Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2016 Nov 26;242(4):355–373. doi: 10.1177/1535370216681549

Major involvement of bacterial components in rheumatoid arthritis and its accompanying oxidative stress, systemic inflammation and hypercoagulability

Etheresia Pretorius 1,, Oore-Ofe Akeredolu 1, Prashilla Soma 1, Douglas B Kell 2,3,4
PMCID: PMC5298544  PMID: 27889698

Abstract

We review the evidence that infectious agents, including those that become dormant within the host, have a major role to play in much of the etiology of rheumatoid arthritis and the inflammation that is its hallmark. This occurs in particular because they can produce cross-reactive (auto-)antigens, as well as potent inflammagens such as lipopolysaccharide that can themselves catalyze further inflammagenesis, including via β-amyloid formation. A series of observables coexist in many chronic, inflammatory diseases as well as rheumatoid arthritis. They include iron dysregulation, hypercoagulability, anomalous morphologies of host erythrocytes, and microparticle formation. Iron dysregulation may be responsible for the periodic regrowth and resuscitation of the dormant bacteria, with concomitant inflammagen production. The present systems biology analysis benefits from the philosophical idea of “coherence,” that reflects the principle that if a series of ostensibly unrelated findings are brought together into a self-consistent narrative, that narrative is thereby strengthened. As such, we provide a coherent and testable narrative for the major involvement of (often dormant) bacteria in rheumatoid arthritis.

Keywords: Rheumatoid arthritis, dormancy, iron dysregulation, atopobiosis, infectious agents, lipopolysaccharides, Proteus, inflammation, comorbidities

Impact statement

Rheumatoid arthritis (RA) is accompanied by long-term inflammation that is mediated by cytokines and cross-reactive (auto-)antigens. Here we suggest one explanation is the presence of a (dormant) microbiome in RA that sheds the highly potent inflammagen, lipopolysaccharide lipopolysaccharides (LPS) to catalyze inflammagenesis, including via β-amyloid formation. We discuss various co-existing features in RA, including iron dysregulation, hypercoagulability, anomalous morphologies of host erythrocytes, and microparticle formation. We review literature and provide coherent evidence that an aberrant blood microbiome in RA has a major involvement in the development, progression, and therefore over-all etiology of the disease.

Introduction: Disease background

RA is a complex and heterogeneous disease, sometimes classified as a syndrome with shared clinical manifestations.1 It is the most common immune-related chronic, inflammatory, autoimmune disease and affects approximately 0.5–1% of the adult population worldwide. It occurs as 20–50 cases per 100,000 annually, most commonly in women over 40.25 Although this is not yet apparently a mainstream recognition, a frankly overwhelming amount of epidemiological and experimental evidence, that we shall review here, indicates a microbial origin for RA. The clinical features of RA involve the presence of systemic inflammation, with various imbalances between pro- and anti-inflammatory cytokine activities, which may lead to multisystem immune complications.4 In RA patients, serum or plasma levels of cytokines are considered to indicate the severity of disease4 and this pathophysiologic presence of pro-inflammatory cytokines is known to be involved in the degradation of bone and cartilage.4 Due to the complexity of the disease, treatment and disease tracking after diagnosis is very difficult. In this article, we shall review briefly current knowledge regarding the involvement of cytokines and other markers in RA, which are also the hallmarks of systemic inflammation in all other inflammatory conditions. We also discuss clot hypercoagulability and platelet and erythrocyte (RBC) changes, that is consequent upon this persistent systemic inflammation, and how microparticle formation (from both platelets and RBCs) is a characteristic feature of RA. We then review a considerable literature (summarized in part in two books6,7) that suggests that the presence of a variety of detritus produced from walls and membranes of Gram-negative and other bacteria (including wall-less forms) may play a fundamental role in RA development, as well as the accompanying cardiovascular disease and systemic inflammation seen in RA. We discuss how the exposure of genetically susceptible individuals to environmental factors (1) that can act as triggers (2), cause an immunological reaction, followed by an autoimmune response (3), can result in RA (4). We review a plethora of evidence, collectively referred to as Ebringer’s theory (5), that points to the environmental trigger as microbial (particularly from e.g. urinary tract infections) (6). We then look at the role of LPS from these microbes (7) in causing an imbalance between pro- and anti-inflammatory cytokines, followed by systemic inflammation, and the effect on the cardiovascular and hematological health of the RA patient (8) (see Figure 1). Finally, recognizing the lack of easy and accessible biomarkers, we suggest that in a truly precision medicine approach,8 hypercoagulability and also microparticle presence, as well as LPS and β-amyloid analysis could play an important role in tracking the progression of the disease. A MEDLINE, Google Scholar, Scopus, and Web of Science search was done to review the literature.

Figure 1.

Figure 1

Genetically susceptible individuals exposed to environmental factors (1) that act as triggers (2) cause an immunological reaction followed by an autoimmune response (3) that may lead to rheumatoid arthritis (RA) (4). We discuss Ebringer’s theory (5), suggesting the cause of the trigger being microbes (6) and the role of LPS (7) that may result in an imbalance between pro- and anti-inflammatory cytokines, followed by systemic inflammation and the effect on the cardiovascular and hematological health of the RA patient (8). (A color version of this figure is available in the online journal.)

Epidemiology

An initial analysis of the potential causes of a disease is wise to explain any unusual epidemiological features it may possess,9 following appropriate controls for their veracity.10 Clearly, within an overall prevalence of ca 0.5–1%, one feature of this disease is its considerable predilection for women (69% of cases in a recent UK survey5) over men, despite some reduction in female prevalence attributed to the use of oral contraceptives.11,12 This is a striking difference of approximately two-or three-fold (see also literature2,1215). Clearly, it might be linked to hormonal differences, or to something on the X-chromosome, but we know of no persuasive study that suggests what that might be.

Twin studies “show concordance rates of 15% to 30% between monozygotic twins and 5% among dizygotic twins,14 suggesting that 50% to 60% of RA cases are due to genetic factors.16,17” Other studies comparing monozygotic twins alone show an occurrence in a second twin, when a first twin manifests the disease, as just 12% in Finland,18 15% for the UK,19 and 10% in Denmark.20 Thus, environmental influences have a major influence. Consequently, the leading hypothesis for RA (and indeed for most other autoimmune disorders) is that RA is the result of an environmental exposure or “trigger” in a genetically more susceptible individual, that causes an immunological reaction to the triggering antigen that happens to share one or more epitopes with a host protein (and see Kell and Pretorius21), thus manifesting the autoimmune responses. What might be the most common triggers? One possibility to link the triggers with infection is to look at the presence of flares. Flares are defined as a worsening of signs and symptoms of sufficient intensity and duration to lead to changes in therapy as per the Outcome Measures in Rheumatology Clinical Trials (OMERACT) RA Flare Definition Working Group, developed at OMERACT 9 in 2008.22 This working Group developed a standardized method for description and measurement of “flare in RA” to guide individual patient treatment.23

A very high proportion of sufferers were actually exposed to an infection before their disease was diagnosed, but sadly these kinds of data are not typically recorded properly in the scientific literature. Consequently, and as this becomes increasingly easy with electronic health records, we do encourage clinical readers to make such analyses available. However, as with several related diseases (e.g. literature21,2428), our role as systems biologists is to put together a coherent, systems biology picture, and with all the evidence that we shall review below, it is very clear indeed that RA is driven by a microbial component. To this end, one very major (and in our view clear) differentiator between women and men is the equally more common, anatomy-based prevalence in women over men of urinary tract infection.2931 This is the first “plank” in Ebringer’s impressive series of arguments (most recently at6,9,32,33) that actually gives a satisfying and coherent account of at least one microbial origin for RA, Proteus spp., that we now review.

Ebringer’s theory (with experiments) that Proteus infection from the urinary tract is a major cause of RA

Ebringer sets his work in a Popperian framework (for reviews of that see literature3436) but we consider that it is more conveniently set in a systems biology manner as a logical series or chain of intellectually linked events, and this is what we do here. Ebringer (we like especially Ebringer et al.32) makes the following 10 claims (the many supporting references are in the paper):

  1. HLA-DR4 lymphocytes injected into a rabbit evoke specific antibodies against Proteus cells (mainly the species mirabilis and vulgaris).

  2. Antibodies to Proteus spp. are present in RA patients from 14 different countries.

  3. Antibodies to Proteus bacteria in RA patients are disease-specific since no such antibodies are found in other conditions.

  4. When RA patients have high titers of antibodies to Proteus such bacteria are found in urinary cultures.

  5. Only Proteus bacteria and no other microbes evoke significantly elevated antibodies in RA patients (this is not 100% true, see below).

  6. A “shared epitope” EQR(K)RAA shows “molecular mimicry” with the related sequence ESRRAL found in Proteus hemolysis.

  7. Proteus urease contains a sequence IRRET which has “molecular mimicry” with the related LRREI found in collagen XI of hyaline cartilage.

  8. Sera obtained from RA patients have cytopathic properties against sheep red cells coated with the cross-reacting EQR(K)RAA and LRREI self-antigen peptides.

  9. Proteus sequences in hemolysin and urease as well as the self-antigens, HLA-DR1/4 and collagen XI, each contain an arginine doublet, thereby providing a substrate for peptidyl arginine deiminase (PAD) to give rise to citrulline, which is the main antigenic component of CCP, antibodies to which are found in early cases of RA.

  10. Antibodies to Proteus come not only from sequences cross reacting to self-antigens but also from non-cross reacting sequences, thereby indicating that active RA patients have been exposed to infection by Proteus.

Taken together, these arguments show strongly that microbes, especially those derived from urinary tract infections, can act as triggers of autoimmunity, via established epitopes or antigens. We should point out, however, that many other studies (and it is highly doubtful that there could be a unitary cause) indicate antibody and PCR-based evidence for the presence or role of other microbes in RA. Both gut dysbioses and a changed oral microbiota have also been implicated. There is also evidence of a significant association between periodontitis and RA.3743 Gut dysbioses are also frequently found in RA individuals44,45 and it was recently shown that dysbioses in RA patients may reflect an unusual abundance of certain rare bacterial lineages.46 Normalizing the gut microbiota was also suggested in assessing prognoses and in the treatment of RA.45,46 Some of these other microbes that are associated with RA are listed in Table 1 (and see also literature21,27,28).

Table 1.

Some other prokaryotic microorganisms besides Proteus spp. that have been implicated in RA

Organism Evidence Selected references
Campylobacter Microbiology 47
Chlamydia trachomatis Synovial tissues 48
Escherichia coli Antibodies 49,50
Multiple organisms Review 27,5153
Mycoplasmas PCR, westerns, antibodies 5457
Porphyromonas gingivalis Antibodies, PCR, culture 39,5863
Staphylococcus aureus Microbiology of hip joint infections 64

This was a binary (presence/absence) assessment of the microbial contribution, but microbes have many properties beyond presence and absence. In particular, a notable and missing feature of most of these studies (including those of Ebringer) involves (i) the physiological state of the organisms in question, and (ii) what causes them to manifest their activities periodically (for instance as the “flares” characteristic of RA). This we therefore discuss next.

Dormancy, resuscitation, and iron dysregulation

Clinical or infection microbiologists typically recognize bacteria as being in one of two physiological macrostates, either being “alive” (on the basis of their being capable of replication, e.g. to form a colony on a petri dish containing a suitable solid medium), or if not being so capable then being assumed “dead.” However, these are not the only two major physiological states, and indeed they are probably the least common in natural environments! Importantly, the definition of these states is operational: the appearance of a microbe’s physiology also depends on the experiment being used to test it and is not of itself an “innate property” of the organism.6567 In environmental microbiology, most microbes are non-growing because they lack the nutrients and/or signaling molecules to replicate, but they are not (irreversibly) “dead.” They are best described as “dormant,” and the means by which they are brought back from an apparent state of non-aliveness to one in which they can be cultivated is conventionally known as “resuscitation.” We demonstrated this in a series of papers in laboratory cultures of various actinobacteria (e.g. literature65,6873), leading to the discovery of an autologous “wake-up” molecule, the “resuscitation promotion factor” or Rpf7478 that was necessary for resuscitating dormant bacteria in the presence of weak nutrient broth. Note that assays must be done under conditions of dilution to extinction,66,67 to avoid the possibility of simple regrowth of small numbers of cells that were always “alive” and never dormant.

In clinical microbiology, the terms “persister” and “persistent” are commonly used to refer to a phenotypically non-growing (but non-dead) subset of microbes, typically those that have been treated with but tolerant to antibiotics (e.g. literature27,28,7986). In general terms, these too are operationally dormant as defined above, though their relative physiological states (e.g. as judged by expression profiles) are not really established. We have recently summarized the evidence for a dormant blood microbiome including in red cells21,27,28 (and see also Damgaard et al.87) to complement other literature pertaining to white cells and tissues (e.g. literature8892). Such dormant cells are, of course, well placed to create inflammation via a continuing shedding of inflammatory agents such as LPS and molecules with antigenic properties as described above. We note that the emergence of infection may also accompany treatment of the disease,9395 although whether this is due to resuscitation (as we suspect96) or reinfection is not yet uniformly clear.

