome of nonspecific increased chemical susceptibility”, in particular, is manifested in immune disorders. The said dispersion might be due to the SNPs of detoxication system enzyme genes (Supplementary Table S5). At least three mutations were found to be RA-associated. Study of the function of the detoxication system can reveal an important link in the provocation of RA by non-genetic factors. It may turn out that ecotoxicant concentrations considered to be safe for the general population are fraught with RA provocation in persons at risk (Figure 3).

3. Concluding Remarks

The non-genetic factors modulate basic processes in the body (Figure 4), with the impact of these factors on the body being absolutely non-specific. The impact of these ordinary nonspecific factors—trivial infections, ecotoxicants in concentrations not exceeding the permissible values, such commonplace events as pregnancy, delivery, menopause—on the loci minoris resistentia of a body at risk of RA is to initiate an imbalanced protective adaptive response, which may provoke the disease onset. The most significant and well-known weak links are undoubtedly SE. The expression of these RA-associated variants of HLA DR B1 alleles is due to the presentation of low-affinity antigens, activation of autoreactive T lymphocytes and ACCP production. The other significant weak link is a poorly controlled and therefore beyond reasonable sufficiency pro-inflammatory response due to the bulk of SNPs accumulated in NFkappaB- and Jak/STAT, cytokine signaling pathways together with insufficient inhibitory control. It should be noted that RA is a phenotypically heterogenic pathology due to the sets of SNPs that differ from case to case. Therefore, hypothetically, there may be an RA subtype with congenital susceptibility to infections due to the SNPs of the genes involved in anti-infective protection and another RA subtype in persons with an imbalance in sex and corticosteroid hormones or an insufficient ecotoxicant detoxication system with matching sets of SNPs. Therefore, it can be assumed that one or another of the discussed effects may come to the fore in certain RA subtypes. Given the growing interest in the preclinical stages of RA, which are known to develop on the mucous membranes, perhaps the most promising line of research is to study the interplay of barrier tissues, the local immune system and the microbiome in persons at risk of RA.
It should be noted that environmental and individual factors affecting the loci minoris resistentia of a body at risk of RA form tangles of interdependencies, thus increasing their impact on the development of the disease (Figure 5).
Many non-genetic factors form the network of interdependent relationships; thus, several interdependent external factors can hit one weak body locus at once, either provoking or reinforcing each other (Figure 6).
Given the fact that the ratio of genetic and non-genetic RA risks is considered to be fifty/fifty, the algorithm for disease risk predicting should include both genetic and non-genetic factors, as well as any laboratory parameters indicating the negative impact of these factors. If we want to track down and, if possible, to prevent negative development of events at the earliest RA stages, the diagnosis of preclinical stages based on the presence of autoantibody/inflammatory markers is somewhat belated, not to mention that articular symptoms may be non-specific. This is not an easy task, keeping in mind the non-specific character of the parameters modified by external factors, the same as in the general population. It may be necessary to scale both the intensity of the external influence and the severity of response to it. The task is further complicated by the fact that the sets of gene SNPs that can lead to the development of RA can vary greatly from person to person. Indeed, RA is characterized by a wide variety of clinical manifestations (phenotypic heterogeneity).
That’s why a number of the more radical thinkers even believe that, in fact, the set of clinical signs that have been known to us since 1782 [177], and which we call "rheumatoid arthritis", is the outcome of many different pathogenic pathways—or, in other words, a syndrome resulting from a number of different diseases [52].

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23158140/s1. References [6,22,52,109,144,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243] are cited in the Supplementary Materials.

Author Contributions

Conceptualization, M.A., Y.R. and W.B.; methodology, M.A., Y.R. and W.B.; writing—original draft preparation, E.T., S.P., G.K., A.V., E.S., M.M. and O.K.; writing—review and editing, M.A., Y.R. and W.B.; visualization, R.L., M.K. and A.N.; supervision, M.A.; project administration, M.A.; funding acquisition, M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Russian Science Foundation (No. 17-15-01099).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data discussed in the review were obtained in publications posted in PubMed.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kobayashi, S.; Momohara, S.; Kamatani, N.; Okamoto, H. Molecular aspects of rheumatoid arthritis: Role of environmental factors. FEBS J. 2008, 275, 4456–4462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Prokunina, L.; Padyukov, L.; Bennet, A.; de Faire, U.; Wiman, B.; Prince, J.; Alfredsson, L.; Klareskog, L.; Alarcón-Riquelme, M. Association of the PD-1.3A allele of the PDCD1 gene in patients with rheumatoid arthritis negative for rheumatoid factor and the shared epitope. Arthritis Rheum. 2004, 50, 1770–1773. [Google Scholar] [CrossRef]
  3. Rosloniec, E.F.; Whittington, K.B.; Zaller, D.M.; Kang, A.H. HLA-DR1 (DRB1*0101) and DR4 (DRB1*0401) use the same anchor residues for binding an immunodominant peptide derived from human type II collagen. J. Immunol. 2002, 168, 253–259. [Google Scholar] [CrossRef] [PubMed]
  4. Hill, J.A.; Wang, D.; Jevnikar, A.M.; Cairns, E.; Bell, D.A. The relationship between predicted peptide-MHC class II affinity and T-cell activation in a HLA-DRbeta1*0401 transgenic mouse model. Arthritis Res. Ther. 2002, 5, R40–R48. [Google Scholar] [CrossRef]
  5. Auger, I.; Sebbag, M.; Vincent, C.; Balandraud, N.; Guis, S.; Nogueira, L.; Svensson, B.; Cantagrel, A.; Serre, G.; Roudier, J. Influence of HLA-DR genes on the production of rheumatoid arthritis-specific autoantibodies to citrullinated fibrinogen. Arthritis Rheum. 2005, 52, 3424–3432. [Google Scholar] [CrossRef] [PubMed]
  6. Gourraud, P.A.; Dieudé, P.; Boyer, J.F.; Nogueira, L.; Cambon-Thomsen, A.; Mazières, B.; Cornélis, F.; Serre, G.; Cantagrel, A.; Constantin, A. A new classification of HLA-DRB1 alleles differentiates predisposing and protective alleles for autoantibody production in rheumatoid arthritis. Arthritis Res. Ther. 2007, 9, R27. [Google Scholar] [CrossRef] [Green Version]
  7. Gourraud, P.A.; Boyer, J.F.; Barnetche, T.; Abbal, M.; Cambon-Thomsen, A.; Cantagrel, A.; Constantin, A. A new classification of HLA-DRB1 alleles differentiates predisposing and protective alleles for rheumatoid arthritis structural severity. Arthritis Rheum. 2006, 54, 593–599. [Google Scholar] [CrossRef]
  8. Ohnishi, Y.; Tsutsumi, A.; Matsumoto, I.; Goto, D.; Ito, S.; Kuwana, M.; Uemura, Y.; Nishimura, Y.; Sumida, T. Altered peptide ligands control type II collagen-reactive T cells from rheumatoid arthritis patients. Mod. Rheumatol. 2006, 16, 226–228. [Google Scholar] [CrossRef]
  9. Sakkas, L.I.; Daoussis, D.; Liossis, S.N.; Bogdanos, D.P. The Infectious Basis of ACPA-Positive Rheumatoid Arthritis. Front. Microbiol. 2017, 8, 1853. [Google Scholar] [CrossRef]
  10. Lundberg, K.; Bengtsson, C.; Kharlamova, N.; Reed, E.; Jiang, X.; Kallberg, H.; Pollak-Dorocic, I.; Israelsson, L.; Kessel, C.; Padyukov, L.; et al. Genetic and environmental determinants for disease risk in subsets of rheumatoid arthritis defined by the anticitrullinated protein/peptide antibody fine specificity profile. Ann. Rheum. Dis. 2012, 72, 652–658. [Google Scholar] [CrossRef] [Green Version]
  11. Fu, J.; Nogueira, S.V.; Drongelen, V.V.; Coit, P.; Ling, S.; Rosloniec, E.F.; Sawalha, A.H.; Holoshitz, J. Shared epitope-aryl hydrocarbon receptor crosstalk underlies the mechanism of gene-environment interaction in autoimmune arthritis. Proc. Natl. Acad. Sci. USA 2018, 115, 4755–4760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chaudhry, W.N.; Concepción-Acevedo, J.; Park, T.; Andleeb, S.; Bull, J.J.; Levin, B.R. Synergy and Order Effects of Antibiotics and Phages in Killing Pseudomonas aeruginosa Biofilms. PLoS ONE 2017, 12, e0168615. [Google Scholar] [CrossRef] [PubMed]