The question then arises as to what kinds of stimuli trigger the resuscitation. Two are well established. One is the stimulation of Gram-negative bacterial growth by the stress hormone noradrenaline (NA) and other auto-inducers.97103 One of the roles of NA is to act as a siderophore,104,105 since it is normally available iron that limits microbial replication in vivo,106111 a phenomenon that adds considerably to the undesirable and purely chemical effects of the second one, which is the presence and availability of unliganded iron that is liable to catalyze the highly damaging Fenton reaction25,112 (see Figure 2 adapted from Kell et al.27).

Figure 2.

Figure 2

A bacterial system contains distinct subpopulations (1), that we classify as culturable, dormant and non-culturable (2). Specific attention is given to persister cells (3), and the inter-relationship (4) between the subpopulations and phenotypic switching between culturability and dormancy (5). Throughout we follow a systems biology approach to suggest resuscitation due to various triggers like iron and noradrenaline (6). (A color version of this figure is available in the online journal.)

Pathophysiologic markers of inflammation in RA

As is well established, inflammatory agents such as LPS lead to the induction of inflammatory cytokines, most commonly IL-6, IL-1β, and TNF-α.129,147,201,142,159 Cytokine-mediated pathways are central to the pathogenesis of RA.119 A MEDLINE, Google Scholar, Scopus, and Web of Science assessment revealed that a plethora of cytokines and markers of inflammation are involved in RA pathology, and importantly, these cytokines are not only localized to the synovial fluid, but are systemically present and detectable in serum samples (see Table 2). Furthermore, a changed systemic cytokine activity is typically associated with oxidative stress, and this is also true in RA individuals.202205

Table 2.

Dysregulated inflammatory markers in Rheumatoid arthritis

Cytokines and other markers of inflammation Effect on RA Selected references
IL-1β Upregulated; strong stimulator of bone resorption. Linked to joint inflammation and cartilage and bone destruction in patients with RA. Present in serum of RA patients. 113118
IL-6 IL-6 is involved in the regulation of immune responses, hematopoiesis and inflammation and is found in abundance in the synovial fluid and serum of patients with RA and the level correlates with the disease activity and joint destruction. Present in serum of RA patients. 119,120,121,113, 114,121128
IL-8 It is a neutrophil-activating peptide and the degree of neutrophil turnover is linked to the anaerobic metabolism of the synovial cavity. Upregulated in RA; and IL-6/IL-1β co-stimulation increases IL-8 production. Present in serum of RA patients. 129,130135
IL-12 Linked to leukocyte migration, bone erosions and angiogenesis in RA. Present in serum of RA patients. 114,136141
IL-15 Upregulated and expressed in synovial fluid. Long-term retention of IL-15/IL-15R α complexes on the surfaces of monocytes and dendritic cells. Present in serum of RA patients. 142,143146
IL-16 Present in human synovial fibroblasts. Present in serum of RA patients. 147,148151
IL-17 Plays key roles in the propagation of joint inflammation, cartilage destruction, and bone erosion; IL-17 inhibits progenitor cells in RA cartilage; has regulatory roles in host defense and chronic inflammation which result in tissue damage and autoimmunity; shares downstream transcription factors with IL-1 and TNF-α; promote osteoclastogenesis. Present in serum of RA patients. 147,152,147,113,114, 123,132,153158
IL-18 Detected in synovial fluid in RA patients. Present in serum of RA patients. 159,137,160165
IL-23 IL-23 is essential for the differentiation of Th17 lymphocytes, a subtype of T lymphocyte implicated in RA and promotes promote osteoclastogenesis Present in serum of RA patients. 123,139,157,166168
IL-27 IL-27 upregulated and produced by antigen-presenting cells. Linked to leukocyte migration, bone erosions and angiogenesis. Present in serum of RA patients. 139,155,169175
IL-7, IL- 10- IL-19, IL-20, IL-22 IL-32, IL-35 All implicated in contributing to the pathogenesis of RA. 138,139,176
Toll-like receptor 2 (TLR-2) and TLR-4 Expressed in inflamed RA synovium, and the expression of these receptors is associated with the presence of inflammatory cytokines. expressed by cells within the RA joint and a variety of endogenous TLR ligands are present within the inflamed joints of patients with RA. 147,177180
TNF-α Promotes systemic inflammation and autoimmune pathology and is one of the cytokines that make up the acute phase reaction. Linked to joint inflammation and cartilage and bone destruction in patients with RA. Present in serum of RA patients. 119,114,117,120, 122,181184
interferon γ (IFNγ) Promotes autoimmune pathology and plays a role in immunity against intracellular pathogens; abundantly expressed in rheumatoid synovitis. Present in serum of RA patients. 113,185188
Prostaglandin E2 Acts as mediator of immune inflammation. 132,189,190
Thromboxane-A2 and COX-2 [cyclooxygenase (COX)-2/ thromboxane A2 (TxA2)] Thromboxane-A2 produced by activated platelets and has prothrombotic properties, while COX-2is responsible for the formation of thromboxane and prostaglandins. 191193
Iron Increased levels in synovial fluid and changed serum ferritin levels 194200

Systemic inflammation, cardiovascular disease, and RA

All of the above-mentioned cytokines and related inflammatory mediators are known to be involved in both systemic inflammation and cardiovascular disease, and it is also known that patients with RA carry an excess risk for cardiovascular disease,119,206210 i.e. that there is a comorbidity. Indeed, cells and cytokines implicated in RA pathogenesis are involved in the development and progression of atherosclerosis, (which is generally recognized as an inflammatory cardiovascular disease208). Analysis of RA patients selected from an RA clinic in South Africa (ethical clearance obtained) confirmed that indeed cardiovascular complications are an important part of the clinical profile of RA patients (see Table 3).

Table 3.

An analysis of the co-existing conditions of rheumatoid arthritis patients patients from a rheumatoid arthritis clinic in South Africa, showing baseline demographics of subjects (n = 38) with Rheumatoid Arthritis

Variable Mean ± (SD) for age, others N (%)
Age, years
 Males 53 ± (13.01)
 Females 55 ± (11.54)
Chronic medication: Females Males
 NSAID 21 (64%) 4 (80%)
 Prednisone 20 (61%) 5 (100%)
 Chloroquine 7 (21%) 2 (40%)
 Salazopyrin 8 (24%) 1 (20%)
 Opioids 13 (39%) 1 (20%)
 Anti-depressants 6 (18%) 1 (20%)
 Acid-reducers 17 (51%) 3 (60%)
 Anti-hypertensives 14 (42%) 5 (100%)
 Statins 10 (30%) 2 (40%)
Co-morbid conditions:
 Hypertension 21 (64%) 5 (100%)
 Dyslipidaemia 12 (36%) 1 (20%)
 Hypothyroidism 4 (12%)
 Asthma 3 (9%)
 Anaemia 3 (9%)
 Diabetes mellitus 1 (3%) 1 (20%)
 Gastric reflux 11 (33%) 3 (60%)

Cardiovascular comorbidities relate more than others to disease activity in RA, and particularly type 2 diabetes and hyperlipidemia were found to be associated with disease activity.211 Cardiovascular disease, and particularly diabetes, has also been linked to gut dysbioses and bacterial translocation.212216 Recently, it was also suggested that a co-morbidity index should be used both at baseline, and as a continuous variable in analyses in RA patients,217 as some co-morbidities are causally associated with RA and many others are related to its treatment.218

Systemic inflammation is entirely consistent with the plethora of diseases and comorbidities that exhibit iron dysregulation,24,112 raised serum ferritin,219 hypercoagulation and hypofibrinolysis,25 and anomalous morphological changes in both erythrocytes and fibrin.220,221 We thus turn to hypercoagulation in RA.

Hypercoagulation, erythrocyte (RBC), and platelet involvement in RA as a result of systemic inflammation

Another hallmark of systemic inflammation (as well as cardiovascular pathology) is clot hypercoagulability, and a hypercoagulable state is also found in RA,222231 together with a decreased clot lysis ability,25,225,226 possibly due to genuine amyloid formation.232 Systemic inflammation, oxidative stress, and hypercoagualability all affect erythrocytes (RBCs) and platelets. One of the changes due to the systemic inflammation and oxidative stress noticeable in RBCs and platelets is the production of cell-derived microparticles (MPs).219

Flow cytometry is the usual method to quantify MP233,234; unfortunately, the small size of these structures and lack of standardization in methodology complicates measurement.235 Mostly, as MP contain surface and cytoplasmic contents of the parent cells and bear phosphatidylserine, antibodies to specific cell surface markers and annexin V can be used for identification235,236 or for tissue factor-dependent FXa generation assays.237 Their sizes can vary but are of the order of 50 to 800 nm238 and are therefore easily detected on and around their mother-cell by scanning electron microscopy.209

In RA, circulating MPs exposing complement components or activator molecules are elevated,239 and their levels are correlated with disease activity.240 These MPs are also of great important in cardiovascular diseases, and this may be one reason for the enhanced cardiovascular morbidity and mortality seen in RA.240 Furthermore, MPs may contribute to the local hypercoagulation and fibrin deposition in inflamed joints of patients with RA.241

Particularly, MPs derived from platelets that are involved in various thrombotic events are elevated in RA patients, and these platelet-derived MPs may be an important role-player in RA240,242,243; they may even be used as a biomarker reflecting systemic cell activation in RA.244,245 Platelet MPs in RA have also been found to be responsible for detrimental effects on endothelial cells, thus supporting their role as biomarkers of vascular damage.246 Although RBC MP formation is not very well described in RA in particular, we recently discussed possible mechanisms by which RBC MP formation may occur in RA.245 The nature and distribution of lipids in RBC bilayers are altered in RA, showing a decreased level of cholesterol and phospholipids when compared to healthy controls.247 Changed levels of cholesterol found in the RBC phospholipid bilayer can determine its capacity to express phosphatidylserine (PS) on the outer leaflet, independent of ATP-driven flippases.248 It is also well known that oxidative stress in RA influences RBC membrane integrity (see Table 4), and this supports the possibility that portions of the RBC membrane may bud off to form RBC MPs.174

Table 4.

Markers taken as indicators of oxidative stress in RBCs

Marker Observed effect On RBCs
Peroxides Increased in RBCs249
Glutathione Oxidized in RBCs249251
Catalase Increased in RBCs250
Superoxide dismutase Increased in RBCs250
Malondialdehyde Increased in RBCs250
Membrane redox system Upregulated in RBCs250
Caspase-3/Calpain Increased in RBCs251
Enzyme activity, e.g. ATPases Altered in RBCs249,251254
2,3 Diphosphoglycerate Decreased in RBCs251,253,254

Pathologic changes in coagulation result directly in abnormal fibrin fiber formation during clotting, and MPs associated with platelets, as well as membrane changes of RBCs, can be visualized by scanning electron microscopy in blood smears from RA individuals. See Figure 3(a) for an example of healthy fibrin fiber formation versus pathological fibrin fiber formation in RA (Figure 3(b)), and a healthy platelet showing a clear cell body and pseudopodia formation with a smooth membrane (Figure 4(a)) versus platelets from RA where activation, spreading, and MP formation are visible (Figure 4(b) and (c)). Red arrows in Figure 4(c) possibly indicate round ultramicrobacteria, which differ from the much more irregularly shaped MPs.

Figure 3.

Figure 3

Fibrin fiber formation in the presence of thrombin (a) healthy fibrin and (b) rheumatoid arthritis fibrin with matted fibrin. Scale: 1 µm

Figure 4.

Figure 4

(a) A healthy platelet with prominent cell body and pseudopodia formation and smooth membrane; (b) two spreaded and activated platelets with microparticle formation (irregularly shaped structures closely associated with membranes (white arrows)) in rheumatoid arthritis; red arrows showing much rounder structures – possibly ultrabacteria; (c) a higher magnification of an RA platelet with microparticles budding off spreaded platelet. The scale bars are (a): 200 nm; (b): 1 µm and (c): 200 nm. (A color version of this figure is available in the online journal.)

Figure 5(a) and (b) shows a typical healthy RBC with no membrane changes, compared to normal RBCs, of a typical RA individual (Figure 5(c) and (d)), where RBC folding due to loss of structural integrity is seen in the presence of fibrin fiber formation (when thrombin is added (Figure 5(c))) and membrane changes are observed with MP formation (Figure 5(d)). The changed membrane surface is also confirmed via roughness analysis of RBC membranes using atomic force microscopy (AFM), where a significantly increased roughness was noted in RA RBCs compared to the case of healthy RBCs (Figure 6).

Figure 5.

Figure 5

(a) and (b) A representative healthy RBC (b is higher magnification showing the membrane); (c and d) A representative rheumatoid arthritis RBC with folding (c) and visible membrane microparticle formation. Scale bars for (a) and (c): 1 µm; scale bars for (b) and (d): 100 nm

Figure 6.

Figure 6

RBC membrane roughness analysis as seen with atomic force microscopy (AFM) of a representative micrograph from a healthy RBC (a) and a rheumatoid arthritis RBC (b). (A color version of this figure is available in the online journal.)

A note on amyloidogenesis and amyloidoses in RA

In a recent development,232,255,256 we have shown that the fibrin dense-matted deposits of the type shown in Figure 4(b) can be generated by tiny amounts of LPS, and that they are actually β-amyloid in nature. This has significant implication more generally,257 including diseases such as sepsis258 and pre-eclampsia.259 Amyloid fibrils of various kinds are widely recognized as inflammatory and cytotoxic (e.g. literature260263). We do not yet know the origins of the amyloid seen in RA, but it has long been known that amyloidoses are an important accompaniment (and potentially exacerbator) of RA (e.g. literature264268).