  13. Diogo, D.; Okada, Y.; Plenge, R.M. Genome-wide association studies to advance our understanding of critical cell types and pathways in rheumatoid arthritis: Recent findings and challenges. Curr. Opin. Rheumatol. 2014, 26, 85–92. [Google Scholar] [CrossRef] [PubMed]
  14. Messemaker, T.C.; Huizinga, T.W.; Kurreeman, F. Immunogenetics of rheumatoid arthritis: Understanding functional implications. J. Autoimmun. 2015, 64, 74–81. [Google Scholar] [CrossRef]
  15. Okada, Y.; Wu, D.; Trynka, G.; Raj, T.; Terao, C.; Ikari, K.; Kochi, Y.; Ohmura, K.; Suzuki, A.; Yoshida, S. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 2014, 506, 376–381. [Google Scholar] [CrossRef]
  16. Ribet, D.; Cossart, P. Pathogen-Mediated Posttranslational Modifications: A Re-emerging Field. Cell 2010, 143, 694–702. [Google Scholar] [CrossRef] [Green Version]
  17. Rahman, M.; McFadden, G. Modulation of NF-κBsignalling by microbial pathogens. Nat. Rev. Microbiol. 2011, 9, 291–306. [Google Scholar] [CrossRef]
  18. Zhao, J.; He, S.; Minassian, A.; Li, J.; Feng, P. Recent advances on viral manipulation of NF-κB signaling pathway. Curr. Opin. Virol. 2015, 15, 103–111. [Google Scholar] [CrossRef] [Green Version]
  19. Jangra, S.; Yuen, K.-S.; Botelho, M.G.; Jin, D.-Y. Epstein-Barr Virus and Innate Immunity: Friends or Foes? Microorganisms 2019, 7, 183. [Google Scholar] [CrossRef] [Green Version]
  20. Sonenshine, D.E.; Macaluso, K.R. Microbial Invasion vs. Tick Immune Regulation. Front. Cell Infect. Microbiol. 2017, 7, 390. [Google Scholar] [CrossRef]
  21. Arch, R.H.; Thompson, C.B. Lymphocyte survival--the struggle against death. Annu. Rev. Cell Dev. Biol. 1999, 15, 113–140. [Google Scholar] [CrossRef] [PubMed]
  22. Barr, T.A.; Brown, S.; Ryan, G.; Zhao, J.; Gray, D. TLR-mediated stimulation of APC: Distinct cytokine responses of B cells and dendritic cells. Eur. J. Immunol. 2007, 37, 3040–3053. [Google Scholar] [CrossRef] [PubMed]
  23. Lindblad, S.S.; Mydel, P.; Jonsson, I.M.; Senior, R.M.; Tarkowski, A.; Bokarewa, M. Smoking and nicotine exposure delay development of collagen-induced arthritis in mice. Arthritis Res. Ther. 2009, 11, R88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Munro, S.K.; Balakrishnan, B.; Lissaman, A.C.; Gujral, P.; Ponnampalam , A.P. Cytokines and pregnancy: Potential regulation by histone deacetylases. Mol. Reprod. Dev. 2021, 88, 321–337. [Google Scholar] [CrossRef] [PubMed]
  25. Collins, K.H.; Herzog, W.; MacDonald, G.Z.; Reimer, R.A.; Rios, J.L.; Smith, I.C.; Zernicke, R.F.; Hart, D.A. Obesity, Metabolic Syndrome, and Musculoskeletal Disease: Common Inflammatory Pathways Suggest a Central Role for Loss of Muscle Integrity. Front. Physiol. 2018, 9, 112. [Google Scholar] [CrossRef]
  26. Carroll, J.E.; Carrillo, C.; Olmstead, R.; Witarama, T.; Breen, E.C.; Yokomizo, M.; Seeman, T.; Irwin, M.R. Sleep deprivation and divergent toll-like receptor-4 activation of cellular inflammation in aging. Sleep 2015, 38, 205–211. [Google Scholar] [CrossRef]
  27. Richardson, M.R.; Churilla, J.R. Sleep Duration and C-Reactive Protein in US Adults. South Med. J. 2017, 110, 314–317. [Google Scholar] [CrossRef]
  28. Dolsen, M.R.; Crosswell, A.D.; Prather, A.A. Links between Stress, Sleep, and Inflammation: Are there Sex Differences? Curr. Psychiatry Rep. 2019, 21, 8. [Google Scholar] [CrossRef]
  29. Frisell, T.; Holmqvist, M.; Källberg, H.; Klareskog, L.; Alfredsson, L.; Askling, J. Familial risks and heritability of rheumatoid arthritis: Role of rheumatoid factor/anti-citrullinated protein antibody status, number and type of affected relatives, sex, and age. Arthritis Rheum. 2013, 65, 2773–2782. [Google Scholar] [CrossRef]
  30. Jiang, X.; Frisell, T.; Askling, J.; Karlson, E.W.; Klareskog, L.; Alfredsson, L.; Källberg, H. To what extent is the familial risk of rheumatoid arthritis explained by established rheumatoid arthritis risk factors? Arthritis Rheumatol. 2015, 67, 352–362. [Google Scholar] [CrossRef] [Green Version]
  31. Abdel-Nasser, A.M.; Rasker, J.J.; Valkenburg, H.A. Epidemiological and clinical aspects relating to the variability of rheumatoid arthritis. Semin. Arthritis Rheum. 1997, 27, 123–140. [Google Scholar] [CrossRef]
  32. Alamanos, Y.; Drosos, A.A. Epidemiology of adult rheumatoid arthritis. Autoimmun. Rev. 2005, 4, 130–136. [Google Scholar] [CrossRef] [PubMed]
  33. Ferucci, E.D.; Templin, D.W.; Lanier, A.P. Rheumatoid arthritis in American Indians and Alaska Natives: A review of the literature. Semin. Arthritis Rheum. 2005, 34, 662–667. [Google Scholar] [CrossRef] [PubMed]
  34. Kochi, Y.; Suzuki, A.; Yamada, R.; Yamamoto, K. Ethnogenetic heterogeneity of rheumatoid arthritis-implications for pathogenesis. Nat. Rev. Rheumatol. 2010, 6, 290–295. [Google Scholar] [CrossRef]
  35. Kim, K.; Bang, S.Y.; Lee, H.S.; Cho, S.K.; Choi, C.B.; Sung, Y.K.; Kim, T.H.; Jun, J.; Yoo, D.H.; Kang, Y.M. High-density genotyping of immune loci in Koreans and Europeans identifies eight new rheumatoid arthritis risk loci. Ann. Rheum. Dis. 2014, 74, e13. [Google Scholar] [CrossRef]
  36. Okada, Y.; Terao, C.; Ikari, K.; Kochi, Y.; Ohmura, K.; Suzuki, A.; Kawaguchi, T.; Stahl, E.A.; Kurreeman, F.A.; Nishida, N. Meta-analysis identifies nine new loci associated with rheumatoid arthritis in the Japanese population. Nat. Genet. 2012, 44, 511–516. [Google Scholar] [CrossRef]
  37. Bergström, U.; Jacobsson, L.T.H.; Nilsson, J.A.; Wirfält, E.; Turesson, C. Smoking, low formal level of education, alcohol consumption, and the risk of rheumatoid arthritis. Scand. J. Rheumatol. 2013, 42, 123–130. [Google Scholar] [CrossRef]
  38. Xu, B.; Lin, J. Characteristics and risk factors of rheumatoid arthritis in the United States: An NHANES analysis. PeerJ 2017, 5, e4035. [Google Scholar] [CrossRef]
  39. Trejo, V.; Derom, C.; Vlietinck, R.; Ollier, W.; Silman, A.; Ebers, G.; Derom, R.; Gregersen, P.K. X chromosome inactivation patterns correlate with fetal-placental anatomy in monozygotic twin pairs: Implications for immune relatedness and concordance for autoimmunity. Mol. Med. 1994, 1, 62–70. [Google Scholar] [CrossRef] [Green Version]
  40. Eriksson, J.G. Early growth and adult health outcomes: Lessons learned from the Helsinki Birth Cohort Study. Matern. Child Nutr. 2005, 1, 149–154. [Google Scholar] [CrossRef]
  41. Eriksson, J.G. Epidemiology, genes and the environment: Lessons learned from the Helsinki Birth Cohort Study. J. Intern. Med. 2007, 261, 418–425. [Google Scholar] [CrossRef] [PubMed]
  42. Stünkel, W.; Pan, H.; Chew, S.B.; Tng, E.; Tan, J.H.; Chen, L.; Joseph, R.; Cheong, C.Y.; Ong, M.L.; Lee, Y.S. Transcriptome changes affecting Hedgehog and cytokine signalling in the umbilical cord: Implications for disease risk. PLoS ONE 2012, 7, e39744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Herberth, G.; Hinz, D.; Röder, S.; Schlink, U.; Sack, U.; Diez, U.; Borte, M.; Lehmann, I. Maternal immune status in pregnancy is related to offspring’s immune responses and atopy risk. Allergy 2011, 66, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  44. Djuardi, Y.; Wibowo, H.; Supali, T.; Ariawan, I.; Bredius, R.G.; Yazdanbakhsh, M.; Rodrigues, L.C.; Sartono, E. Determinants of the relationship between cytokine production in pregnant women and their infants. PLoS ONE 2009, 4, e7711. [Google Scholar] [CrossRef] [Green Version]
  45. Jenmalm, M.C. Childhood immune maturation and allergy development: Regulation by maternal immunity and microbial exposure. Am. J. Reprod. Immunol. 2011, 66, 75–80. [Google Scholar] [CrossRef] [Green Version]
  46. Jaakkola, J.J.; Gissler, M. Maternal smoking in pregnancy as a determinant of rheumatoid arthritis and other inflammatory polyarthropathies during the first 7 years of life. Int. J. Epidemiol. 2005, 34, 664–671. [Google Scholar] [CrossRef]
  47. Rotroff, D.M.; Joubert, B.R.; Marvel, S.W.; Håberg, S.E.; Wu, M.C.; Nilsen, R.M.; Ueland, P.M.; Nystad, W.; London, S.J.; Motsinger-Reif, A. Maternal smoking impacts key biological pathways in newborns through epigenetic modification in Utero. BMC Genom. 2016, 17, 976. [Google Scholar] [CrossRef] [Green Version]
  48. Carlens, C.; Jacobsson, L.; Brandt, L.; Cnattingius, S.; Stephansson, O.; Askling, J. Perinatal characteristics, early life infections and later risk of rheumatoid arthritis and juvenile idiopathic arthritis. Ann. Rheum. Dis. 2009, 68, 1159–1164. [Google Scholar] [CrossRef]
  49. Hughes, G.C.; Choubey, D. Modulation of autoimmune rheumatic diseases by oestrogen and progesterone. Nat. Rev. Rheumatol. 2014, 10, 740–751. [Google Scholar] [CrossRef]
  50. Pierdominici, M.; Maselli, A.; Colasanti, T.; Giammarioli, A.M.; Delunardo, F.; Vacirca, D.; Sanchez, M.; Giovannetti, A.; Malorni, W.; Ortona, E. Estrogen receptor profiles in human peripheral blood lymphocytes. Immunol. Lett. 2010, 132, 79–85. [Google Scholar] [CrossRef]