Present treatments treat the symptoms

There are two main pharmacological treatments for RA, viz. small molecule disease-modifying anti-rheumatic drugs (so-called sDMARDs) and biologicals (bDMARDs). We shall discuss these in general terms, especially for the former from the perspective of potential antibacterial effects. However, we need to start with present metrics for the severity of disease.

Metrics of disease severity

Given that there seems little general recognition of the real causes of RA, and that the symptoms including pain can be very severe, it is understandable, as this overall section implies, that many treatments are aimed at alleviating the symptoms. We also recognize that at least some of those that “work,” may do so, if inadvertently, by treating the causes as well. But to begin with, we need to consider the metrics of disease severity in common use.

A great many approaches to assessing disease severity exist, and the tendency to prescribe more drugs to those with more severe disease is necessarily269 a confounding factor.270 Databases for medical claims around disease severity have value,270 but they tend to lack information on important clinical variables, such as the number of tender and swollen joints, which would traditionally be used to assess disease severity in RA.271 Where comparisons exist, the information regarding, e.g. the presence of swollen joints and disease severity, are not well correlated. Objective measures of variables such as inflammatory cytokines are attractive, but for the patient, the severity, especially of pain, is subjective, and patient-assessed severity indices are consequently common. This said, the patient global assessment (PGA)272 is noteworthy273 as while using seven objective criteria based on stiffness and swelling, pain (being presumably too subjective) is not among them. Other studies did find some correlations between swelling, stiffness, and pain of joints.274,275

The European League Against Rheumatism (EULAR) has a number of useful recommendations, including treatment/management,276 efficacy,277,278 and joint imaging.279 For our present purposes, these will suffice entirely.

Small molecules (sDMARDs) and their role(s) as antibacterials

The chief recommendations276 are to start early and monitor frequently. These are seen as the strategy of first resort, with methrotrexate, sulfasalazine, and leflunomide being seen as front line drugs (possibly along with low-dose glucocorticoids). It is highly noteworthy that sulfasalazine is in fact an antibiotic (one of the first), as it is split in the intestine into aspirin and the antibiotic sulphapyridine,280,281 while methotrexate shows antibiotic activity against organisms as diverse as S. aureus282 and Plasmodium vivax.283 As mentioned above, “first treatments” are the least likely to be confounded by bias occasioned by the fact that later treatments will be favored by patients with more severe or refractory disease, a phenomenon that probably confounded a study of minocycline and doxycycline.284 However, and while – like other drugs285,286 – they probably have multiple effects, a good many studies indicate the utility of the antibiotics minocycline and doxycycline in treating RA (e.g. literature287294). After sDMARDs have been tried, the recommendation276is to move to a biological.

Biologicals (bDMARDs)

The chief biologicals276,277,295298 are inhibitors of TNF-α, including monoclonals, and inhibitors of the IL-6 receptor; they all decrease inflammatory symptoms, and it is unclear whether they might have any direct or indirect antibacterial effects. Their chief issue is that, as proteins, they can themselves cause (auto) antigens to be raised,299301 while, as mentioned above, any dampening of immune system response may increase the likelihood of novel or emergent infection.

Iron chelation as a therapeutic?

If the “dormant microbial” hypothesis is correct, it is to be predicted that nutritional and/or pharmacological iron chelators would be of clinical benefit. Although we are not aware of any clinical trials to this end, a number of papers indicate the therapeutic benefits of chelators in RA (e.g. literature302305 Iron chelation may also have a useful mechanistic role in iron redistribution.306308 Thus, as with Alzheimer’s disease, where the established benefits309 of iron chelation languished for decades,310,311 it would seem that iron withholding (e.g. literature312,313) might be an important therapeutic strategy in RA that is well worth exploring.

In a similar vein, we would predict or anticipate a great preponderance of RA in those with iron overload diseases, as indeed seems to be the case.314,315

A role for lipopolysacharides (LPS) in RA

Recently, we summarized the evidence for a significant involvement of lipopolysaccharide shed by dormant and resuscitating bacteria as underpinning the chronic inflammation characteristic of a variety of diseases, and suggested that LPS may play a role in the pathogenesis of RA.21 The presence and role of LPS may be supported by a recent review that provided evidence for the involvement of a microbiome in inflammatory arthritis and rheumatic diseases.316 Recently, Scher et al.317 also found strong correlates between the presence of Prevotella copri and new-onset untreated RA patients.

Certainly, LPS is also known to upregulate all of the cytokines upregulated in RA and mentioned in Table 1. In our recent review,21 we also focused on the fact that antibodies could be generated to LPS that – like the anti-Proteus antibodies mentioned in detail above – might also serve as autoantibodies of significance in RA and in particular during the flares (that may be ascribed to periods of particular resuscitation activity).

The generalized LPS also exerts its effects via activation of cytokines such as IL-6, and TNF-α in response to LPS,115 IL-8,318 IL-12,319 IL-15,320 thereby exciting the innate immune response. The scheme is typically as follows (extensively discussed in Kell and Pretorius21):

  • LPS binds to the toll-like receptor 4 (TLR4).321325

  • Production of a variety of pro-inflammatory cytokines,326328 where NF-κB plays a prominent role329,330 via a set of canonical pathways illustrated in Figure 7.

  • NF-κB translocates to the nucleus to turn on a great many genes in a frequency-dependent fashion, including in particular TNF-α and IL-6.331333

  • At high concentrations of LPS,334,335 it also activates a “non-canonical” inflammasome pathway, which is independent of TLR4336,337 (see Figure 8).

Figure 7.

Figure 7

The LPS-mediated cellular production of inflammatory cytokines. Canonical pathway of LPS-mediated release and nuclear translocation of NF-κB (based on O'Neill et al.327) taken from Kell and Pretorius.21 (A color version of this figure is available in the online journal.)

Figure 8.

Figure 8

The intracellular LPS-mediated activation of caspase-1 leading to IL-1β production (after Latz et al.338) taken from Kell and Pretorius.21 (A color version of this figure is available in the online journal.)

Finally (see above), LPS may catalyze the formation of inflammatory and cytotoxic β-amyloids. Consequently, we might again suggest that appropriate antibacterials and iron chelators (that can hitchhike on the necessary transporters282,339343) would serve to lower this stimulus, and in contrast to the biologics actually strike at the root causes of the disease.

Further tests of our hypothesis

While we have adduced much evidence in favor of the view that recurring infection by (resuscitating dormant) bacteria is in fact a, if not the, major and ultimate cause of RA, albeit there is not a unitary “cause,” our views do come with multiple predictions that may be tested (of course some have been already, see above, in many cases extensively).

  • Bacteria should be detectable in relevant tissues of RA patients, whether by culture or by molecular methods (e.g. macromolecular sequencing or antibodies).

  • Relevant products such as LPS and other antigens should be detectable in patients vs. controls.

  • Their numbers (bacteria and/or inflammatory products) should increase with disease severity and during “flares.”

  • Their numbers and activity (hence disease prevalence/severity) should correlate with free iron levels.

  • Treatments that lower the activity of bacteria and/or their products should be of significant therapeutic benefit.

  • These may include iron withholding, antibacterial, anti-LPS, and anti-amyloid treatments.

Although probably not yet seen as mainstream, a number of therapeutic strategies based on these and other ideas (including the roles of vitamin D metabolites, that for reasons of space we do not discuss here) do indeed seem to have enjoyed success (e.g. literature344348).

Concluding remarks

As mentioned previously,27 while it can be difficult (but cf349) to ascribe causality in complex biochemical networks, an accepted strategy within the philosophy of science, that rather accurately describes the systems biology approach, is to the effect that if a series of ostensibly unrelated findings are brought together into a self-consistent narrative, that narrative is thereby strengthened. This is known as “coherence.”350353 We have sought, we believe successfully, to bring together the evidence for a coherent narrative that links infection, microbial dormancy, iron dysregulation, and inflammation as part of the etiology of RA. Importantly, the proposals can easily be tested further, both diagnostically and therapeutically.

Acknowledgments

We thank the National Research Foundation (NRF) and Medical Research Council (MRC) of South Africa for supporting this collaboration. This article is paper 10 in the series “The dormant blood microbiome in chronic, inflammatory disease.”

Authors’ contributions

All authors participated in the design, interpretation of the studies, and analysis of the data and review of the manuscript. EP wrote the paper and prepared images; DBK wrote and edited the paper; OA prepared samples: PS identified patients, collected samples and verified the clinical relevance.

Declaration of Conflicting Interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Ethical approval

Ethical approval was granted at the University of Pretoria (Human Ethics Committee: Faculty of Health Sciences): E Pretorius.

Funding

This work was also a contribution from the Manchester Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) (BBSRC grant BB/M017702/1).