  51. Ortona, E.; Pierdominici, M.; Maselli, A.; Veroni, C.; Aloisi, F.; Shoenfeld, Y. Sex-based differences in autoimmune diseases. Ann. Ist Super Sanita. 2016, 52, 205–212. [Google Scholar] [CrossRef] [PubMed]
  52. Østensen, M.; Förger, F.; Nelson, J.L.; Schuhmacher, A.; Hebisch, G.; Villiger, P.M. Pregnancy in patients with rheumatic disease: Anti-inflammatory cytokines increase in pregnancy and decrease postpartum. Ann. Rheum. Dis. 2005, 64, 839–844. [Google Scholar] [CrossRef] [PubMed]
  53. Wan, J.; Hu, Z.; Zeng, K.; Yin, Y.; Zhao, M.; Chen, M.; Chen, Q. The reduction in circulating levels of estrogen and progesterone in women with preeclampsia. Pregnancy Hypertens. 2018, 11, 18–25. [Google Scholar] [CrossRef]
  54. Kuciene, R.; Dulskiene, V. Associations of maternal gestational hypertension with high blood pressure and overweight/obesity in their adolescent offspring: A retrospective cohort study. Sci Rep. 2022, 12, 3800. [Google Scholar] [CrossRef]
  55. Ho, J.T.; Lewis, J.G.; O’Loughlin, P.; Bagley, C.J.; Romero, R.; Dekker, G.A.; Torpy, D.J. Reduced maternal corticosteroid-binding globulin and cortisol levels in pre-eclampsia and gamete recipient pregnancies. Clin. Endocrinol. 2007, 66, 869–877. [Google Scholar] [CrossRef]
  56. Cherak, S.J.; Giesbrecht, G.F.; Metcalfe, A.; Ronksley, P.E.; Malebranche, M.E. The effect of gestational period on the association between maternal prenatal salivary cortisol and birth weight: A systematic review and meta-analysis. Psychoneuroendocrinology 2018, 94, 49–62. [Google Scholar] [CrossRef]
  57. Kellgren, J.H.; Lawrence, J.S.; Aitken-Swan, J. Rheumatic complaints in an urban population. Ann. Rheum. Dis. 1953, 12, 5–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Gabriel, S.E.; Crowson, C.S.; O’Fallon, W.M. The epidemiology of rheumatoid arthritis in Rochester, Minnesota, 1955-1985. Arthritis. Rheum. 1999, 42, 415–420. [Google Scholar] [CrossRef]
  59. Lindstrom, T.M.; Robinson, W.H. Rheumatoid arthritis: A role for immunosenescence? J. Am. Geriatr. Soc. 2010, 58, 1565–1575. [Google Scholar] [CrossRef] [Green Version]
  60. Weyand, C.M.; Goronzy, J.J. Aging of the Immune System. Mechanisms and Therapeutic Targets. Ann. Am. Thorac. Soc. 2016, 13, S422–S428. [Google Scholar] [CrossRef] [Green Version]
  61. Andersen, C.J.; Murphy, K.E.; Fernandez, M.L. Impact of Obesity and Metabolic Syndrome on Immunity. Adv. Nutr. 2016, 7, 66–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Lemerle, J.; Arleevskaya, M.I.; Brooks, W.H.; Renaudineau, Y. Effects of environmental factors and omega-3 fatty acids on rheumatoid arthritis. Ann. Joint 1. 2016, 1. [Google Scholar] [CrossRef]
  63. Sköldstam, L.; Hagfors, L.; Johansson, G. An experimental study of a Mediterranean diet intervention for patients with rheumatoid arthritis. Ann. Rheum. Dis. 2003, 62, 208–214. [Google Scholar] [CrossRef]
  64. Casas, R.; Sacanella, E.; Estruch, R. The immune protective effect of the Mediterranean diet against chronic low-grade inflammatory diseases. Endocr. Metab. Immune Disord. Drug Targets 2014, 14, 245–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Rosell, M.; Wesley, A.M.; Rydin, K.; Klareskog, L.; Alfredsson, L.; EIRA study group. Dietary fish and fish oil and the risk of rheumatoid arthritis. Epidemiology 2009, 20, 896–901. [Google Scholar] [CrossRef]
  66. Thorburn, A.N.; Macia, L.; Mackay, C.R. Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity 2014, 40, 833–842. [Google Scholar] [CrossRef] [Green Version]
  67. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef]
  68. Vinolo, M.A.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef] [Green Version]
  69. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; de Roos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  70. Sparks, J.A.; Barbhaiya, M.; Tedeschi, S.K.; Leatherwood, C.L.; Tabung, F.K.; Speyer, C.B.; Malspeis, S.; Costenbader, K.H.; Karlson, E.W.; Lu, B. Inflammatory dietary pattern and risk of developing rheumatoid arthritis in women. Clin. Rheumatol. 2019, 38, 243–250. [Google Scholar] [CrossRef]
  71. Müller, S.; Quast, T.; Schröder, A.; Hucke, S.; Klotz, L.; Jantsch, J.; Gerzer, R.; Hemmersbach, R.; Kolanus, W. Salt-Dependent Chemotaxis of Macrophages. PLoS ONE 2013, 8, e73439. [Google Scholar] [CrossRef] [Green Version]
  72. Zhang, W.C.; Du, L.J.; Zheng, X.J.; Chen, X.Q.; Shi, C.; Chen, B.Y.; Sun, X.N.; Li, C.; Zhang, Y.Y.; Liu, Y.; et al. Elevated sodium chloride drives type I interferon signaling in macrophages and increases antiviral resistance. J. Biol. Chem. 2018, 293, 030–1039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Binger, K.J.; Gebhardt, M.; Heinig, M.; Rintisch, C.; Schroeder, A.; Neuhofer, W.; Hilgers, K.; Manzel, A.; Schwartz, K.; Kleinewietfeld, M.; et al. High salt reduces the activation of IL-4– and IL-13–stimulated macrophages. J. Clin. Investig. 2015, 125, 4223–4238. [Google Scholar] [CrossRef] [PubMed]
  74. Barbaro, N.R.; Jason, D.; Foss, J.D.; Kryshta, D.O.; Tsyba, N.; Kumaresan, S.; Xiao, L.; Mernaugh, R.L.; Itani, H.A.; Loperena, R.; et al. Dendritic Cell Amiloride-Sensitive Channels Mediate Sodium Induced Inflammation and Hypertension. Cell Rep. 2017, 21, 1009–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Wu, C.; Yosef, N.; Thalhamer, T.; Zhu, C.; Xiao, S.; Kishi, Y.; Regev, A.; Kuchroo, V. Induction of pathogenic Th17 cells by inducible salt sensing kinase SGK1. Nature 2013, 496, 513–517. [Google Scholar] [CrossRef] [Green Version]
  76. Hernandez, A.L.; Kitz, A.; Wu, C.; Lowther, D.E.; Rodriguez, D.M.; Vudattu, N.; Deng, S.; Herold, K.C.; Kuchroo, V.K.; Kleinewietfeld, M.; et al. Sodium chloride inhibits the suppressive function of FOXP3+ regulatory T cells. J. Clin. Investig. 2015, 125, 4212–4222. [Google Scholar] [CrossRef] [Green Version]
  77. Jung, S.M.; Kim, Y.; Kim, J.; Jung, H.; Yi, H.; Rim, Y.A.; Park, N.; Kwok, S.K.; Park, S.H.; Ju, J.H. Sodium Chloride Aggravates Arthritis via Th17 Polarization. Yonsei Med. J. 2019, 60, 88–97. [Google Scholar] [CrossRef]
  78. Sehnert, B.; Pohle, S.; Heuberger, C.; Rzepka, R.; Seidl, M.; Nimmerjahn, F.; Chevalier, N.; Titze, J.; Voll, R.E. Low-Salt Diet Attenuates B-Cell- and Myeloid-Cell-Driven Experimental Arthritides by Affecting Innate as Well as Adaptive Immune Mechanisms. Front. Immunol. 2021, 12, 765741. [Google Scholar] [CrossRef]
  79. Yi, B.; Titze, J.; Rykova, M.; Feuerecker, M.; Vassilieva, G.; Nichiporuk, I.; Schelling, G.; Morukov, B.; Choukèr, A. Effects of dietary salt levels on monocytic cells and immune responses in healthy human subjects: A longitudinal study. Transl. Res. 2015, 166, 103–110. [Google Scholar] [CrossRef] [Green Version]
  80. Vitales-Noyola, M.; Layseca-Espinosa, E.; Baranda, L.; Abud-Mendoza, C.; Niño-Moreno, P.; Monsiváis-Urenda, A.; Rosenstein, Y.; González-Amaro, R. Analysis of Sodium Chloride Intake and Treg/Th17 Lymphocytes in Healthy Individuals and Patients with Rheumatoid Arthritis or Systemic Lupus Erythematosus. J. Immunol. Res. 2018, 2018, 9627806. [Google Scholar] [CrossRef]
  81. Minamino, H.; Katsushima, M.; Hashimoto, M.; Fujita, Y.; Yoshida, T.; Ikeda, K.; Isomura, N.; Oguri, Y.; Yamamoto, W.; Watanabe, R.; et al. Urinary sodium-to-potassium ratio associates with hypertension and current disease activity in patients with rheumatoid arthritis: A cross-sectional study. Arthritis Res. Ther. 2021, 23, 96. [Google Scholar] [CrossRef] [PubMed]
  82. Salgado, E.; Bes-Rastrollo, M.; de Irala, J.; Carmona, L.; Gómez-Reino, J.J. High Sodium Intake Is Associated with Self-Reported Rheumatoid Arthritis: A Cross Sectional and Case Control Analysis Within the SUN Cohort. Medicine 2015, 94, e0924. [Google Scholar] [CrossRef] [PubMed]
  83. Sundström, B.; Johansson, I.; Rantapää-Dahlqvist, S. Interaction between dietary sodium and smoking increases the risk for rheumatoid arthritis: Results from a nested case-control study. Rheumatology 2015, 54, 487–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Jiang, X.; Sundström, B.; Alfredsson, L.; Klareskog, L.; Rantapää-Dahlqvist, S.; Bengtsson, C. High sodium chloride consumption enhances the effects of smoking but does not interact with SGK1 polymorphisms in the development of ACPA-positive status in patients with RA. Ann. Rheum. Dis. 2016, 75, 943–946. [Google Scholar] [CrossRef]
  85. Scrivo, R.; Massaro, L.; Barbati, C.; Vomero, M.; Ceccarelli, F.; Spinelli, F.R.; Riccieri, V.; Spagnoli, A.; Alessandri, C.; Desideri, G.; et al. The role of dietary sodium intake on the modulation of T helper 17 cells and regulatory T cells in patients with rheumatoid arthritis and systemic lupus erythematosus. PLoS ONE 2017, 12, e0184449. [Google Scholar] [CrossRef]
  86. Pedersen, M.; Jacobsen, S.; Klarlund, M.; Pedersen, B.V.; Wiik, A.; Wohlfahrt, J.; Frisch, M. Environmental risk factors differ between rheumatoid arthritis with and without auto-antibodies against cyclic citrullinated peptides. Arthritis Res. Ther. 2006, 8, R133. [Google Scholar] [CrossRef] [Green Version]
  87. Heliövaara, M.; Aho, K.; Knekt, P.; Impivaara, O.; Reunanen, A.; Aromaa, A. Coffee consumption, rheumatoid factor, and the risk of rheumatoid arthritis. Ann. Rheum. Dis. 2000, 59, 631–635. [Google Scholar] [CrossRef] [Green Version]
  88. Mikuls, T.R.; Cerhan, J.R.; Criswell, L.A.; Merlino, L.; Mudano, A.S.; Burma, M.; Folsom, A.R.; Saag, K.G. Coffee, tea, and caffeine consumption and risk of rheumatoid arthritis: Results from the Iowa Women’s Health Study. Arthritis Rheum. 2002, 46, 83–91. [Google Scholar] [CrossRef]
  89. Pedersen, M.; Stripp, C.; Klarlund, M.; Olsen, S.F.; Tjønneland, A.M.; Frisch, M. Diet and risk of rheumatoid arthritis in a prospective cohort. J. Rheumatol. 2005, 32, 1249–1252. [Google Scholar]
  90. Rambod, M.; Nazarinia, M.; Raieskarimian, F. The impact of dietary habits on the pathogenesis of rheumatoid arthritis: A case-control study. Clin. Rheumatol. 2018, 37, 2643–2648. [Google Scholar] [CrossRef]
  91. Sharif, K.; Watad, A.; Bragazzi, N.L.; Adawi, M.; Amital, H.; Shoenfeld, Y. Coffee and autoimmunity: More than a mere hot beverage! Autoimmun. Rev. 2017, 16, 712–721. [Google Scholar] [CrossRef] [PubMed]
  92. Di Dalmazi, G.; Hirshberg, J.; Lyle, D.; Freij, J.B.; Caturegli, P. Reactive oxygen species in organ-specific autoimmunity. Auto Immun. Highlights 2016, 7, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Priftis, A.; Stagos, D.; Konstantinopoulos, K.; Tsitsimpikou, C.; Spandidos, D.A.; Tsatsakis, A.M.; Tzatzarakis, M.N.; Kouretas, D. Comparison of antioxidant activity between green and roasted coffee beans using molecular methods. Mol. Med. Rep. 2015, 12, 7293–7302. [Google Scholar] [CrossRef] [Green Version]
  94. Di Giuseppe, D.; Alfredsson, L.; Bottai, M.; Askling, J.; Wolk, A. Long term alcohol intake and risk of rheumatoid arthritis in women: A population based cohort study. BMJ 2012, 345, e4230. [Google Scholar] [CrossRef] [Green Version]
  95. Larsson, I.; Andersson, M.; BARFOT study group. Reasons to stop drinking alcohol among patients with rheumatoid arthritis in Sweden: A mixed-methods study. BMJ Open 2018, 8, e024367. [Google Scholar] [CrossRef] [Green Version]
  96. Seitz, S.; Schneider, C.K.; Malotka, J.; Nong, X.; Engel, A.G.; Wekerle, H.; Hohlfeld, R.; Dornmair, K. Reconstitution of paired T cell receptor alpha- and beta-chains from microdissected single cells of human inflammatory tissues. Proc. Natl. Acad. Sci. USA 2006, 103, 12057–12062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Jonsson, I.M.; Verdrengh, M.; Brisslert, M.; Lindblad, S.; Bokarewa, M.; Islander, U.; Carlsten, H.; Ohlsson, C.; Nandakumar, K.S.; Holmdahl, R.; et al. Ethanol prevents development of destructive arthritis. Proc. Natl. Acad. Sci. USA 2007, 104, 258–263. [Google Scholar] [CrossRef] [Green Version]
  98. Waldschmidt, T.J.; Cook, R.T.; Kovacs, E.J. Alcohol and inflammation and immune responses: Summary of the 2006 Alcohol and Immunology Research Interest Group (AIRIG) meeting. Alcohol 2008, 42, 137–142. [Google Scholar] [CrossRef] [Green Version]
  99. Carlens, C.; Hergens, M.P.; Grunewald, J.; Ekbom, A.; Eklund, A.; Höglund, C.O.; Askling, J. Smoking, use of moist snuff, and risk of chronic inflammatory diseases. Am. J. Respir. Crit. Care Med. 2010, 181, 1217–1222. [Google Scholar] [CrossRef]
  100. Chiang, Y.C.; Yen, Y.H.; Chang, W.C.; Cheng, K.J.; Chang, W.P.; Chen, H.Y. The association between urbanization and rheumatoid arthritis in Taiwan. Int. J. Clin. Pharmacol. Ther. 2016, 54, 1–10. [Google Scholar] [CrossRef]
  101. Ziarko, M.; Mojs, E.; Kaczmarek, Ł.D.; Warchol-Biedermann, K.; Malak, R.; Lisinski, P.; Samborski, W. Do urban and rural residents living in Poland differ in their ways of coping with chronic diseases? Eur. Rev. Med. Pharmacol. Sci. 2015, 9, 4227–4234. [Google Scholar]
  102. Neovius, M.; Simard, J.F.; Askling, J.; ARTIS study group. Nationwide prevalence of rheumatoid arthritis and penetration of disease-modifying drugs in Sweden. Ann. Rheum. Dis. 2011, 70, 624–629. [Google Scholar] [CrossRef] [PubMed]
  103. Szabo, S.; Yoshida, M.; Filakovszky, J.; Juhasz, G. “Stress” is 80 Years Old: From Hans Selye Original Paper in 1936 to Recent Advances in GI Ulceration. Curr. Pharm. Des. 2017, 23, 4029–4041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Khanfer, R.; Carroll, D.; Lord, J.M.; Phillips, A.C. Reduced neutrophil superoxide production among healthy older adults in response to acute psychological stress. Int. J. Psychophysiol. 2012, 86, 238–244. [Google Scholar] [CrossRef]
  105. Maxwell, L.; Barrett, B.; Chase, J.; Brown, R.; Ewers, T. Self-Reported Mental Health Predicts Acute Respiratory Infection. WMJ 2015, 114, 100–104. [Google Scholar]
  106. Cohen, S.; Tyrrell, D.A.; Smith, A.P. Psychological stress and susceptibility to the common cold. N. Engl. J. Med. 1991, 325, 606–612. [Google Scholar] [CrossRef]
  107. Grinde, B. Herpesviruses: Latency and reactivation-viral strategies and host response. J. Oral. Microbiol. 2013, 5, 1–9. [Google Scholar] [CrossRef] [Green Version]
  108. Arleevskaya, M.I.; Kravtsova, O.A.; Lemerle, J.; Renaudineau, Y.; Tsibulkin, A.P. How Rheumatoid Arthritis Can Result from Provocation of the Immune System by Microorganisms and Viruses. Front. Microbiol. 2016, 7, 1296. [Google Scholar] [CrossRef] [Green Version]
  109. Arleevskaya, M.I.; Gabdoulkhakova, A.G.; Filina, Y.V.; Miftakhova, R.R.; Bredberg, A.; Tsybulkin, A.P. A transient peak of infections during onset of rheumatoid arthritis: A 10-year prospective cohort study. BMJ Open. 2014, 4, e005254. [Google Scholar] [CrossRef] [Green Version]
  110. Arleevskaya, M.I.; Shafigullina, A.Z.; Filina, Y.V.; Lemerle, J.; Renaudineau, Y. Associations between Viral Infection History Symptoms, Granulocyte Reactive Oxygen Species Activity, and Active Rheumatoid Arthritis Disease in Untreated Women at Onset: Results from a Longitudinal Cohort Study of Tatarstan Women. Front. Immunol. 2017, 8, 1725. [Google Scholar] [CrossRef]
  111. Uffelmann, E.; Huang, Q.Q.; Munung, N.S.; de Vries, J.; Okada, Y.; Martin, A.R.; Martin, H.C.; Lappalainen, T.; Posthuma, D. Genome-wide association studies. Nat. Rev. Methods Primers 2021, 1, 59. [Google Scholar] [CrossRef]
  112. Tuaeva, N.O.; Nosyrev, A.E.; Renaudineau, Y.; Arleevskaya, M.I. Psychological stress and rheumatoid arthritis-interference of pathogenetic mechanisms. Rheumatol. Sci. Pract. 2019, 57, 83–90. [Google Scholar] [CrossRef]
  113. Bryant, P.A.; Trinder, J.; Curtis, N. Sick and tired: Does sleep have a vital role in the immune system? Nat. Rev. Immunol. 2004, 4, 457–467. [Google Scholar] [CrossRef] [PubMed]
  114. Cutolo, M.; Sulli, A.; Pizzorni, C.; Secchi, M.E.; Soldano, S.; Seriolo, B.; Straub, R.H.; Otsa, K.; Maestroni, G.J. Circadian rhythms: Glucocorticoids and arthritis. Ann. N. Y. Acad. Sci. 2006, 1069, 289–299. [Google Scholar] [CrossRef]
  115. Morgan, E.; Schumm, L.P.; McClintock, M.; Waite, L.; Lauderdale, D.S. Sleep Characteristics and Daytime Cortisol Levels in Older Adults. Sleep 2007, 40, zsx043. [Google Scholar] [CrossRef] [Green Version]
  116. Dimitrov, S.; Lange, T.; Fehm, H.L.; Born, J. A regulatory role of prolactin, growth hormone, and corticosteroids for human T-cell production of cytokines. Brain Behav. Immun. 2004, 18, 368–374. [Google Scholar] [CrossRef]
  117. Marshall, L.; Born, J. Brain-immune interactions in sleep. Int. Rev. Neurobiol. 2002, 52, 93–131. [Google Scholar] [CrossRef]
  118. Spiegel, K.; Sheridan, J.F.; Van Cauter, E. Effect of sleep deprivation on response to immunization. JAMA 2002, 288, 1471–1472. [Google Scholar] [CrossRef]
  119. Lange, T.; Perras, B.; Fehm, H.L.; Born, J. Sleep enhances the human antibody response to hepatitis A vaccination. Psychosom. Med. 2003, 65, 831–835. [Google Scholar] [CrossRef]
  120. Walsh, C.P.; Lim, A.; Marsland, A.L.; Ferrell, R.E.; Manuck, S.B. Circulating Interleukin-6 concentration covaries inversely with self-reported sleep duration as a function of polymorphic variation in the glucocorticoid receptor. Brain Behav. Immun. 2019, 78, 21–30. [Google Scholar] [CrossRef]