References

  • 1.Chemin K, Klareskog L, Malmstrom V. Is rheumatoid arthritis an autoimmune disease? Curr Opin Rheumatol 2016; 28: 181–8. [DOI] [PubMed] [Google Scholar]
  • 2.Carmona L, Cross M, Williams B, Lassere M, March L. Rheumatoid arthritis. Best Pract Res Clin Rheumatol 2010; 24: 733–45. [DOI] [PubMed] [Google Scholar]
  • 3.Terao C, Ikari K, Nakayamada S, Takahashi Y, Yamada R, Ohmura K, Hashimoto M, Furu M, Ito H, Fujii T, Yoshida S, Saito K, Taniguchi A, Momohara S, Yamanaka H, Mimori T, Matsuda F. A twin study of rheumatoid arthritis in the Japanese population. Mod Rheumatol 2016;26:685–9. [DOI] [PubMed]
  • 4.Mateen S, Zafar A, Moin S, Khan AQ, Zubair S. Understanding the role of cytokines in the pathogenesis of rheumatoid arthritis. Clin Chim Acta 2016;455:161–71. [DOI] [PubMed]
  • 5.Humphreys JH, Verstappen SM, Hyrich KL, Chipping JR, Marshall T, Symmons DP. The incidence of rheumatoid arthritis in the UK: comparisons using the 2010 ACR/EULAR classification criteria and the 1987 ACR classification criteria. Results from the Norfolk Arthritis Register. Ann Rheum Dis 2013; 72: 1315–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ebringer A. Rheumatoid arthritis and Proteus, London: Springer, 2012. [Google Scholar]
  • 7.Poehlmann KM. Rheumatoid arthritis: the infection connection, Torrance, CA: Satori Press, 2012. [Google Scholar]
  • 8.Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med 2015; 372: 793–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ebringer A, Rashid T. Rheumatoid arthritis is caused by Proteus: the molecular mimicry theory and Karl Popper. Front Biosci 2009; 1: 577–86. [DOI] [PubMed] [Google Scholar]
  • 10.Symmons DPM. Epidemiology research in rheumatology-progress and pitfalls. Nat Rev Rheumatol 2015; 11: 631–8. [DOI] [PubMed] [Google Scholar]
  • 11.Symmons D, Turner G, Webb R, Asten P, Barrett E, Lunt M, Scott D, Silman A. The prevalence of rheumatoid arthritis in the United Kingdom: new estimates for a new century. Rheumatology 2002; 41: 793–800. [DOI] [PubMed] [Google Scholar]
  • 12.Gibofsky A. Overview of epidemiology, pathophysiology, and diagnosis of rheumatoid arthritis. Am J Manag Care 2012; 18(13 Suppl): S295–302. [PubMed] [Google Scholar]
  • 13.Tobón GJ, Youinou P, Saraux A. The environment, geo-epidemiology, and autoimmune disease: rheumatoid arthritis. J Autoimmun 2010; 35: 10–4. [DOI] [PubMed] [Google Scholar]
  • 14.Charles J, Britt H, Pan Y. Rheumatoid arthritis. Aust Fam Physician 2013; 42: 765–765. [PubMed] [Google Scholar]
  • 15.Crane MM, Juneja M, Allen J, Kurrasch RH, Chu ME, Quattrocchi E, Manson SC, Chang DJ. Epidemiology and treatment of new-onset and established rheumatoid arthritis in an insured US population. Arthritis Care Res 2015; 67: 1646–55. [DOI] [PubMed] [Google Scholar]
  • 16.Silman AJ, Pearson JE. Epidemiology and genetics of rheumatoid arthritis. Arthritis Res 2002;4(Suppl 3):S265–72. [DOI] [PMC free article] [PubMed]
  • 17.McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med 2011; 365: 2205–19. [DOI] [PubMed] [Google Scholar]
  • 18.Aho K, Koskenvuo M, Tuominen J, Kaprio J. Occurrence of rheumatoid arthritis in a nationwide series of twins. J Rheumatol 1986; 13: 899–902. [PubMed] [Google Scholar]
  • 19.Silman AJ, MacGregor AJ, Thomson W, Holligan S, Carthy D, Farhan A, Ollier WE. Twin concordance rates for rheumatoid arthritis: results from a nationwide study. Br J Rheumatol 1993; 32: 903–7. [DOI] [PubMed] [Google Scholar]
  • 20.Svendsen AJ, Holm NV, Kyvik K, Petersen PH, Junker P. Relative importance of genetic effects in rheumatoid arthritis: historical cohort study of Danish nationwide twin population. BMJ 2002; 324: 264–6. [PMC free article] [PubMed] [Google Scholar]
  • 21.Kell DB, Pretorius E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: the central roles of LPS and LPS-induced cell death. Integr Biol 2015; 7: 1339–77. [DOI] [PubMed] [Google Scholar]
  • 22.Alten R, Pohl C, Choy EH, Christensen R, Furst DE, Hewlett SE, Leong A, May JE, Sanderson TC, Strand V, Woodworth TG, Bingham CO., 3rd Developing a construct to evaluate flares in rheumatoid arthritis: a conceptual report of the OMERACT RA flare definition working group. J Rheumatol 2011; 38: 1745–50. [DOI] [PubMed] [Google Scholar]
  • 23.Bingham CO, 3rd, Pohl C, Woodworth TG, Hewlett SE, May JE, Rahman MU, Witter JP, Furst DE, Strand CV, Boers M, Alten RE. Developing a standardized definition for disease “flare” in rheumatoid arthritis (OMERACT 9 Special Interest Group). J Rheumatol 2009; 36: 2335–41. [DOI] [PubMed] [Google Scholar]
  • 24.Kell DB. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics 2009; 2: 2–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kell DB, Pretorius E. The simultaneous occurrence of both hypercoagulability and hypofibrinolysis in blood and serum during systemic inflammation, and the roles of iron and fibrin(ogen). Integr Biol 2015; 7: 24–52. [DOI] [PubMed] [Google Scholar]
  • 26.Funke C, Schneider SA, Berg D, Kell DB. Genetics and iron in the systems biology of Parkinson's disease and some related disorders. Neurochem Int 2013; 62: 637–52. [DOI] [PubMed] [Google Scholar]
  • 27.Kell DB, Potgieter M, Pretorius E. Individuality, phenotypic differentiation, dormancy and ‘persistence’ in culturable bacterial systems: commonalities shared by environmental, laboratory, and clinical microbiology. F1000Research 2015; 6: 179–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Potgieter M, Bester J, Kell DB, Pretorius E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS Microbiol Rev 2015; 39: 567–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Foxman B. Epidemiology of urinary tract infections: incidence, morbidity, and economic costs. Am J Med 2002;113(Suppl 1A):5S–13. [DOI] [PubMed]
  • 30.Foxman B. The epidemiology of urinary tract infection. Nat Rev Urol 2010; 7: 653–60. [DOI] [PubMed] [Google Scholar]
  • 31.Vasudevan M. Urinary tract infection: an overview of the infection and the associated risk factors. J Microbiol Exp 2014; 1: 8–22. [Google Scholar]
  • 32.Ebringer A, Rashid T, Wilson C. Rheumatoid arthritis, Proteus, anti-CCP antibodies and Karl Popper. Autoimmun Rev 2010; 9: 216–23. [DOI] [PubMed] [Google Scholar]
  • 33.Ebringer A, Rashid T. Rheumatoid arthritis is caused by a Proteus urinary tract infection. Apmis 2014; 122: 363–8. [DOI] [PubMed] [Google Scholar]
  • 34.Medawar P. Pluto's republic, Oxford: Oxford University Press, 1982. [Google Scholar]
  • 35.Kell DB, Oliver SG. Here is the evidence, now what is the hypothesis? The complementary roles of inductive and hypothesis-driven science in the post-genomic era. Bioessays 2004; 26: 99–105. [DOI] [PubMed] [Google Scholar]
  • 36.Kell DB. Scientific discovery as a combinatorial optimisation problem: how best to navigate the landscape of possible experiments? Bioessays 2012; 34: 236–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mercado FB, Marshall RI, Klestov AC, Bartold PM. Relationship between rheumatoid arthritis and periodontitis. J Periodontol 2001; 72: 779–87. [DOI] [PubMed] [Google Scholar]
  • 38.Mercado F, Marshall RI, Klestov AC, Bartold PM. Is there a relationship between rheumatoid arthritis and periodontal disease? J Clin Periodontol 2000; 27: 267–72. [DOI] [PubMed] [Google Scholar]
  • 39.de Smit M, Westra J, Vissink A, Doornbos-van der Meer B, Brouwer E, van Winkelhoff AJ. Periodontitis in established rheumatoid arthritis patients: a cross-sectional clinical, microbiological and serological study. Arthritis Res Ther 2012; 14: R222–R222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Choi IA, Kim JH, Kim YM, Lee JY, Kim KH, Lee EY, Lee EB, Lee YM, Song YW. Periodontitis is associated with rheumatoid arthritis: a study with longstanding rheumatoid arthritis patients in Korea. Korean J Intern Med 2016; 31(5): 977–86.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fuggle NR, Smith TO, Kaul A, Sofat N. Hand to mouth: a systematic review and meta-analysis of the association between rheumatoid arthritis and periodontitis. Front Immunol 2016; 7: 80–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Scher JU, Ubeda C, Equinda M, Khanin R, Buischi Y, Viale A, Lipuma L, Attur M, Pillinger MH, Weissmann G, Littman DR, Pamer EG, Bretz WA, Abramson SB. Periodontal disease and the oral microbiota in new-onset rheumatoid arthritis. Arthritis Rheum 2012; 64: 3083–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bialowas K, Swierkot J, Radwan-Oczko M. [Role of Porphyromonas gingivalis in rheumatoid arthritis and inflammatory spondyloarthropathies]. Postepy Hig Med Dosw 2014; 68: 1171–9. [DOI] [PubMed] [Google Scholar]
  • 44.Maeda Y, Kumanogoh A, Takeda K. Altered composition of gut microbiota in rheumatoid arthritis patients. Nihon Rinsho Meneki Gakkai Kaishi 2016; 39: 59–63. [DOI] [PubMed] [Google Scholar]
  • 45.Zhang X, Zhang D, Jia H. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med 2015;21:895–905. [DOI] [PubMed]
  • 46.Chen J, Wright K, Davis JM, Jeraldo P, Marietta EV, Murray J, Nelson H, Matteson EL, Taneja V. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med 2016; 8: 43–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Leirisalo-Repo M. Early arthritis and infection. Curr Opin Rheumatol 2005; 17: 433–9. [DOI] [PubMed] [Google Scholar]
  • 48.Carter JD, Gerard HC, Whittum-Hudson JA, Hudson AP. The molecular basis for disease phenotype in chronic Chlamydia-induced arthritis. Int J Clin Rheumtol 2012; 7: 627–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Newkirk MM, Goldbach-Mansky R, Senior BW, Klippel J, Schumacher HR, Jr., El-Gabalawy HS. Elevated levels of IgM and IgA antibodies to Proteus mirabilis and IgM antibodies to Escherichia coli are associated with early rheumatoid factor (RF)-positive rheumatoid arthritis. Rheumatology 2005; 44: 1433–41. [DOI] [PubMed] [Google Scholar]
  • 50.Newkirk MM, Zbar A, Baron M, Manges AR. Distinct bacterial colonization patterns of Escherichia coli subtypes associate with rheumatoid factor status in early inflammatory arthritis. Rheumatology 2010; 49: 1311–6. [DOI] [PubMed] [Google Scholar]
  • 51.Hill Gaston JS, Lillicrap MS. Arthritis associated with enteric infection. Best Pract Res Clin Rheumatol 2003; 17: 219–39. [DOI] [PubMed] [Google Scholar]
  • 52.Astrauskiene D, Bernotiene E. New insights into bacterial persistence in reactive arthritis. Clin Exp Rheumatol 2007; 25: 470–9. [PubMed] [Google Scholar]
  • 53.Amital H, Govoni M, Maya R, Meroni PL, Ori B, Shoenfeld Y, Tincani A, Trotta F, Sarzi-Puttini P, Atzeni F. Role of infectious agents in systemic rheumatic diseases. Clin Exp Rheumatol 2008; 26(1 Suppl 48): S27–32. [PubMed] [Google Scholar]
  • 54.Schaeverbeke T, Renaudin H, Clerc M, Lequen L, Vernhes JP, De Barbeyrac B, Bannwarth B, Bebear C, Dehais J. Systematic detection of mycoplasmas by culture and polymerase chain reaction (PCR) procedures in 209 synovial fluid samples. Br J Rheumatol 1997; 36: 310–4. [DOI] [PubMed] [Google Scholar]
  • 55.Sawitzke A, Joyner D, Knudtson K, Mu HH, Cole B. Anti-MAM antibodies in rheumatic disease: evidence for a MAM-like superantigen in rheumatoid arthritis? J Rheumatol 2000; 27: 358–64. [PubMed] [Google Scholar]
  • 56.da Rocha Sobrinho HM, Jarach R, da Silva NA, Shio MT, Jancar S, Timenetsky J, Oliveira MAP, Dorta ML, Ribeiro-Dias F. Mycoplasmal lipid-associated membrane proteins and Mycoplasma arthritidis mitogen recognition by serum antibodies from patients with rheumatoid arthritis. Rheumatol Int 2011; 31: 951–7. [DOI] [PubMed] [Google Scholar]
  • 57.Haier J, Nasralla M, Franco AR, Nicolson GL. Detection of mycoplasmal infections in blood of patients with rheumatoid arthritis. Rheumatology 1999; 38: 504–9. [DOI] [PubMed] [Google Scholar]
  • 58.Martinez-Martinez RE, Abud-Mendoza C, Patiño-Marin N, Rizo-Rodríguez JC, Little JW, Loyola-Rodríguez JP. Detection of periodontal bacterial DNA in serum and synovial fluid in refractory rheumatoid arthritis patients. J Clin Periodontol 2009; 36: 1004–10. [DOI] [PubMed] [Google Scholar]