  121. Abell, J.G.; Shipley, M.J.; Ferrie, J.E.; Kivimäki, M.; Kumari, M. Recurrent short sleep, chronic insomnia symptoms and salivary cortisol: A 10-year follow-up in the Whitehall II study. Psychoneuroendocrinology 2016, 68, 91–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Irwin, M.R.; Wang, M.; Campomayor, C.O.; Collado-Hidalgo, A.; Cole, S. Sleep deprivation and activation of morning levels of cellular and genomic markers of inflammation. Arch. Intern. Med. 2006, 166, 1756–1762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Rigobon, A.V.; Kanagasabai, T.; Taylor, V.H. Obesity moderates the complex relationships between inflammation, oxidative stress, sleep quality and depressive symptoms. BMC Obes. 2018, 5, 32. [Google Scholar] [CrossRef] [PubMed]
  124. Jansen, E.C.; Dunietz, G.L.; Chervin, R.D.; Baylin, A.; Baek, J.; Banker, M.; Song, P.X.K.; Cantoral, A.; Tellez Rojo, M.M.; Peterson, K.E. Adiposity in Adolescents: The Interplay of Sleep Duration and Sleep Variability. J.Pediatr. 2018, 203, 309–316. [Google Scholar] [CrossRef] [Green Version]
  125. Okun, M.L.; Hall, M.; Coussons-Read, M.E. Sleep disturbances increase interleukin-6 production during pregnancy: Implications for pregnancy complications. Reprod. Sci. 2007, 14, 560–567. [Google Scholar] [CrossRef]
  126. Okun, M.L.; Roberts, J.M.; Marsland, A.L.; Hall, M. How disturbed sleep may be a risk factor for adverse pregnancy outcomes. Obstet. Gynecol. Surv. 2009, 64, 273–280. [Google Scholar] [CrossRef] [Green Version]
  127. Simpson, N.; Dinges, D.F. Sleep and inflammation. Nutr. Rev. 2007, 65, S244–S252. [Google Scholar] [CrossRef]
  128. Hall, M.H.; Smagula, S.F.; Boudreau, R.M.; Ayonayon, H.N.; Goldman, S.E.; Harris, T.B.; Naydeck, B.L.; Rubin, S.M.; Samuelsson, L.; Satterfield, S.; et al. Association between sleep duration and mortality is mediated by markers of inflammation and health in older adults: The Health, Aging and Body Composition Study. Sleep 2015, 38, 189–195. [Google Scholar] [CrossRef]
  129. Sariyildiz, M.A.; Batmaz, I.; Bozkurt, M.; Bez, Y.; Cetincakmak, M.G.; Yazmalar, L.; Ucar, D.; Celepkolu, T. Sleep quality in rheumatoid arthritis: Relationship between the disease severity, depression, functional status and the quality of life. J. Clin. Med. Res. 2014, 6, 44–52. [Google Scholar] [CrossRef] [Green Version]
  130. Løppenthin, K.; Esbensen, B.A.; Jennum, P.; Østergaard, M.; Tolver, A.; Thomsen, T.; Midtgaard, J. Sleep quality and correlates of poor sleep in patients with rheumatoid arthritis. Clin. Rheumatol. 2015, 34, 2029–2039. [Google Scholar] [CrossRef]
  131. Austad, C.; Kvien, T.; Olsen, I.C.; Uhlig, T. Sleep disturbance in patients with rheumatoid arthritis is related to fatigue, disease activity, and other patient-reported outcomes. Scand. J. Rheumatol. 2017, 46, 95–103. [Google Scholar] [CrossRef] [PubMed]
  132. Grabovac, I.; Haider, S.; Berner, C.; Lamprecht, T.; Fenzl, K.H.; Erlacher, L.; Quittan, M.; Dorner, T.E. Sleep Quality in Patients with Rheumatoid Arthritis and Associations with Pain, Disability, Disease Duration, and Activity. J. Clin. Med. 2018, 7, 336. [Google Scholar] [CrossRef] [Green Version]
  133. Luyster, F.S.; Chasens, E.R.; Wasko, M.C.; Dunbar-Jacob, J. Sleep quality and functional disability in patients with rheumatoid arthritis. J. Clin. Sleep Med. 2011, 7, 49–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zoli, A.; Lizzio, M.M.; Ferlisi, E.M.; Massafra, V.; Mirone, L.; Barini, A.; Scuderi, F.; Bartolozzi, F.; Magaró, M. ACTH, cortisol and prolactin in active rheumatoid arthritis. Clin. Rheumatol. 2002, 21, 289–293. [Google Scholar] [CrossRef] [PubMed]
  135. Zoli, A.; Ferlisi, E.M.; Lizzio, M.; Altomonte, L.; Mirone, L.; Barini, A.; Scuderi, F.; Bartolozzi, F.; Magaro, M. Prolactin/cortisol ratio in rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2002, 966, 508–512. [Google Scholar] [CrossRef] [PubMed]
  136. Sulli, A.; Maestroni, G.J.; Villaggio, B.; Hertens, E.; Craviotto, C.; Pizzorni, C.; Briata, M.; Seriolo, B.; Cutolo, M. Melatonin serum levels in rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2002, 966, 276–283. [Google Scholar] [CrossRef]
  137. Cutolo, M.; Villaggio, B.; Otsa, K.; Aakre, O.; Sulli, A.; Seriolo, B. Altered circadian rhythms in rheumatoid arthritis patients play a role in the disease’s symptoms. Autoimmun. Rev. 2005, 4, 497–502. [Google Scholar] [CrossRef]
  138. Bach, J.F. Infections and autoimmune diseases. J. Autoimmun. 2005, 25, 74–80. [Google Scholar] [CrossRef]
  139. Kivity, S.; Agmon-Levin, N.; Blank, M.; Shoenfeld, Y. Infections and autoimmunity--friends or foes? Trends Immunol. 2009, 30, 409–414. [Google Scholar] [CrossRef]
  140. Vojdani, A.A. Potential Link between Environmental Triggers and Autoimmunity. Autoimmune Dis. 2014, 2014, 437231. [Google Scholar] [CrossRef] [Green Version]
  141. Floreani, A.; Franceschet, I.; Cazzagon, N.; Spinazzè, A.; Buja, A.; Furlan, P.; Baldo, V.; Gershwin, M.E. Extrahepatic Autoimmune Conditions Associated with Primary Biliary Cirrhosis. Clinic. Rev. Allerg. Immunol. 2015, 48, 192–197. [Google Scholar] [CrossRef] [PubMed]
  142. Arleevskaya, M.I.; Gabdoulkhakova, A.G.; Filina, J.V.; Zabotin, A.I.; Tsibulkin, A.P. Mononuclear Phagocytes in Rheumatoid Arthritis Patients and their Relatives—Family Similarity. Open Rheumatol. J. 2011, 5, 36–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Larionova, R.V.; Arleevskaya, M.I.; Kravtsova, O.A.; Validov, S.; Renaudineau, Y. In seroconverted rheumatoid arthritis patients a multi-reactive anti-herpes IgM profile is associated with disease activity. Clin. Immunol. 2019, 200, 19–23. [Google Scholar] [CrossRef]
  144. Ip, W.K.; Lau, Y.L.; Chan, S.Y.; Mok, C.C.; Chan, D.; Tong, K.K.; Lau, C.S. Mannose-binding lectin and rheumatoid arthritis in southern Chinese. Arthritis Rheum. 2000, 43, 1679–1687. [Google Scholar] [CrossRef]
  145. Jacobsen, S.; Madsen, H.O.; Klarlund, M.; Jensen, T.; Skjødt, H.; Jensen, K.E.; Svejgaard, A.; Garred, P.; TIRA Group. The influence of mannose binding lectin polymorphisms on disease outcome in early polyarthritis. TIRA Group. J. Rheumatol. 2001, 28, 935–942. [Google Scholar]
  146. Olsen, H.G.; Lien, S.; Gautier, M.; Nilsen, H.; Roseth, A.; Berg, P.R.; Sundsaasen, K.K.; Svendsen, M.; Meuwissen, T.H. Mapping of a milk production quantitative trait locus to a 420-kb region on bovine chromosome 6. Genetics 2005, 169, 275–283. [Google Scholar] [CrossRef] [Green Version]
  147. Plenge, R.M. Rheumatoid arthritis genetics: 2009 update. Curr.Rheumatol. Rep. 2009, 11, 351–356. [Google Scholar] [CrossRef] [PubMed]
  148. Diogo, D.; Bastarache, L.; Liao, K.P.; Graham, R.R.; Fulton, R.S.; Greenberg, J.D.; Eyre, S.; Bowes, J.; Cui, J.; Lee, A.; et al. TYK2 Protein-Coding Variants Protect against Rheumatoid Arthritis and Autoimmunity, with No Evidence of Major Pleiotropic Effects on Non-Autoimmune Complex Traits. PLoS ONE 2015, 10, e0122271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Oki, M.; Watanabe, N.; Owada, T.; Oya, Y.; Ikeda, K.; Saito, Y.; Matsumura, R.; Seto, Y.; Iwamoto, I.; Nakajima, H. A functional polymorphism in B and T lymphocyte attenuator is associated with susceptibility to rheumatoid arthritis. Clin. Dev. Immunol. 2011, 305656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Mattey, D.L.; Hassell, A.B.; Plant, M.; Dawes, P.T.; Ollier, W.R.; Jones, P.W.; Fryer, A.A.; Alldersea, J.E.; Strange, R.C. Association of polymorphism in glutathione S-transferase loci with susceptibility and outcome in rheumatoid arthritis: Comparison with the shared epitope. Ann. Rheum. Dis. 1999, 58, 164–168. [Google Scholar] [CrossRef] [Green Version]
  151. Nemec, P.; Goldbergová, M.; Swobodnik, T.; Polásková, D.; Soucek, M.; Vasků, A. Polymorfizmus v promotorovéoblasti genu pro MMP-2 u revmatoidníartritidy. VnitrLek 2006, 52, 348–354. [Google Scholar]
  152. Ling, S.; Li, Z.; Borschukova, O.; Xiao, L.; Pumpens, P.; Holoshitz, J. The rheumatoid arthritis shared epitope increases cellular susceptibility to oxidative stress by antagonizing an adenosine-mediated anti-oxidative pathway. Arthritis Res. Ther. 2007, 9, R5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Arleevskaya, M.I.; Albina, S.; Larionova, R.V.; Gabdoulkhakova, A.G.; Lemerle, J.; Renaudineau, Y. Prevalence and Incidence of Upper Respiratory Tract Infection Events Are Elevated Prior to the Development of Rheumatoid Arthritis in First-Degree Relatives. Front. Immunol. 2018, 9, 2771. [Google Scholar] [CrossRef] [PubMed]
  154. Toussirot, E.; Wendling, D.; Tiberghien, P.; Luka, J.; Roudier, J. Decreased T cell precursor frequencies to Epstein-Barr virus glycoprotein Gp110 in peripheral blood correlate with disease activity and severity in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2000, 59, 533–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Saal, J.G.; Krimmel, M.; Steidle, M.; Gerneth, F.; Wagner, S.; Fritz, P.; Koch, S.; Zacher, J.; Sell, S.; Einsele, H.; et al. Synovial Epstein-Barr virus infection increases the risk of rheumatoid arthritis in individuals with the shared HLA-DR4 epitope. Arthritis Rheum. 1999, 42, 1485–1496. [Google Scholar] [CrossRef]