  • 59.Mikuls TR, Payne JB, Reinhardt RA, Thiele GM, Maziarz E, Cannella AC, Holers VM, Kuhn KA, O'Dell JR. Antibody responses to Porphyromonas gingivalis (P. gingivalis) in subjects with rheumatoid arthritis and periodontitis. Int Immunopharmacol 2009; 9: 38–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hitchon CA, Chandad F, Ferucci ED, Willemze A, Ioan-Facsinay A, van der Woude D, Markland J, Robinson D, Elias B, Newkirk M, Toes RM, Huizinga TW, El-Gabalawy HS. Antibodies to Porphyromonas gingivalis are associated with anticitrullinated protein antibodies in patients with rheumatoid arthritis and their relatives. J Rheumatol 2010; 37: 1105–12. [DOI] [PubMed] [Google Scholar]
  • 61.Mikuls TR, Thiele GM, Deane KD, Payne JB, O'Dell JR, Yu F, Sayles H, Weisman MH, Gregersen PK, Buckner JH, Keating RM, Derber LA, Robinson WH, Holers VM, Norris JM. Porphyromonas gingivalis and disease-related autoantibodies in individuals at increased risk of rheumatoid arthritis. Arthritis Rheum 2012; 64: 3522–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Li S, Yu Y, Yue Y, Liao H, Xie W, Thai J, Mikuls TR, Thiele GM, Duryee MJ, Sayles H, Payne JB, Klassen LW, O'Dell JR, Zhang Z, Su K. autoantibodies from single circulating plasmablasts react with citrullinated antigens and Porphyromonas gingivalis in rheumatoid arthritis. Arthritis Rheumatol 2016; 68: 614–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kharlamova N, Jiang X, Sherina N, Potempa B, Israelsson L, Quirke AM, Eriksson K, Yucel-Lindberg T, Venables PJ, Potempa J, Alfredsson L, Lundberg K. Antibodies to Porphyromonas gingivalis indicate interaction between oral infection, smoking, and risk genes in rheumatoid arthritis etiology. Arthritis Rheumatol 2016; 68: 604–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Schrama JC, Lutro O, Langvatn H, Hallan G, Espehaug B, Sjursen H, Engesaeter LB, Fevang BT. Bacterial findings in infected hip joint replacements in patients with rheumatoid arthritis and osteoarthritis: a study of 318 revisions for infection reported to the Norwegian arthroplasty register. ISRN Orthop 2012; 2012: 437675–437675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kaprelyants AS, Kell DB. Dormancy in stationary-phase cultures of Micrococcus luteus: flow cytometric analysis of starvation and resuscitation. Appl Env Microbiol 1993; 59: 3187–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kell DB, Kaprelyants AS, Weichart DH, Harwood CL, Barer MR. Viability and activity in readily culturable bacteria: a review and discussion of the practical issues. Antonie van Leeuwenhoek 1998; 73: 169–87. [DOI] [PubMed] [Google Scholar]
  • 67.Kell DB, Young M. Bacterial dormancy and culturability: the role of autocrine growth factors. Curr Opin Microbiol 2000; 3: 238–43. [DOI] [PubMed] [Google Scholar]
  • 68.Kaprelyants AS, Kell DB. Rapid assessment of bacterial viability and vitality using rhodamine 123 and flow cytometry. J Appl Bacteriol 1992; 72: 410–22. [Google Scholar]
  • 69.Kaprelyants AS, Mukamolova GV, Kell DB. Estimation of dormant Micrococcus luteus cells by penicillin lysis and by resuscitation in cell-free spent medium at high dilution. FEMS Microbiol Lett 1994; 115: 347–52. [Google Scholar]
  • 70.Kaprelyants AS, Mukamolova GV, Davey HM, Kell DB. Quantitative analysis of the physiological heterogeneity within starved cultures of Micrococcus luteus using flow cytometry and cell sorting. Appl Env Microbiol 1996; 62: 1311–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Mukamolova GV, Kaprelyants AS, Kell DB. Secretion of an antibacterial factor during resuscitation of dormant cells in Micrococcus luteus cultures held in an extended stationary phase. Antonie van Leeuwenhoek 1995; 67: 289–95. [DOI] [PubMed] [Google Scholar]
  • 72.Mukamolova GV, Yanopolskaya ND, Kell DB, Kaprelyants AS. On resuscitation from the dormant state of Micrococcus luteus. Antonie van Leeuwenhoek 1998; 73: 237–43. [DOI] [PubMed] [Google Scholar]
  • 73.Mukamolova GV, Kaprelyants AS, Kell DB, Young M. Adoption of the transiently non-culturable state – a bacterial survival strategy? Adv Micr Physiol 2003; 47: 65–129. [DOI] [PubMed] [Google Scholar]
  • 74.Mukamolova GV, Kaprelyants AS, Young DI, Young M, Kell DB. A bacterial cytokine. Proc Natl Acad Sci 1998; 95: 8916–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mukamolova GV, Kormer SS, Kell DB, Kaprelyants AS. Stimulation of the multiplication of Micrococcus luteus by an autocrine growth factor. Arch Microbiol 1999; 172: 9–14. [DOI] [PubMed] [Google Scholar]
  • 76.Mukamolova GV, Turapov OA, Young DI, Kaprelyants AS, Kell DB, Young M. A family of autocrine growth factors in Mycobacterium tuberculosis. Mol Microbiol 2002; 46: 623–35. [DOI] [PubMed] [Google Scholar]
  • 77.Mukamolova GV, Turapov OA, Kazarian K, Telkov M, Kaprelyants AS, Kell DB, Young M. The rpf gene of Micrococcus luteus encodes an essential secreted growth factor. Mol Microbiol 2002; 46: 611–21. [DOI] [PubMed] [Google Scholar]
  • 78.Mukamolova GV, Murzin AG, Salina EG, Demina GR, Kell DB, Kaprelyants AS, Young M. Muralytic activity of Micrococcus luteus Rpf and its relationship to physiological activity in promoting bacterial growth and resuscitation. Mol Microbiol 2006; 59: 84–98. [DOI] [PubMed] [Google Scholar]
  • 79.Allison KR, Brynildsen MP, Collins JJ. Heterogeneous bacterial persisters and engineering approaches to eliminate them. Curr Opin Microbiol 2011; 14: 593–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Fauvart M, De Groote VN, Michiels J. Role of persister cells in chronic infections: clinical relevance and perspectives on anti-persister therapies. J Med Microbiol 2011; 60(Pt 6): 699–709. [DOI] [PubMed] [Google Scholar]
  • 81.Kester JC, Fortune SM. Persisters and beyond: mechanisms of phenotypic drug resistance and drug tolerance in bacteria. Crit Rev Biochem Mol Biol 2014; 49: 91–101. [DOI] [PubMed] [Google Scholar]
  • 82.Lewis K. Persister cells, dormancy and infectious disease. Nat Rev Microbiol 2007; 5: 48–56. [DOI] [PubMed] [Google Scholar]
  • 83.Lewis K. Persister cells. Annu Rev Microbiol 2010; 64: 357–72. [DOI] [PubMed] [Google Scholar]
  • 84.Wood TK, Knabel SJ, Kwan BW. Bacterial persister cell formation and dormancy. Appl Environ Microbiol 2013; 79: 7116–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zhang Y, Yew WW, Barer MR. Targeting persisters for tuberculosis control. Antimicrob Agents Chemother 2012; 56(5): 2223–30.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Holden DW. Persisters unmasked. Science 2015; 347: 30–2. [DOI] [PubMed] [Google Scholar]
  • 87.Damgaard C, Magnussen K, Enevold C, Nilsson M, Tolker-Nielsen T, Holmstrup P, Nielsen CH. Viable bacteria associated with red blood cells and plasma in freshly drawn blood donations. PLoS One 2015; 10: e0120826.–e0120826.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Domingue GJ, Ghoniem GM, Bost KL, Fermin C, Human LG. Dormant microbes in interstitial cystitis. J Urol 1995; 153: 1321–6. [PubMed] [Google Scholar]
  • 89.Domingue GJ, Woody HB. Bacterial persistence and expression of disease. Clin Microbiol Rev 1997; 10: 320–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Domingue GJ., Sr Cryptic bacterial infection in chronic prostatitis: diagnostic and therapeutic implications. Curr Opin Urol 1998; 8: 45–9. [DOI] [PubMed] [Google Scholar]
  • 91.Mattman L. Cell wall deficient forms: stealth pathogens, 3rd ed Boca Raton: CRC Press, 2001. [Google Scholar]
  • 92.Thwaites GE, Gant V. Are bloodstream leukocytes Trojan Horses for the metastasis of Staphylococcus aureus? Nat Rev Microbiol 2011; 9: 215–22. [DOI] [PubMed] [Google Scholar]
  • 93.Curtis JR, Patkar N, Xie A, Martin C, Allison JJ, Saag M, Shatin D, Saag KG. Risk of serious bacterial infections among rheumatoid arthritis patients exposed to tumor necrosis factor alpha antagonists. Arthritis Rheum 2007; 56: 1125–33. [DOI] [PubMed] [Google Scholar]
  • 94.Bernatsky S, Habel Y, Rahme E. Observational studies of infections in rheumatoid arthritis: a metaanalysis of tumor necrosis factor antagonists. J Rheumatol 2010; 37: 928–31. [DOI] [PubMed] [Google Scholar]
  • 95.Lanternier F, Tubach F, Ravaud P, Salmon D, Dellamonica P, Bretagne S, Couret M, Bouvard B, Debandt M, Gueit I, Gendre JP, Leone J, Nicolas N, Che D, Mariette X, Lortholary O. Research Axed on Tolerance of Biotherapies Group. Incidence and risk factors of Legionella pneumophila pneumonia during anti-tumor necrosis factor therapy: a prospective French study. Chest 2013; 144: 990–8. [DOI] [PubMed] [Google Scholar]
  • 96.Tubach F, Salmon D, Ravaud P, Allanore Y, Goupille P, Breban M, Pallot-Prades B, Pouplin S, Sacchi A, Chichemanian RM, Bretagne S, Emilie D, Lemann M, Lortholary O, Mariette X. Research Axed on Tolerance of Biotherapies Group. Risk of tuberculosis is higher with anti-tumor necrosis factor monoclonal antibody therapy than with soluble tumor necrosis factor receptor therapy: the three-year prospective French Research Axed on Tolerance of Biotherapies registry. Arthritis Rheum 2009; 60: 1884–94.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Freestone PP, Haigh RD, Williams PH, Lyte M. Stimulation of bacterial growth by heat-stable, norepinephrine-induced autoinducers. FEMS Microbiol Lett 1999; 172: 53–60. [DOI] [PubMed] [Google Scholar]
  • 98.Freestone PPE, Sandrini SM, Haigh RD, Lyte M. Microbial endocrinology: how stress influences susceptibility to infection. Trends Microbiol 2008; 16: 55–64. [DOI] [PubMed] [Google Scholar]
  • 99.Lyte M, Ernst S. Catecholamine-induced growth of Gram-negative bacteria. Life Sci 1992; 50: 203–12. [DOI] [PubMed] [Google Scholar]
  • 100.Lyte M, Ernst S. Alpha-adrenergic and beta-adrenergic-receptor involvement in catecholamine-induced growth of gram-negative bacteria. Biochem Biophys Res Commun 1993; 190: 447–52. [DOI] [PubMed] [Google Scholar]
  • 101.Lyte M. Microbial endocrinology and infectious disease in the 21st century. Trends Microbiol 2004; 12: 14–20. [DOI] [PubMed] [Google Scholar]
  • 102.Lyte M. Microbial endocrinology: host-microbiota neuroendocrine interactions influencing brain and behavior. Gut Microbes 2014; 5: 381–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Reissbrodt R, Rienaecker I, Romanova JM, Freestone PPE, Haigh RD, Lyte M, Tschäpe H, Williams PH. Resuscitation of Salmonella enterica serovar typhimurium and enterohemorrhagic Escherichia coli from the viable but nonculturable state by heat-stable enterobacterial autoinducer. Appl Environ Microbiol 2002; 68: 4788–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Freestone PPE, Haigh RD, Williams PH, Lyte M. Involvement of enterobactin in norepinephrine-mediated iron supply from transferrin to enterohaemorrhagic Escherichia coli. FEMS Microbiol Lett 2003; 222: 39–43. [DOI] [PubMed] [Google Scholar]
  • 105.Sandrini SM, Shergill R, Woodward J, Muralikuttan R, Haigh RD, Lyte M, Freestone PP. Elucidation of the mechanism by which catecholamine stress hormones liberate iron from the innate immune defense proteins transferrin and lactoferrin. J Bacteriol 2010; 192: 587–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Armitage AE, Drakesmith H. The battle for iron. Science 2014; 346: 1299–300. [DOI] [PubMed] [Google Scholar]
  • 107.Fischbach MA, Lin HN, Liu DR, Walsh CT. How pathogenic bacteria evade mammalian sabotage in the battle for iron. Nat Chem Biol 2006; 2: 132–8. [DOI] [PubMed] [Google Scholar]
  • 108.Haley KP, Skaar EP. A battle for iron: host sequestration and Staphylococcus aureus acquisition. Microbes Infect 2012; 14: 217–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Subashchandrabose S, Mobley HLT. Back to the metal age: battle for metals at the host-pathogen interface during urinary tract infection. Metallomics 2015; 7: 935–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Nairz M, Schroll A, Sonnweber T, Weiss G. The struggle for iron – a metal at the host-pathogen interface. Cell Microbiol 2010; 12: 1691–702. [DOI] [PubMed] [Google Scholar]