  156. Wu, F.; Zhang, W.; Zhang, L.; Wu, J.; Li, C.; Meng, X.; Wang, X.; He, P.; Zhang, J. NRAMP1, VDR, HLA-DRB1, and HLA-DQB1 gene polymorphisms in susceptibility to tuberculosis among the Chinese Kazakh population: A case-control study. Biomed. Res. Int. 2013, 2013, 484535. [Google Scholar] [CrossRef]
  157. Du, J.; Liu, J.; Gu, J.; Zhu, P. HLA-DRB1*09 is associated with increased incidence of cytomegalovirus infection and disease after allogeneic hematopoietic stem cell transplantation. Biol. Blood. Marrow. Transplant. 2007, 13, 1417–1421. [Google Scholar] [CrossRef] [Green Version]
  158. Kekik, C.; Besisik, S.K.; Seyhun, Y.; Oguz, F.S.; Sargin, D.; Carin, M.N. Relationship between HLA tissue type, CMV infection, and acute graft-vs-host disease after allogeneic hematopoietic stem cell transplantation: Single-center experience. Transplant Proc. 2009, 41, 3859–3862. [Google Scholar] [CrossRef]
  159. Kallio-Laine, K.; Seppänen, M.; Aittoniemi, J.; Kautiainen, H.; Seppälä, I.; Valtonen, V.; Färkkilä, M.; Kalso, E.; Lokki, M.L. HLA-DRB1*01 allele and low plasma immunoglobulin G1 concentration may predispose to herpes-associated recurrent lymphocytic meningitis. Hum. Immunol. 2010, 71, 179–181. [Google Scholar] [CrossRef]
  160. Acar, K.; Akı, S.Z.; Ozkurt, Z.N.; Bozday, G.; Rota, S.; TürközSucak, G. Factors associated with cytomegalovirus reactivation following allogeneic hematopoietic stem cell transplantation: Human leukocyte antigens might be among the risk factors. Turk. J. Haematol. 2014, 31, 276–285. [Google Scholar] [CrossRef]
  161. Hart, J.E.; Laden, F.; Puett, R.C.; Costenbader, K.H.; Karlson, E.W. Exposure to traffic pollution and increased risk of rheumatoid arthritis. Environ. Health Perspect. 2009, 117, 1065–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Essouma, M.; Noubiap, J.J. Is air pollution a risk factor for rheumatoid arthritis? J. Inflamm. 2015, 12, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Stolt, P.; Källberg, H.; Lundberg, I.; Sjögren, B.; Klareskog, L.; Alfredsson, L.; EIRA study group. Silica exposure is associated with increased risk of developing rheumatoid arthritis: Results from the Swedish EIRA study. Ann. Rheum. Dis. 2005, 64, 582–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Sverdrup, B.; Källberg, H.; Bengtsson, C.; Lundberg, I.; Padyukov, L.; Alfredsson, L.; Klareskog, L.; Epidemiological Investigation of Rheumatoid Arthritis Study Group. Association between occupational exposure to mineral oil and rheumatoid arthritis: Results from the Swedish EIRA case-control study. Arthritis Res. Ther. 2005, 7, R1296–R1303. [Google Scholar] [CrossRef] [Green Version]
  165. Miller, F.W. Is occupational exposure to mineral oil a risk factor for rheumatoid arthritis? Nat. Clin. Pract. Rheumatol. 2006, 2, 130–131. [Google Scholar] [CrossRef]
  166. Sverdrup, B.M.; Källberg, H.; Klareskog, L.; Epidemiological Investigation of Rheumatoid Arthritis Study Group. Usage of skin care products and risk of rheumatoid arthritis: Results from the Swedish EIRA study. Arthritis Res. Ther. 2012, 14, R41. [Google Scholar] [CrossRef] [Green Version]
  167. Shin, J.; Lee, J.; Lee, J.; Ha, E.H. Association between Exposure to Ambient Air Pollution and Rheumatoid Arthritis in Adults. Int. J. Environ. Res. Public Health 2019, 16, 1227. [Google Scholar] [CrossRef] [Green Version]
  168. Murphy, D.; Bellis, K.; Hutchinson, D. Vapour, gas, dust and fume occupational exposures in male patients with rheumatoid arthritis resident in Cornwall (UK) and their association with rheumatoid factor and anti-cyclic protein antibodies: A retrospective clinical study. BMJ Open 2018, 8, e021754. [Google Scholar] [CrossRef] [Green Version]
  169. Balogh, E.; Veale, D.J.; McGarry, T.; Orr, C.; Szekanecz, Z.; Ng, C.T.; Fearon, U.; Biniecka, M. Oxidative stress impairs energy metabolism in primary cells and synovial tissue of patients with rheumatoid arthritis. Arthritis Res. Ther. 2018, 20, 95. [Google Scholar] [CrossRef]
  170. Khojah, H.M.; Ahmed, S.; Abdel-Rahman, M.S.; Hamza, A.B. Reactive oxygen and nitrogen species in patients with rheumatoid arthritis as potential biomarkers for disease activity and the role of antioxidants. Free Radic. Biol. Med. 2016, 97, 285–291. [Google Scholar] [CrossRef]
  171. Bonizzi, G.; Piette, J.; Merville, M.P.; Bours, V. Cell type-specific role for reactive oxygen species in nuclear factor-kappaB activation by interleukin-1. Biochem. Pharmacol. 2000, 59, 7–11. [Google Scholar] [CrossRef]
  172. Bonizzi, G.; Piette, J.; Schoonbroodt, S.; Greimers, R.; Havard, L.; Merville, M.P.; Bours, V. Reactive oxygen intermediate-dependent NF-kappaB activation by interleukin-1beta requires 5-lipoxygenase or NADPH oxidase activity. Mol. Cell. Biol. 1999, 19, 1950–1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Torices, S.; Julia, A.; Muñoz, P.; Varela, I.; Balsa, A.; Marsal, S.; Fernández-Nebro, A.; Blanco, F.; López-Hoyos, M.; Martinez-Taboada, V.; et al. A functional variant of TLR10 modifies the activity of NFkB and may help predict a worse prognosis in patients with rheumatoid arthritis. Arthritis Res. Ther. 2016, 18, 221. [Google Scholar] [CrossRef]
  174. Cantagrel, A.; Degboé, Y. New autoantibodies associated with rheumatoid arthritis recognize posttranslationally modified self-proteins. Jt. Bone Spine 2016, 83, 11–17. [Google Scholar] [CrossRef] [PubMed]
  175. Verheul, M.K.; Böhringer, S.; van Delft, M.A.M.; Jones, J.D.; Rigby, W.F.C.; Gan, R.W.; Holers, V.M.; Edison, J.D.; Deane, K.D.; Janssen, K.M.J.; et al. Triple Positivity for Anti-Citrullinated Protein Autoantibodies, Rheumatoid Factor, and Anti-Carbamylated Protein Antibodies Conferring High Specificity for Rheumatoid Arthritis: Implications for Very Early Identification of At-Risk Individuals. Arthritis Rheumatol. 2018, 70, 1721–1731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Jónsson, H.; Helgason, J. Rheumatoid arthritis in an Icelandic textbook from 1782. Scand. J. Rheumatol. 1996, 25, 134–137. [Google Scholar] [CrossRef] [PubMed]
  177. Weyand, C.M.; Klimiuk, P.A.; Goronzy, J.J. Heterogeneity of rheumatoid arthritis: From phenotypes to genotypes. Springer Semin. Immunopathol. 1998, 20, 5–22. [Google Scholar] [CrossRef]
  178. Lan, K.C.; Lai, Y.J.; Cheng, H.H.; Tsai, N.C.; Su, Y.T.; Tsai, C.C.; Hsu, T.Y. Levels of sex steroid hormones and their receptors in women with preeclampsia. Reprod. Biol. Endocrinol. 2020, 18, 12. [Google Scholar] [CrossRef] [Green Version]
  179. Visser, N.; van Rijn, B.B.; Rijkers, G.T.; Franx, A.; Bruinse, H.W. Inflammatory changes in preeclampsia: Current understanding of the maternal innate and adaptive immune response. Obstet. Gynecol. Surv. 2007, 62, 191–201. [Google Scholar] [CrossRef]
  180. Henley, D.; Brown, S.; Pennell, C.; Lye, S.; Torpy, D.J. Evidence for central hypercortisolism and elevated blood pressure in adolescent offspring of mothers with pre-eclampsia. Clin. Endocrinol. (Oxf.) 2016, 85, 583–589. [Google Scholar] [CrossRef]
  181. Kalpakcioglu, B.; Senel, K. The interrelation of glutathione reductase, catalase, glutathione peroxidase, superoxide dismutase, and glucose-6-phosphate in the pathogenesis of rheumatoid arthritis. Clin. Rheumatol. 2008, 27, 141–145. [Google Scholar] [CrossRef] [PubMed]
  182. Bijl, M.; Horst, G.; Limburg, P.C.; Kallenberg, C.G. Effects of smoking on activation markers, Fas expression and apoptosis of peripheral blood lymphocytes. Eur. J. Clin. Investig. 2001, 31, 550–553. [Google Scholar] [CrossRef] [PubMed]
  183. Sopori, M. Effects of cigarette smoke on the immune system. Nat. Rev. Immunol. 2002, 2, 372–377. [Google Scholar] [CrossRef] [PubMed]
  184. Reynolds, H.Y. Lung inflammation: Normal host defense or a complication of some diseases? Annu. Rev. Med. 1987, 38, 295–323. [Google Scholar] [CrossRef]
  185. Wegner, N.; Lundberg, K.; Kinloch, A.; Fisher, B.; Malmström, V.; Feldmann, M.; Venables, P.J. Autoimmunity to specific citrullinated proteins gives the first clues to the etiology of rheumatoid arthritis. Immunol. Rev. 2010, 233, 34–54. [Google Scholar] [CrossRef]
  186. Zeilinger, S.; Kühnel, B.; Klopp, N.; Baurecht, H.; Kleinschmidt, A.; Gieger, C.; Weidinger, S.; Lattka, E.; Adamski, J.; Peters, A.; et al. Tobacco smoking leads to extensive genome-wide changes in DNA methylation. PLoS One. 2013, 8, e63812. [Google Scholar] [CrossRef]
  187. Padyukov, L.; Silva, C.; Stolt, P.; Alfredsson, L.; Klareskog, L. A gene-environment interaction between smoking and shared epitope genes in HLA-DR provides a high risk of seropositive rheumatoid arthritis. Arthritis Rheum. 2004, 50, 3085–3092. [Google Scholar] [CrossRef]
  188. Park, B.; Koo, S.M.; An, J.; Lee, M.; Kang, H.Y.; Qiao, D.; Cho, M.H.; Sung, J.; Silverman, E.K.; Yang, H.J.; et al. Genome-wide assessment of gene-by-smoking interactions in COPD. Sci. Rep. 2018, 8, 9319. [Google Scholar] [CrossRef]
  189. Chen, J.; Huang, F.; Liu, M.; Duan, X.; Xiang, Z. Genetic polymorphism of glutathione S-transferase T1 and the risk of rheumatoid arthritis: A meta-analysis. Clin. Exp. Rheumatol. 2012, 30, 741–747. [Google Scholar]
  190. Brown, K.D.; Claudio, E.; Siebenlist, U. The roles of the classical and alternative nuclear factor-kappaB pathways: Potential implications for autoimmunity and rheumatoid arthritis. Arthritis Res Ther. 2008, 10, 212. [Google Scholar] [CrossRef] [Green Version]
  191. Świerkot, J.; Nowak, B.; Czarny, A.; Zaczyńska, E.; Sokolik, R.; Madej, M.; Korman, L.; Sebastian, A.; Wojtala, P.; Lubiński, Ł.; et al. The Activity of JAK/STAT and NF-κB in Patients with Rheumatoid Arthritis. Adv Clin Exp Med. 2016, 25, 709–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Anisman, H.; Merali, Z. Cytokines, stress and depressive illness: Brain-immune interactions. Ann Med. 2003, 35, 2–11. [Google Scholar] [CrossRef] [PubMed]
  193. Aschbacher, K.; Epel, E.; Wolkowitz, O.M.; Prather, A.A.; Puterman, E.; Dhabhar, F.S. Maintenance of a positive outlook during acute stress protects against pro-inflammatory reactivity and future depressive symptoms. Brain Behav. Immun. 2012, 26, 346–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Bulatov, E.; Khaiboullina, S.; dos Reis, H.-G.; Palotás, A.; Venkataraman, K.; Vijayalakshmi, M.; Rizvanov, A. Ubiquitin-Proteasome System: Promising Therapeutic Targets in Autoimmune and Neurodegenerative Diseases. BioNanoSci 2016, 6, 341–344. [Google Scholar] [CrossRef]
  195. Straub, R.H.; Schölmerich, J.; Zietz, B. Replacement therapy with DHEA plus corticosteroids in patients with chronic inflammatory diseases--substitutes of adrenal and sex hormones. Z. Rheumatol. 2000, 59 (Suppl. 2), 108–118. [Google Scholar] [CrossRef]
  196. Cutolo, M.; Sulli, A.; Pizzorni, C.; Craviotto, C.; Straub, R.H. Hypothalamic-pituitary-adrenocortical and gonadal functions in rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2003, 992, 107–117. [Google Scholar] [CrossRef]
  197. Masi, A.T.; Rehman, A.A.; Chatterton, R.T.; Wang, H.; Goertzen, N.J.; Elmore, K.B.; Aldag, J.C. Controlled Cohort Study of Serum Gonadal and Adrenocortical Steroid Levels in Males Prior to Onset of Rheumatoid Arthritis (pre-RA): A Comparison to pre-RA Females and Sex Differences among the Study Groups. Int. J. Rheumatol. 2013, 2013, 284145. [Google Scholar] [CrossRef] [Green Version]
  198. Cutolo, M.; Foppiani, L.; Prete, C.; Ballarino, P.; Sulli, A.; Villaggio, B.; Seriolo, B.; Giusti, M.; Accardo, S. Hypothalamic-pituitary-adrenocortical axis function in premenopausal women with rheumatoid arthritis not treated with glucocorticoids. J. Rheumatol. 1999, 26, 282–288. [Google Scholar]
  199. Straub, R.H.; Paimela, L.; Peltomaa, R.; Schölmerich, J.; Leirisalo-Repo, M. Inadequately low serum levels of steroid hormones in relation to interleukin-6 and tumor necrosis factor in untreated patients with early rheumatoid arthritis and reactive arthritis. Arthritis Rheum. 2002, 46, 654–662. [Google Scholar] [CrossRef]
  200. Jorgensen, C.; Sany, J. Modulation of the immune response by the neuro-endocrine axis in rheumatoid arthritis. Clin. Exp. Rheumatol. 1994, 12, 435–441. [Google Scholar]
  201. Geenen, R.; Van Middendorp, H.; Bijlsma, J.W. The impact of stressors on health status and hypothalamic-pituitary-adrenal axis and autonomic nervous system responsiveness in rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2006, 1069, 77–97. [Google Scholar] [CrossRef]
  202. Kozaci, D.L.; Chernajovsky, Y.; Chikanza, I.C. The differential expression of corticosteroid receptor isoforms in corticosteroid-resistant and -sensitive patients with rheumatoid arthritis. Rheumatology (Oxford) 2007, 46, 579–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Chatzikyriakidou, A.; Georgiou, I.; Voulgari, P.V.; Georgiadis, A.N.; Argyriou, E.S.; Drosos, A.A. Glucocorticoid receptor variants may predispose to rheumatoid arthritis susceptibility. Scand. J. Rheumatol. 2009, 38, 1–5. [Google Scholar] [CrossRef] [PubMed]
  204. van Oosten, M.J.; Dolhain, R.J.; Koper, J.W.; van Rossum, E.F.; Emonts, M.; Han, K.H.; Wouters, J.M.; Hazes, J.M.; Lamberts, S.W.; Feelders, R.A. Polymorphisms in the glucocorticoid receptor gene that modulate glucocorticoid sensitivity are associated with rheumatoid arthritis. Arthritis Res. Ther. 2010, 12, R159. [Google Scholar] [CrossRef] [Green Version]
  205. Donn, R.; Payne, D.; Ray, D. Glucocorticoid receptor gene polymorphisms and susceptibility to rheumatoid arthritis. Clin. Endocrinol. (Oxf) 2007, 67, 342–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Adlan, A.M.; Lip, G.Y.; Paton, J.F.; Kitas, G.D.; Fisher, J.P. Autonomic function and rheumatoid arthritis: A systematic review. Semin. Arthritis Rheum. 2014, 44, 283–304. [Google Scholar] [CrossRef] [PubMed]
  207. Kosek, E.; Altawil, R.; Kadetoff, D.; Finn, A.; Westman, M.; Le Maître, E.; Andersson, M.; Jensen-Urstad, M.; Lampa, J. Evidence of different mediators of central inflammation in dysfunctional and inflammatory pain--interleukin-8 in fibromyalgia and interleukin-1 β in rheumatoid arthritis. J. Neuroimmunol. 2015, 280, 49–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Koopman, F.A.; Tang, M.W.; Vermeij, J.; de Hair, M.J.; Choi, I.Y.; Vervoordeldonk, M.J.; Gerlag, D.M.; Karemaker, J.M.; Tak, P.P. Autonomic Dysfunction Precedes Development of Rheumatoid Arthritis: A Prospective Cohort Study. EBioMedicine 2016, 6, 231–237. [Google Scholar] [CrossRef] [Green Version]
  209. Igari, T.; Takeda, M.; Obara, K.; Ono, S. Catecholamine metabolism in the patients with rheumatoid arthritis. Tohoku J. Exp. Med. 1977, 122, 9–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Vlcek, M.; Rovensky, J.; Eisenhofer, G.; Radikova, Z.; Penesova, A.; Kerlik, J.; Imrich, R. Autonomic nervous system function in rheumatoid arthritis. Cell Mol. Neurobiol. 2012, 32, 897–901. [Google Scholar] [CrossRef]
  211. Rovensky, J.; Imrich, R.; Penesova, A.; Radikova, Z.; Scipova, A.; Vlcek, M.; Vigas, M. Adrenomedullary response to hypoglycemia in first-degree relatives of patients with rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2008, 1148, 552–555. [Google Scholar] [CrossRef] [PubMed]
  212. Imrich, R.; Rovensky, J.; Malis, F.; Zlnay, M.; Killinger, Z.; Kvetnansky, R.; Huckova, M.; Vigas, M.; Macho, L.; Koska, J. Low levels of dehydroepiandrosterone sulphate in plasma, and reduced sympathoadrenal response to hypoglycaemia in premenopausal women with rheumatoid arthritis. Ann. Rheum. Dis. 2005, 64, 202–206. [Google Scholar] [CrossRef] [PubMed]
  213. Wahle, M.; Hanefeld, G.; Brunn, S.; Straub, R.H.; Wagner, U.; Krause, A.; Häntzschel, H.; Baerwald, C.G. Failure of catecholamines to shift T-cell cytokine responses toward a Th2 profile in patients with rheumatoid arthritis. Arthritis Res. Ther. 2006, 8, R138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Wahle, M.; Pierer, M.; Krause, A.; Kolker, S.; Baerwald, C.G. Decreased catecholamine-induced cell death in B lymphocytes from patients with rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2002, 966, 425–428. [Google Scholar] [CrossRef]
  215. Baerwald, C.G.; Wahle, M.; Ulrichs, T.; Jonas, D.; von Bierbrauer, A.; von Wichert, P.; Burmester, G.R.; Krause, A. Reduced catecholamine response of lymphocytes from patients with rheumatoid arthritis. Immunobiology 1999, 200, 77–91. [Google Scholar] [CrossRef]
  216. Elbeialy, A.; Elbarbary, M.; Kamel, M. Peripheral beta-endorphin in rheumatoid arthritis. A correlation with the disease activity. Scand. J. Rheumatol. 1997, 26, 88–91. [Google Scholar] [CrossRef]
  217. Denko, C.W.; Aponte, J.; Gabriel, P.; Petricevic, M. Serum beta-endorphin in rheumatic disorders. J. Rheumatol. 1982, 9, 827–833. [Google Scholar]
  218. Almay, B.G.L.; Johansson, F.; Von Knorring, L.; T erenius, L.; Wahlstrom, A. Endorphins in chronic pain. I. Differences in CSF endorphin levels between organic and psychogenic pain syndromes. Pain 1978, 5, 153–162. [Google Scholar] [CrossRef]
  219. Wiedermann, C.J.; Sacerdote, P.; Mur, E.; Kinigadner, U.; Wicker, T.; Panerai, A.E.; Braunsteiner, H. Decreased immunoreactive beta-endorphin in mononuclear leucocytes from patients with rheumatic diseases. Clin. Exp. Immunol. 1992, 87, 178–182. [Google Scholar] [CrossRef]