  • 111.Reid DW, Anderson GJ, Lamont IL. Role of lung iron in determining the bacterial and host struggle in cystic fibrosis. Am J Physiol Lung Cell Mol Physiol 2009; 297: L795–802. [DOI] [PubMed] [Google Scholar]
  • 112.Kell DB. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson’s, Huntington’s, Alzheimer’s, prions, bactericides, chemical toxicology and others as examples. Arch Toxicol 2010; 577: 825–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Roberts CA, Dickinson AK, Taams LS. The interplay between monocytes/macrophages and CD4(+) T cell subsets in rheumatoid arthritis. Front Immunol 2015; 6: 571–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Furst DE, Emery P. Rheumatoid arthritis pathophysiology: update on emerging cytokine and cytokine-associated cell targets. Rheumatology 2014; 53: 1560–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ruscitti P, Cipriani P, Carubbi F, Liakouli V, Zazzeroni F. The role of IL-1beta in the bone loss during rheumatic diseases. Mediators Inflamm 2015;2015:782382. [DOI] [PMC free article] [PubMed]
  • 116.Lee B, Kim TH, Jun JB, Yoo DH, Woo JH, Choi SJ, Lee YH, Song GG, Sohn J, Park-Min KH, Ivashkiv LB, Ji JD. Direct inhibition of human RANK+ osteoclast precursors identifies a homeostatic function of IL-1beta. J Immunol 2010; 185: 5926–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Joosten LA. Excessive interleukin-1 signaling determines the development of Th1 and Th17 responses in chronic inflammation. Arthritis Rheum 2010; 62: 320–2. [DOI] [PubMed] [Google Scholar]
  • 118.Eklund KK, Leirisalo-Repo M, Ranta P, Maki T, Kautiainen H, Hannonen P, Korpela M, Hakala M, Jarvinen P, Mottonen T. Serum IL-1beta levels are associated with the presence of erosions in recent onset rheumatoid arthritis. Clin Exp Rheumatol 2007; 25: 684–9. [PubMed] [Google Scholar]
  • 119.McInnes IB, Buckley CD, Isaacs JD. Cytokines in rheumatoid arthritis – shaping the immunological landscape. Nat Rev Rheumatol 2016; 12: 63–8. [DOI] [PubMed] [Google Scholar]
  • 120.Rossi D, Modena V, Sciascia S, Roccatello D. Rheumatoid arthritis: biological therapy other than anti-TNF. Int Immunopharmacol 2015; 27: 185–8. [DOI] [PubMed] [Google Scholar]
  • 121.Huizinga TW. Personalized medicine in rheumatoid arthritis: is the glass half full or half empty? J Intern Med 2015; 277: 178–87. [DOI] [PubMed] [Google Scholar]
  • 122.Kim GW, Lee NR, Pi RH, Lim YS, Lee YM, Lee JM, Jeong HS, Chung SH. IL-6 inhibitors for treatment of rheumatoid arthritis: past, present, and future. Arch Pharm Res 2015; 38: 575–84. [DOI] [PubMed] [Google Scholar]
  • 123.Siebert S, Tsoukas A, Robertson J, McInnes I. Cytokines as therapeutic targets in rheumatoid arthritis and other inflammatory diseases. Pharmacol Rev 2015; 67: 280–309. [DOI] [PubMed] [Google Scholar]
  • 124.Puchner A, Bluml S. IL-6 blockade in chronic inflammatory diseases. Wien Med Wochenschr 2015; 165: 14–22. [DOI] [PubMed] [Google Scholar]
  • 125.Calabrese LH, Rose-John S. IL-6 biology: implications for clinical targeting in rheumatic disease. Nat Rev Rheumatol 2014; 10: 720–7. [DOI] [PubMed] [Google Scholar]
  • 126.Li F, Xu J, Zheng J, Sokolove J, Zhu K, Zhang Y, Sun H, Evangelou E, Pan Z. Association between interleukin-6 gene polymorphisms and rheumatoid arthritis in Chinese Han population: a case-control study and a meta-analysis. Sci Rep 2014; 4: 5714–5714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Srirangan S, Choy EH. The role of interleukin 6 in the pathophysiology of rheumatoid arthritis. Ther Adv Musculoskelet Dis 2010; 2: 247–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Madhok R, Crilly A, Watson J, Capell HA. Serum interleukin 6 levels in rheumatoid arthritis: correlations with clinical and laboratory indices of disease activity. Ann Rheum Dis 1993; 52: 232–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Cho ML, Ju JH, Kim HR, Oh HJ, Kang CM, Jhun JY, Lee SY, Park MK, Min JK, Park SH, Lee SH, Kim HY. Toll-like receptor 2 ligand mediates the upregulation of angiogenic factor, vascular endothelial growth factor and interleukin-8/CXCL8 in human rheumatoid synovial fibroblasts. Immunol Lett 2007; 108: 121–8. [DOI] [PubMed] [Google Scholar]
  • 130.Caiello I, Minnone G, Holzinger D, Vogl T, Prencipe G, Manzo A, De Benedetti F, Strippoli R. IL-6 amplifies TLR mediated cytokine and chemokine production: implications for the pathogenesis of rheumatic inflammatory diseases. PLoS One 2014; 9: e107886–e107886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Laveti D, Kumar M, Hemalatha R, Sistla R, Naidu VG, Talla V, Verma V, Kaur N, Nagpal R. Anti-inflammatory treatments for chronic diseases: a review. Inflamm Allergy Drug Targets 2013; 12: 349–61. [DOI] [PubMed] [Google Scholar]
  • 132.Paradowska A, Maślińiski W, Grzybowska-Kowalczyk A, Łacki J. The function of interleukin 17 in the pathogenesis of rheumatoid arthritis. Arch Immunol Ther Exp 2007; 55: 329–34. [DOI] [PubMed] [Google Scholar]
  • 133.Williams RO. Pathogenesis and therapy of rheumatoid arthritis. Ernst Schering Found Symp Proc 2006;1:107–30. [DOI] [PubMed]
  • 134.Troughton PR, Platt R, Bird H, el-Manzalawi E, Bassiouni M, Wright V. Synovial fluid interleukin-8 and neutrophil function in rheumatoid arthritis and seronegative polyarthritis. Br J Rheumatol 1996; 35: 1244–51. [DOI] [PubMed] [Google Scholar]
  • 135.Kaneko S, Satoh T, Chiba J, Ju C, Inoue K, Kagawa J. Interleukin-6 and interleukin-8 levels in serum and synovial fluid of patients with osteoarthritis. Cytokines Cell Mol Ther 2000; 6: 71–9. [DOI] [PubMed] [Google Scholar]
  • 136.Shen L, Zhang H, Zhou X, Liu R. Association between polymorphisms of interleukin 12 and rheumatoid arthritis associated biomarkers in a Chinese population. Cytokine 2015; 76: 363–7. [DOI] [PubMed] [Google Scholar]
  • 137.Angelo HD, Gomes S, II, Oliveira RD, Louzada-Junior P, Donadi EA, Crovella S, Maia MM, de Souza PR, Sandrin-Garcia P. Interleukin-18, interleukin-12B and interferon-gamma gene polymorphisms in Brazilian patients with rheumatoid arthritis: a pilot study. Tissue Antigens 2015; 86: 276–8. [DOI] [PubMed] [Google Scholar]
  • 138.Nakano S, Morimoto S, Suzuki S, Tsushima H, Yamanaka K, Sekigawa I, Takasaki Y. Immunoregulatory role of IL-35 in T cells of patients with rheumatoid arthritis. Rheumatology 2015; 54: 1498–506. [DOI] [PubMed] [Google Scholar]
  • 139.Pope RM, Shahrara S. Possible roles of IL-12-family cytokines in rheumatoid arthritis. Nat Rev Rheumatol 2013; 9: 252–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ernestam S, af Klint E, Catrina AI, Sundberg E, Engstrom M, Klareskog L, Ulfgren AK. Synovial expression of IL-15 in rheumatoid arthritis is not influenced by blockade of tumour necrosis factor. Arthritis Res Ther 2006; 8: R18–R18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ebrahimi AA, Noshad H, Sadreddini S, Hejazi MS, Mohammadzadeh Sadigh Y, Eshraghi Y, Ghojazadeh M. Serum levels of TNF-alpha, TNF-alphaRI, TNF-alphaRII and IL-12 in treated rheumatoid arthritis patients. Iran J Immunol 2009; 6: 147–53. [PubMed] [Google Scholar]
  • 142.Gonzalez-Alvaro I, Ortiz AM, Garcia-Vicuna R, Balsa A, Pascual-Salcedo D, Laffon A. Increased serum levels of interleukin-15 in rheumatoid arthritis with long-term disease. Clin Exp Rheumatol 2003; 21: 639–42. [PubMed] [Google Scholar]
  • 143.Jung YO, Cho ML, Kang CM, Jhun JY, Park JS, Oh HJ, Min JK, Park SH, Kim HY. Toll-like receptor 2 and 4 combination engagement upregulate IL-15 synergistically in human rheumatoid synovial fibroblasts. Immunol Lett 2007; 109: 21–7. [DOI] [PubMed] [Google Scholar]
  • 144.Kageyama Y, Takahashi M, Torikai E, Suzuki M, Ichikawa T, Nagafusa T, Koide Y, Nagano A. Treatment with anti-TNF-alpha antibody infliximab reduces serum IL-15 levels in patients with rheumatoid arthritis. Clin Rheumatol 2007; 26: 505–9. [DOI] [PubMed] [Google Scholar]
  • 145.Yang XK, Xu WD, Leng RX, Liang Y, Liu YY, Fang XY, Feng CC, Li R, Cen H, Pan HF, Ye DQ. Therapeutic potential of IL-15 in rheumatoid arthritis. Hum Immunol 2015; 76: 812–8. [DOI] [PubMed] [Google Scholar]
  • 146.Waldmann TA. Targeting the interleukin-15 system in rheumatoid arthritis. Arthritis Rheum 2005; 52: 2585–8. [DOI] [PubMed] [Google Scholar]
  • 147.Cho ML, Jung YO, Kim KW, Park MK, Oh HJ, Ju JH, Cho YG, Min JK, Kim SI, Park SH, Kim HY. IL-17 induces the production of IL-16 in rheumatoid arthritis. Exp Mol Med 2008; 40: 237–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Murota A, Suzuki K, Kassai Y, Miyazaki T, Morita R, Kondo Y, Takeshita M, Niki Y, Yoshimura A, Takeuchi T. Serum proteomic analysis identifies interleukin 16 as a biomarker for clinical response during early treatment of rheumatoid arthritis. Cytokine 2016; 78: 87–93. [DOI] [PubMed] [Google Scholar]
  • 149.Warstat K, Hoberg M, Rudert M, Tsui S, Pap T, Angres B, Essl M, Smith TJ, Cruikshank WW, Klein G, Gay S, Aicher WK. Transforming growth factor beta1 and laminin-111 cooperate in the induction of interleukin-16 expression in synovial fibroblasts from patients with rheumatoid arthritis. Ann Rheum Dis 2010; 69: 270–5. [DOI] [PubMed] [Google Scholar]
  • 150.Lard LR, Roep BO, Toes RE, Huizinga TW. Enhanced concentrations of interleukin 16 are associated with joint destruction in patients with rheumatoid arthritis. J Rheumatol 2004; 31(1): 35–9.. [PubMed] [Google Scholar]
  • 151.Kaufmann J, Franke S, Kientsch-Engel R, Oelzner P, Hein G, Stein G. Correlation of circulating interleukin 16 with proinflammatory cytokines in patients with rheumatoid arthritis. Rheumatology 2001; 40: 474–5. [DOI] [PubMed] [Google Scholar]
  • 152.Lubberts E, Koenders MI, van den Berg WB. The role of T-cell interleukin-17 in conducting destructive arthritis: lessons from animal models. Arthritis Res Ther 2005; 7: 29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Schminke B, Trautmann S, Mai B, Miosge N, Blaschke S. Interleukin 17 inhibits progenitor cells in rheumatoid arthritis cartilage. Eur J Immunol 2016; 46: 440–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Shabgah AG, Fattahi E, Shahneh FZ. Interleukin-17 in human inflammatory diseases. Postepy Dermatol Alergol 2014; 31: 256–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Baek SH, Lee SG, Park YE, Kim GT, Kim CD, Park SY. Increased synovial expression of IL-27 by IL-17 in rheumatoid arthritis. Inflamm Res 2012; 61: 1339–45. [DOI] [PubMed] [Google Scholar]
  • 156.Hwang SY, Kim JY, Kim KW, Park MK, Moon Y, Kim WU, Kim HY. IL-17 induces production of IL-6 and IL-8 in rheumatoid arthritis synovial fibroblasts via NF-kappaB- and PI3-kinase/Akt-dependent pathways. Arthritis Res Ther 2004; 6: R120–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Paradowska-Gorycka A, Grzybowska-Kowalczyk A, Wojtecka-Lukasik E, Maslinski S. IL-23 in the pathogenesis of rheumatoid arthritis. Scand J Immunol 2010; 71: 134–45. [DOI] [PubMed] [Google Scholar]
  • 158.Roşu A, Margaritescu C, Stepan A, Musetescu A, Ene M. IL-17 patterns in synovium, serum and synovial fluid from treatment-naive, early rheumatoid arthritis patients. Rom J Morphol Embryol 2012; 53: 73–80. [PubMed] [Google Scholar]
  • 159.Gracie JA, Forsey RJ, Chan WL, Gilmour A, Leung BP, Greer MR, Kennedy K, Carter R, Wei XQ, Xu D, Field M, Foulis A, Liew FY, McInnes IB. A proinflammatory role for IL-18 in rheumatoid arthritis. J Clin Invest 1999; 104: 1393–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Yang Z, Cao J, Yu C, Yang Q, Zhang Y, Han L. Caspase-1 mediated interleukin-18 activation in neutrophils promotes the activity of rheumatoid arthritis in a NLRP3 inflammasome independent manner. Joint Bone Spine 2016;83:282–9. [DOI] [PubMed]
  • 161.Zhang QG, Qian J, Zhu YC. Targeting bromodomain-containing protein 4 (BRD4) benefits rheumatoid arthritis. Immunol Lett 2015; 166: 103–8. [DOI] [PubMed] [Google Scholar]
  • 162.Cai LP, Zhou LJ, Lu SY, Liang YE, Chen XY, Liu L, Lin J. Association of IL-18 promoter gene polymorphisms with rheumatoid arthritis: a meta-analysis. Mol Biol Rep 2014; 41: 8211–7. [DOI] [PubMed] [Google Scholar]