  220. Yin, H.; Yu, M.; Cheng, H.; Zhang, F.; Gao, Y.; Lin, J.; Han, B.; Zhu, L. Beta-endorphin prevents collagen induced arthritis by neuroimmuno-regulation pathway. Neuroendocrinol. Lett. 2005, 26, 739–744. [Google Scholar]
  221. Mousa, S.A.; Straub, R.H.; Schäfer, M.; Stein, C. Beta-endorphin, Met-enkephalin and corresponding opioid receptors within synovium of patients with joint trauma, osteoarthritis and rheumatoid arthritis. Ann. Rheum. Dis. 2007, 66, 871–879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Takeba, Y.; Suzuki, N.; Kaneko, A.; Asai, T.; Sakane, T. Endorphin and enkephalin ameliorate excessive synovial cell functions in patients with rheumatoid arthritis. J. Rheumatol. 2001, 28, 2176–2183. [Google Scholar] [PubMed]
  223. Neeck, G.; Kluter, A.; Dotzlaw, H.; Eggert, M. Involvement of the glucocorticoid receptor in the pathogenesis of rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2002, 966, 491–495. [Google Scholar] [CrossRef] [PubMed]
  224. Waisberg, M.; Joseph, P.; Hale, B.; Beyersmann, D. Molecular and cellular mechanisms of cadmium carcinogenesis. Toxicology 2003, 192, 95–117. [Google Scholar] [CrossRef]
  225. Romoser, A.A.; Chen, P.L.; Berg, J.M.; Seabury, C.; Ivanov, I.; Criscitiello, M.F.; Sayes, C.M. Quantum dots trigger immunomodulation of the NFκB pathway in human skin cells. Mol. Immunol. 2011, 48, 1349–1359. [Google Scholar] [CrossRef] [Green Version]
  226. Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef]
  227. Lewis, J.B.; Wataha, J.C.; McCloud, V.; Lockwood, P.E.; Messer, R.L.; Tseng, W.Y. Au(III), Pd(II), Ni(II), and Hg(II) alter NF kappa B signaling in THP1 monocytic cells. J. Biomed. Mater. Res. A 2005, 74, 474–481. [Google Scholar] [CrossRef]
  228. Rudolf, E.; Cervinka, M. Nickel modifies the cytotoxicity of hexavalent chromium in human dermal fibroblasts. Toxicol. Lett. 2010, 197, 143–150. [Google Scholar] [CrossRef]
  229. Vivot, K.; Langlois, A.; Bietiger, W.; Dal, S.; Seyfritz, E.; Pinget, M.; Jeandidier, N.; Maillard, E.; Gies, J.P.; Sigrist, S. Pro-inflammatory and pro-oxidant status of pancreatic islet in vitro is controlled by TLR-4 and HO-1 pathways. PLoS One 2014, 9, e107656. [Google Scholar] [CrossRef]
  230. Shrivastava, H.Y.; Ravikumar, T.; Shanmugasundaram, N.; Babu, M.; Unni Nair, B. Cytotoxicity studies of chromium(III) complexes on human dermal fibroblasts. Free Radic. Biol. Med. 2005, 38, 58–69. [Google Scholar] [CrossRef]
  231. Altaf, H.; Revell, P.A. Evidence for active antigen presentation by monocyte/macrophages in response to stimulation with particles: The expression of NFκB transcription factors and costimulatory molecules. Inflammopharmacology 2013, 21, 279–290. [Google Scholar] [CrossRef] [PubMed]
  232. Rudolf, E.; Cervinka, M. The role of intracellular zinc in chromium(VI)-induced oxidative stress, DNA damage and apoptosis. Chem. Biol. Interact. 2006, 62, 212–227. [Google Scholar] [CrossRef] [PubMed]
  233. Zhou, E.; Li, Y.; Wei, Z.; Fu, Y.; Lei, H.; Zhang, N.; Yang, Z.; Xie, G. Schisantherin A protects lipopolysaccharide-induced acute respiratory distress syndrome in mice through inhibiting NF-κB and MAPKs signaling pathways. Int. Immunopharmacol. 2014, 22, 133–140. [Google Scholar] [CrossRef] [PubMed]
  234. Fubini, B.; Hubbard, A. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) generation by silica in inflammation and fibrosis. Free Radic Biol Med. 2003, 34, 1507–1516. [Google Scholar] [CrossRef]
  235. Tawfik, A.A.; Shiraishi, N.; Ninomiya, S.; Tajima, M.; Inomata, M.; Kitano, S. Activation of nuclear factor kappa B and induction of migration inhibitory factor in tumors by surgical stress of laparotomy versus carbon dioxide pneumoperitoneum: An animal experiment. Surg. Endosc. 2010, 24, 578–583. [Google Scholar] [CrossRef] [PubMed]
  236. Shilpa, G.; Renjitha, J.; Saranga, R.; Sajin, F.K.; Nair, M.S.; Joy, B.; Sasidhar, B.S.; Priya, S. Epoxyazadiradione Purified from the Azadirachtaindica Seed Induced Mitochondrial Apoptosis and Inhibition of NFκB Nuclear Translocation in Human Cervical Cancer Cells. Phytother. Res. 2017, 31, 1892–1902. [Google Scholar] [CrossRef]
  237. Yu, M.; Wang, Q.; Ma, Y.; Li, L.; Yu, K.; Zhang, Z.; Chen, G.; Li, X.; Xiao, W.; Xu, P.; et al. Aryl Hydrocarbon Receptor Activation Modulates Intestinal Epithelial Barrier Function by Maintaining Tight Junction Integrity. Int. J. Biol. Sci. 2018, 14, 69–77. [Google Scholar] [CrossRef]
  238. Zouridakis, A.; Simos, Y.V.; Verginadis, I.I.; Charalabopoulos, K.; Ragos, V.; Dounousi, E.; Boudouris, G.; Karkabounas, S.; Evangelou, A.; Peschos, D. Correlation of bioelectrical impedance analysis phase angle with changes in oxidative stress on end-stage renal disease patients, before, during, and after dialysis. Ren. Fail. 2016, 38, 738–743. [Google Scholar] [CrossRef] [Green Version]
  239. Ponnappan, U.; Yull, F.E.; Soderberg, L.S. Inhaled isobutyl nitrite inhibited macrophage inducible nitric oxide by blocking NFkappaB signaling and promoting degradation of inducible nitric oxide synthase-2. Int. Immunopharmacol. 2004, 4, 1075–1082. [Google Scholar] [CrossRef]
  240. Huang, Y.; Davidson, G.; Li, J.; Yan, Y.; Chen, F.; Costa, M.; Chen, L.C.; Huang, C. Activation of nuclear factor-kappaB and not activator protein-1 in cellular response to nickel compounds. Environ. Health Perspect. 2002, 110, 835–839. [Google Scholar] [CrossRef] [Green Version]
  241. Fetisova, I.N.; Mezhinsky, S.; Chasha, T.V.; Ratnikova, S.Y.; Fetisov, N.S. Polymorphism of detoxification system genes. Bull. Ivanovo Med. Acad. 2014, 19, 50–58. (In Russian) [Google Scholar]
  242. Yun, B.R.; El-Sohemy, A.; Cornelis, M.C.; Cornelis, M.C.; Bae, S.C. Glutathione S-transferase M1, T1, and P1 genotypes and rheumatoid arthritis. J. Rheumatol. 2005, 32, 992–997. [Google Scholar] [PubMed]
  243. Pawlik, A.; Wrzesniewska, J.; Fiedorowicz-Fabrycy, I.; Gawronska-Szklarz, B. The MDR1 3435 polymorphism in patients with rheumatoid arthritis. Int. J. Clin. Pharmacol. Ther. 2004, 42, 496–503. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Results of PubMed publications searching with keywords (A) “RheumatoidArthritis and risk factors” (10,120 results in 1969–2022), (B) “Rheumatoid arthritis andenvironment” (2933 results in 1946–2022).
Figure 1. Results of PubMed publications searching with keywords (A) “RheumatoidArthritis and risk factors” (10,120 results in 1969–2022), (B) “Rheumatoid arthritis andenvironment” (2933 results in 1946–2022).
Ijms 23 08140 g001
Figure 2. Search results in the GWAS Catalog: SNPs of 14 genes are associated with both RA and distress. The numbers of SNPs associated both with RA and distress are circled in red.
Figure 2. Search results in the GWAS Catalog: SNPs of 14 genes are associated with both RA and distress. The numbers of SNPs associated both with RA and distress are circled in red.
Ijms 23 08140 g002
Figure 3. Possible mechanisms of implementation of triggering role of ecotoxicants in RA. OS*—oxidative stress.
Figure 3. Possible mechanisms of implementation of triggering role of ecotoxicants in RA. OS*—oxidative stress.
Ijms 23 08140 g003
Figure 4. Mosaic of weak links of the body at RA risk.
Figure 4. Mosaic of weak links of the body at RA risk.
Ijms 23 08140 g004
Figure 5. Network of challenging non-genetic factors.
Figure 5. Network of challenging non-genetic factors.
Ijms 23 08140 g005
Figure 6. Impact of non-genetic factors on weak links of the body under RA risk.
Figure 6. Impact of non-genetic factors on weak links of the body under RA risk.
Ijms 23 08140 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Arleevskaya, M.; Takha, E.; Petrov, S.; Kazarian, G.; Renaudineau, Y.; Brooks, W.; Larionova, R.; Korovina, M.; Valeeva, A.; Shuralev, E.; et al. Interplay of Environmental, Individual and Genetic Factors in Rheumatoid Arthritis Provocation. Int. J. Mol. Sci. 2022, 23, 8140. https://doi.org/10.3390/ijms23158140

AMA Style

Arleevskaya M, Takha E, Petrov S, Kazarian G, Renaudineau Y, Brooks W, Larionova R, Korovina M, Valeeva A, Shuralev E, et al. Interplay of Environmental, Individual and Genetic Factors in Rheumatoid Arthritis Provocation. International Journal of Molecular Sciences. 2022; 23(15):8140. https://doi.org/10.3390/ijms23158140

Chicago/Turabian Style

Arleevskaya, Marina, Elena Takha, Sergey Petrov, Gevorg Kazarian, Yves Renaudineau, Wesley Brooks, Regina Larionova, Marina Korovina, Anna Valeeva, Eduard Shuralev, and et al. 2022. "Interplay of Environmental, Individual and Genetic Factors in Rheumatoid Arthritis Provocation" International Journal of Molecular Sciences 23, no. 15: 8140. https://doi.org/10.3390/ijms23158140

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

Article Metrics

Back to TopTop