  • 163.Gurram VC, Polipalli SK, Karra VK, Puppala M, Pandey SK, Kapoor S, Kumar A, Godi S. Genetic polymorphism of interleukin-18 gene promoter region in rheumatoid arthritis patients from southern India. J Clin Diagn Res 2014; 8: Sc01–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Wen D, Liu J, Du X, Dong JZ, Ma CS. Association of interleukin-18 (-137G/C) polymorphism with rheumatoid arthritis and systemic lupus erythematosus: a meta-analysis. Int Rev Immunol 2014; 33: 34–44. [DOI] [PubMed] [Google Scholar]
  • 165.Bresnihan B, Roux-Lombard P, Murphy E, Kane D, FitzGerald O, Dayer JM. Serum interleukin 18 and interleukin 18 binding protein in rheumatoid arthritis. Ann Rheum Dis 2002; 61: 726–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Zaky DS, El-Nahrery EM. Role of interleukin-23 as a biomarker in rheumatoid arthritis patients and its correlation with disease activity. Int Immunopharmacol 2016; 31: 105–8. [DOI] [PubMed] [Google Scholar]
  • 167.Patel DD, Kuchroo VK. Th17 Cell pathway in human immunity: lessons from genetics and therapeutic interventions. Immunity 2015; 43: 1040–51. [DOI] [PubMed] [Google Scholar]
  • 168.Abu Al Fadl EM, Fattouh M, Allam AA. High IL-23 level is a marker of disease activity in rheumatoid arthritis. Egypt J Immunol 2013; 20: 85–92. [PubMed] [Google Scholar]
  • 169.Meka RR, Venkatesha SH, Dudics S, Acharya B, Moudgil KD. IL-27-induced modulation of autoimmunity and its therapeutic potential. Autoimmun Rev 2015; 14: 1131–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Wynick C, Petes C, Gee K. Interleukin-27 mediates inflammation during chronic disease. J Interferon Cytokine Res 2014; 34: 741–9. [DOI] [PubMed] [Google Scholar]
  • 171.Yan J, Shang X, Rong X. Relationship between IL-27 gene polymorphism and susceptibility of rheumatoid arthritis in Chinese Han population. Int J Clin Exp Med 2015; 8: 6262–6. [PMC free article] [PubMed] [Google Scholar]
  • 172.Paradowska-Gorycka A, Raszkiewicz B, Jurkowska M, Felis-Giemza A, Romanowska-Prochnicka K, Manczak M, Olesinska M. Association of single nucleotide polymorphisms in the IL27 gene with rheumatoid arthritis. Scand J Immunol 2014; 80: 298–305. [DOI] [PubMed] [Google Scholar]
  • 173.Gong F, Pan YH, Huang X, Chen J, Xiao JH, Zhu HY. Interleukin-27 as a potential therapeutic target for rheumatoid arthritis: has the time come? Clin Rheumatol 2013; 32: 1425–8. [DOI] [PubMed] [Google Scholar]
  • 174.Tanida S, Yoshitomi H, Ishikawa M, Kasahara T, Murata K, Shibuya H, Ito H, Nakamura T. IL-27-producing CD14(+) cells infiltrate inflamed joints of rheumatoid arthritis and regulate inflammation and chemotactic migration. Cytokine 2011; 55: 237–44. [DOI] [PubMed] [Google Scholar]
  • 175.Shen H, Xia L, Xiao W, Lu J. Increased levels of interleukin-27 in patients with rheumatoid arthritis. Arthritis Rheum 2011; 63: 860–1. [DOI] [PubMed] [Google Scholar]
  • 176.Alghasham A, Rasheed Z. Therapeutic targets for rheumatoid arthritis: progress and promises. Autoimmunity 2014; 47: 77–94. [DOI] [PubMed] [Google Scholar]
  • 177.Radstake TR, Roelofs MF, Jenniskens YM, Oppers-Walgreen B, van Riel PL, Barrera P, Joosten LA, van den Berg WB. Expression of toll-like receptors 2 and 4 in rheumatoid synovial tissue and regulation by proinflammatory cytokines interleukin-12 and interleukin-18 via interferon-gamma. Arthritis Rheum 2004; 50: 3856–65. [DOI] [PubMed] [Google Scholar]
  • 178.Abdollahi-Roodsaz S, Joosten LA, Helsen MM, Walgreen B, van Lent PL, van den Bersselaar LA, Koenders MI, van den Berg WB. Shift from toll-like receptor 2 (TLR-2) toward TLR-4 dependency in the erosive stage of chronic streptococcal cell wall arthritis coincident with TLR-4-mediated interleukin-17 production. Arthritis Rheum 2008; 58: 3753–64. [DOI] [PubMed] [Google Scholar]
  • 179.Kim KW, Cho ML, Lee SH, Oh HJ, Kang CM, Ju JH, Min SY, Cho YG, Park SH, Kim HY. Human rheumatoid synovial fibroblasts promote osteoclastogenic activity by activating RANKL via TLR-2 and TLR-4 activation. Immunol Lett 2007; 110: 54–64. [DOI] [PubMed] [Google Scholar]
  • 180.Huang QQ, Pope RM. The role of toll-like receptors in rheumatoid arthritis. Curr Rheumatol Rep 2009; 11: 357–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Venkatesha SH, Dudics S, Acharya B, Moudgil KD. Cytokine-modulating strategies and newer cytokine targets for arthritis therapy. Int J Mol Sci 2015; 16: 887–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Selmi C, Generali E, Massarotti M, Bianchi G, Scire CA. New treatments for inflammatory rheumatic disease. Immunol Res 2014; 60: 277–88. [DOI] [PubMed] [Google Scholar]
  • 183.Moots RJ, Mays R, Stephens J, Tarallo M. Burden of dose escalation with tumour necrosis factor inhibitors in rheumatoid arthritis: a systematic review of frequency and costs. Clin Exp Rheumatol 2015; 33: 737–45. [PubMed] [Google Scholar]
  • 184.Danis VA, Franic GM, Rathjen DA, Laurent RM, Brooks PM. Circulating cytokine levels in patients with rheumatoid arthritis: results of a double blind trial with sulphasalazine. Ann Rheum Dis 1992; 51: 946–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Rodriguez-Carrio J, Alperi-Lopez M, Lopez P, Ballina-Garcia FJ, Abal F, Suarez A. Antibodies to high-density lipoproteins are associated with inflammation and cardiovascular disease in rheumatoid arthritis patients. Transl Res 2015; 166: 529–39. [DOI] [PubMed] [Google Scholar]
  • 186.Carvalheiro H, Duarte C, Silva-Cardoso S, da Silva JA, Souto-Carneiro MM. CD8+ T cell profiles in patients with rheumatoid arthritis and their relationship to disease activity. Arthritis Rheumatol 2015; 67: 363–71. [DOI] [PubMed] [Google Scholar]
  • 187.Karonitsch T, Dalwigk K, Byrne R, Niedereiter B, Cetin E, Wanivenhaus A, Scheinecker C, Smolen JS, Kiener HP. IFN-gamma promotes fibroblast-like synoviocytes motility. Ann Rheum Dis 2010; 69: A63–A63. [Google Scholar]
  • 188.Pavlovic V, Dimic A, Milenkovic S, Krtinic D. Serum levels of IL-17, IL-4, and INFgamma in Serbian patients with early rheumatoid arthritis. J Res Med Sci 2014; 19: 18–22. [PMC free article] [PubMed] [Google Scholar]
  • 189.Jia XY, Chang Y, Sun XJ, Dai X, Wei W. The role of prostaglandin E2 receptor signaling of dendritic cells in rheumatoid arthritis. Int Immunopharmacol 2014; 23: 163–9. [DOI] [PubMed] [Google Scholar]
  • 190.Sakata D, Yao C, Narumiya S. Prostaglandin E2, an immunoactivator. J Pharmacol Sci 2010; 112: 1–5. [DOI] [PubMed] [Google Scholar]
  • 191.Huang QC, Huang RY. The cyclooxygenase-2/thromboxane A2 pathway: a bridge from rheumatoid arthritis to lung cancer? Cancer Lett 2014; 354: 28–32. [DOI] [PubMed] [Google Scholar]
  • 192.Fan HW, Liu GY, Zhao CF, Li XF, Yang XY. Differential expression of COX-2 in osteoarthritis and rheumatoid arthritis. Genet Mol Res 2015; 14: 12872–9. [DOI] [PubMed] [Google Scholar]
  • 193.Wang MJ, Huang Y, Huang RY, Chen XM, Zhou YY, Yu WL, Chu YL, Huang QC. Determination of role of thromboxane A2 in rheumatoid arthritis. Discov Med 2015; 19(102): 23–32. [PubMed] [Google Scholar]
  • 194.Blake DR, Bacon PA, Eastham EJ, Brigham K. Synovial fluid ferritin in rheumatoid arthritis. BMJ 1980; 281: 715–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Rothwell RS, Davis P. Relationship between serum ferritin, anemia, and disease activity in acute and chronic rheumatoid arthritis. Rheumatol Int 1981; 1: 65–7. [DOI] [PubMed] [Google Scholar]
  • 196.Biemond P, Swaak AJG, Vaneijk HG, Koster JF. Intraarticular ferritin-bound iron in rheumatoid arthritis – a factor that increases oxygen free radical-induced tissue destruction. Arthritis Rheumat 1986; 29: 1187–93. [DOI] [PubMed] [Google Scholar]
  • 197.Palermo C, Maddali Bongi S, Bianucci G. Relationship between serum ferritin, iron stores and disease activity in rheumatoid arthritis. Ric Clin Lab 1986; 16: 463–9. [DOI] [PubMed] [Google Scholar]
  • 198.Biemond P, Swaak AJG, Vaneijk HG, Koster JF. Superoxide dependent iron release from ferritin in inflammatory diseases. Free Radical Biol Med 1988; 4: 185–98. [DOI] [PubMed] [Google Scholar]
  • 199.Abe E, Arai M. Synovial fluid ferritin in traumatic hemarthrosis, rheumatoid arthritis and osteoarthritis. Tohoku J Exp Med 1992; 168: 499–505. [DOI] [PubMed] [Google Scholar]
  • 200.Yildirim K, Karatay S, Melikoglu MA, Gureser G, Ugur M, Senel K. Associations between acute phase reactant levels and disease activity score (DAS28) in patients with rheumatoid arthritis. Ann Clin Lab Sci 2004; 34: 423–6. [PubMed] [Google Scholar]
  • 201.de Bont N, Netea MG, Rovers C, Smilde T, Hijmans A, Demacker PN, van der Meer JW, Stalenhoef AF. LPS-induced release of IL-1 beta, IL-1Ra, IL-6, and TNF-alpha in whole blood from patients with familial hypercholesterolemia: no effect of cholesterol-lowering treatment. J Interferon Cytokine Res 2006; 26: 101–7. [DOI] [PubMed] [Google Scholar]
  • 202.van Breukelen-van der Stoep DF, Klop B, van Zeben D, Hazes JM, Castro Cabezas M. Cardiovascular risk in rheumatoid arthritis: how to lower the risk? Atherosclerosis 2013; 231: 163–72. [DOI] [PubMed] [Google Scholar]
  • 203.Wruck CJ, Fragoulis A, Gurzynski A, Brandenburg LO, Kan YW, Chan K, Hassenpflug J, Freitag-Wolf S, Varoga D, Lippross S, Pufe T. Role of oxidative stress in rheumatoid arthritis: insights from the Nrf2-knockout mice. Ann Rheum Dis 2011; 70: 844–50. [DOI] [PubMed] [Google Scholar]
  • 204.Veselinovic M, Barudzic N, Vuletic M, Zivkovic V, Tomic-Lucic A, Djuric D, Jakovljevic V. Oxidative stress in rheumatoid arthritis patients: relationship to diseases activity. Mol Cell Biochem 2014; 391: 225–32. [DOI] [PubMed] [Google Scholar]
  • 205.Vasanthi P, Nalini G, Rajasekhar G. Status of oxidative stress in rheumatoid arthritis. Int J Rheum Dis 2009; 12: 29–33. [DOI] [PubMed] [Google Scholar]
  • 206.Nurmohamed MT, Heslinga M, Kitas GD. Cardiovascular comorbidity in rheumatic diseases. Nat Rev Rheumatol 2015; 11: 693–704. [DOI] [PubMed] [Google Scholar]
  • 207.Hollan I, Dessein PH, Ronda N, Wasko MC, Svenungsson E, Agewall S, Cohen-Tervaert JW, Maki-Petaja K, Grundtvig M, Karpouzas GA, Meroni PL. Prevention of cardiovascular disease in rheumatoid arthritis. Autoimmun Rev 2015; 14: 952–69. [DOI] [PubMed] [Google Scholar]
  • 208.Skeoch S, Bruce IN. Atherosclerosis in rheumatoid arthritis: is it all about inflammation? Nat Rev Rheumatol 2015; 11: 390–400. [DOI] [PubMed] [Google Scholar]
  • 209.Gasparyan AY, Stavropoulos-Kalinoglou A, Mikhailidis DP, Douglas KM, Kitas GD. Platelet function in rheumatoid arthritis: arthritic and cardiovascular implications. Rheumatol Int 2011; 31: 153–64. [DOI] [PubMed] [Google Scholar]
  • 210.Nurmohamed MT. Cardiovascular risk in rheumatoid arthritis. Autoimmun Rev 2009; 8: 663–7. [DOI] [PubMed] [Google Scholar]
  • 211.Crepaldi G, Scirè CA, Carrara G, Sakellariou G, Caporali R, Hmamouchi I, Dougados M, Montecucco C. Cardiovascular comorbidities relate more than others with disease activity in rheumatoid arthritis. PLoS One 2016; 11: e0146991–e0146991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermudez-Humaran LG, Smirnova N, Berge M, Sulpice T, Lahtinen S, Ouwehand A, Langella P, Rautonen N, Sansonetti PJ, Burcelin R. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol Med 2011; 3: 559–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Amar J, Lange C, Payros G, Garret C, Chabo C, Lantieri O, Courtney M, Marre M, Charles MA, Balkau B, Burcelin R. Blood microbiota dysbiosis is associated with the onset of cardiovascular events in a large general population: the D.E.S.I.R. study. PLoS One 2013; 8: e54461–e54461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Amar J, Serino M, Lange C, Chabo C, Iacovoni J, Mondot S, Lepage P, Klopp C, Mariette J, Bouchez O, Perez L, Courtney M, Marre M, Klopp P, Lantieri O, Dore J, Charles M, Balkau B, Burcelin R. Involvement of tissue bacteria in the onset of diabetes in humans: evidence for a concept. Diabetologia 2011; 54: 3055–61. [DOI] [PubMed] [Google Scholar]
  • 215.Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011; 472: 57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 2013; 368: 1575–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.El Miedany Y. Co-morbidity index in rheumatoid arthritis: time to think. Clin Rheumatol 2015; 34: 1995–2000. [DOI] [PubMed] [Google Scholar]
  • 218.Gullick NJ, Scott DL. Co-morbidities in established rheumatoid arthritis. Best Pract Res Clin Rheumatol 2011; 25: 469–83. [DOI] [PubMed] [Google Scholar]
  • 219.Kell DB, Pretorius E. Serum ferritin is an important disease marker, and is mainly a leakage product from damaged cells. Metallomics 2014; 6: 748–73. [DOI] [PubMed] [Google Scholar]
  • 220.Pretorius E, Kell DB. Diagnostic morphology: biophysical indicators for iron-driven inflammatory diseases. Integrative Biol 2014; 6: 486–510. [DOI] [PubMed] [Google Scholar]
  • 221.Pretorius E, Olumuyiwa-Akeredolu OO, Mbotwe S, Bester J. Erythrocytes and their role as health indicator: using structure in a patient-orientated precision medicine approach. Blood Rev 2016;30:263–74. [DOI] [PubMed]
  • 222.Mukubo Y, Kawamata M. Perioperative hypercoagulability in patients with rheumatoid arthritis: sonoclot study. J Anesthes 2004; 18: 62–4. [DOI] [PubMed] [Google Scholar]
  • 223.Shoenfeld Y, Gerli R, Doria A, Matsuura E, Cerinic MM, Ronda N, Jara LJ, Abu-Shakra M, Meroni PL, Sherer Y. Accelerated atherosclerosis in autoimmune rheumatic diseases. Circulation 2005; 112: 3337–47. [DOI] [PubMed] [Google Scholar]
  • 224.Bisoendial RJ, Stroes ESG, Tak PP. Where the immune response meets the vessel wall. Neth J Med 2009; 67: 328–33. [PubMed] [Google Scholar]
  • 225.Bisoendial RJ, Levi M, Tak PP, Stroes ESG. The prothrombotic state in rheumatoid arthritis: an additive risk factor for adverse cardiovascular events. Semin Thromb Hemost 2010; 36: 452–7. [DOI] [PubMed] [Google Scholar]
  • 226.Bisoendial RJ, Stroes ESG, Kastelein JJ, Tak PP. Targeting cardiovascular risk in rheumatoid arthritis: a dual role for statins. Nat Rev Rheumatol 2010; 6: 157–64. [DOI] [PubMed] [Google Scholar]
  • 227.Bisoendial RJ, Stroes ESG, Tak PP. Critical determinants of cardiovascular risk in rheumatoid arthritis. Curr Pharmaceut Des 2011; 17: 21–6. [DOI] [PubMed] [Google Scholar]
  • 228.Chung WS, Peng CL, Lin CL, Chang YJ, Chen YF, Chiang JY, Sung FC, Kao CH. Rheumatoid arthritis increases the risk of deep vein thrombosis and pulmonary thromboembolism: a nationwide cohort study. Ann Rheum Dis 2014;73:1774–80. [DOI] [PubMed]
  • 229.Ingegnoli F, Fantini F, Favalli EG, Soldi A, Griffini S, Galbiati V, Meroni PL, Cugno M. Inflammatory and prothrombotic biomarkers in patients with rheumatoid arthritis: effects of tumor necrosis factor-alpha blockade. J Autoimmun 2008; 31: 175–9. [DOI] [PubMed] [Google Scholar]
  • 230.Jin T, Bokarewa M, Amu S, Tarkowski A. Impact of short-term therapies with biologics on prothrombotic biomarkers in rheumatoid arthritis. Clin Exp Rheumatol 2009; 27: 491–4. [PubMed] [Google Scholar]
  • 231.Di Franco M, Gambardella L, AC DIL, Malorni W, Valesini G, Straface E. Possible implication of red blood cells in the prothrombotic risk in early rheumatoid arthritis. J Rheumatol 2015; 42: 1352–4. [DOI] [PubMed] [Google Scholar]
  • 232.Pretorius E, Mbotwe S, Bester J, Robinson C, Kell DB. Acute induction of anomalous blood clotting by highly substoichiometric levels of bacterial lipopolysaccharide (LPS). bioRxiv. Epub ahead of print 16 May 2016. DOI: http://dx.doi.org/10.1101/053538. [DOI] [PMC free article] [PubMed]
  • 233.Konokhova AI, Chernova DN, Moskalensky AE, Strokotov DI, Yurkin MA, Chernyshev AV, Maltsev VP. Super-resolved calibration-free flow cytometric characterization of platelets and cell-derived microparticles in platelet-rich plasma. Cytometry A 2016; 89: 159–68. [DOI] [PubMed] [Google Scholar]
  • 234.Davey HM, Kell DB. Flow cytometry and cell sorting of heterogeneous microbial populations: the importance of single-cell analysis. Microbiol Rev 1996; 60: 641–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Ardoin SP, Shanahan JC, Pisetsky DS. The role of microparticles in inflammation and thrombosis. Scand J Immunol 2007; 66: 159–65. [DOI] [PubMed] [Google Scholar]
  • 236.Simak J, Gelderman MP. Cell membrane microparticles in blood and blood products: potentially pathogenic agents and diagnostic markers. Transfus Med Rev 2006; 20: 1–26. [DOI] [PubMed] [Google Scholar]
  • 237.Hisada Y, Alexander W, Kasthuri R, Voorhees P, Mobarrez F, Taylor A, McNamara C, Wallen H, Witkowski M, Key NS, Rauch U, Mackman N. Measurement of microparticle tissue factor activity in clinical samples: a summary of two tissue factor-dependent FXa generation assays. Thromb Res 2016; 139: 90–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Goubran HA, Burnouf T, Stakiw J, Seghatchian J. Platelet microparticle: a sensitive physiological “fine tuning” balancing factor in health and disease. Transfus Apher Sci 2015; 52: 12–8. [DOI] [PubMed] [Google Scholar]
  • 239.van Eijk IC, Tushuizen ME, Sturk A, Dijkmans BA, Boers M, Voskuyl AE, Diamant M, Wolbink GJ, Nieuwland R, Nurmohamed MT. Circulating microparticles remain associated with complement activation despite intensive anti-inflammatory therapy in early rheumatoid arthritis. Ann Rheum Dis 2010; 69: 1378–82. [DOI] [PubMed] [Google Scholar]
  • 240.Knijff-Dutmer EA, Koerts J, Nieuwland R, Kalsbeek-Batenburg EM, van de Laar MA. Elevated levels of platelet microparticles are associated with disease activity in rheumatoid arthritis. Arthritis Rheum 2002; 46: 1498–503. [DOI] [PubMed] [Google Scholar]
  • 241.Berckmans RJ, Nieuwland R, Tak PP, Boing AN, Romijn FP, Kraan MC, Breedveld FC, Hack CE, Sturk A. Cell-derived microparticles in synovial fluid from inflamed arthritic joints support coagulation exclusively via a factor VII-dependent mechanism. Arthritis Rheum 2002; 46: 2857–66. [DOI] [PubMed] [Google Scholar]
  • 242.Gitz E, Pollitt AY, Gitz-Francois JJ, Alshehri O, Mori J, Montague S, Nash GB, Douglas MR, Gardiner EE, Andrews RK, Buckley CD, Harrison P, Watson SP. CLEC-2 expression is maintained on activated platelets and on platelet microparticles. Blood 2014; 124: 2262–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Nurden AT. Platelets, inflammation and tissue regeneration. Thromb Haemost 2011;105(Suppl 1):S13–33. [DOI] [PubMed]
  • 244.Sellam J, Proulle V, Jungel A, Ittah M, Miceli Richard C, Gottenberg JE, Toti F, Benessiano J, Gay S, Freyssinet JM, Mariette X. Increased levels of circulating microparticles in primary Sjogren's syndrome, systemic lupus erythematosus and rheumatoid arthritis and relation with disease activity. Arthritis Res Ther 2009; 11: R156–R156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Olumuyiwa-Akeredolu OO, Pretorius E. Platelet and red blood cell interactions and their role in rheumatoid arthritis. Rheumatol Int 2015; 35: 1955–64. [DOI] [PubMed] [Google Scholar]
  • 246.Rodríguez-Carrio J, Alperi-López M, Lopez P, Alonso-Castro S, Carro-Esteban SR, Ballina-Garcíá FJ, Suarez A. Altered profile of circulating microparticles in rheumatoid arthritis patients. Clin Sci 2015; 128: 437–48. [DOI] [PubMed] [Google Scholar]
  • 247.Vijayakumar D, Suresh K, Manoharan S. Altered pattern of lipids in plasma and erythrocyte membranes of rheumatoid arthritis patients. Ind J Clin Biochem 2005; 20: 52–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.van Zwieten R, Bochem AE, Hilarius PM, van Bruggen R, Bergkamp F, Hovingh GK, Verhoeven AJ. The cholesterol content of the erythrocyte membrane is an important determinant of phosphatidylserine exposure. Biochim Biophys Acta MolCell Biol Lipids 2012; 1821: 1493–500. [DOI] [PubMed] [Google Scholar]
  • 249.Kosenko EA, Aliev G, Tikhonova LA, Li Y, Poghosyan AC, Kaminsky YG. Antioxidant status and energy state of erythrocytes in alzheimer dementia: probing for markers. CNS Neurol Disord 2012; 11: 926–32. [DOI] [PubMed] [Google Scholar]
  • 250.Maurya PK, Kumar P, Chandra P. Biomarkers of oxidative stress in erythrocytes as a function of human age. World J Methodol 2015; 5: 216–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Kosenko EA, Aliev G, Kaminsky YG. Relationship between chronic disturbance of 2,3-diphosphoglycerate metabolism in erythrocytes and alzheimer disease. CNS Neurol Disord Drug Targets 2016;15:113–23. [DOI] [PubMed]
  • 252.Tikhonova LA, Kaminsky YG, Reddy VP, Li Y, Solomadin IN, Kosenko EA, et al. Impact of amyloid β25-35 on membrane stability, energy metabolism, and antioxidant enzymes in erythrocytes. Am J Alzheimer's Dis Other Dement 2014; 29: 685–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Kaminsky YG, Reddy VP, Ashraf G, Ahmad A, Benberin VV, Kosenko EA, et al. Age-related defects in erythrocyte 2,3-diphosphoglycerate metabolism in dementia. Aging Dis 2013; 4: 244–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Kosenko EA, Solomadin IN, Tikhonova LA, Reddy VP, Aliev G, Kaminsky YG. Pathogenesis of alzheimer disease: role of oxidative stress, amyloid-β peptides, systemic ammonia and erythrocyte energy metabolism. CNS Neurol Disord 2014; 13: 112–9. [DOI] [PubMed] [Google Scholar]
  • 255.Kell DB, Pretorius E. Proteins behaving badly. Substoichiometric molecular control and amplification of the initiation and nature of amyloid fibril formation: lessons from and for blood clotting. Prog Biophys Mol Biol. Epub ahead of print 21 August 2016. DOI: 10.1016/j.pbiomolbio.2016.08.006. [DOI] [PubMed]
  • 256.Pretorius E, Mbotwe S, Bester J, Robinson CJ, Kell DB. Acute induction of anomalous and amyloidogenic blood clotting by molecular amplification of highly substoichiometric levels of bacterial lipopolysaccharide. J R Soc Interface 2016;13:20160539. [DOI] [PMC free article] [PubMed]
  • 257.Kell DB, Pretorius E. Substoichiometric molecular control and amplification of the initiation and nature of amyloid fibril formation: lessons from and for blood clotting. bioRxiv 2016; 2016: 054734–054734. [DOI] [PubMed] [Google Scholar]
  • 258.Kell DB, Pretorius E. To what extent are the terminal stages of sepsis, septic shock, SIRS, and multiple organ dysfunction syndrome actually driven by a toxic prion/amyloid form of fibrin? bioRxiv preprint. bioRxiv 2016; 2016: 057851–057851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Kell DB, Kenny LC. A dormant microbial component in the development of pre-eclampsia. BioRxiv preprint. bioRxiv 2016; 2016: 057356–057356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Espargaró A, Busquets MA, Estelrich J, Sabate R. Key Points concerning amyloid infectivity and prion-like neuronal invasion. Front Mol Neurosci 2016; 9: 29–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Fernández MS. Human IAPP amyloidogenic properties and pancreatic beta-cell death. Cell Cal 2014; 56: 416–27. [DOI] [PubMed] [Google Scholar]
  • 262.Ow SY, Dunstan DE. A brief overview of amyloids and Alzheimer's disease. Protein Sci 2014; 23: 1315–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Stefani M. Structural features and cytotoxicity of amyloid oligomers: implications in Alzheimer's disease and other diseases with amyloid deposits. Prog Neurobiol 2012; 99: 226–45. [DOI] [PubMed] [Google Scholar]
  • 264.Husby G. Amyloidosis and rheumatoid arthritis. Clin Exp Rheumatol 1985; 3: 173–80. [PubMed] [Google Scholar]
  • 265.O'Hara R, Murphy EP, Whitehead AS, FitzGerald O, Bresnihan B. Acute-phase serum amyloid A production by rheumatoid arthritis synovial tissue. Arthritis Res 2000; 2: 142–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Cunnane G, Whitehead AS. Amyloid precursors and amyloidosis in rheumatoid arthritis. Baillieres Best Pract Res Clin Rheumatol 1999; 13: 615–28. [DOI] [PubMed] [Google Scholar]
  • 267.Kobayashi H, Tada S, Fuchigami T, Okuda Y, Takasugi K, Matsumoto T, Iida M, Aoyagi K, Iwashita A, Daimaru Y, Fujishima M. Secondary amyloidosis in patients with rheumatoid arthritis: diagnostic and prognostic value of gastroduodenal biopsy. Br J Rheumatol 1996; 35: 44–9. [DOI] [PubMed] [Google Scholar]
  • 268.Nakamura T. Amyloid A amyloidosis secondary to rheumatoid arthritis: pathophysiology and treatments. Clin Exp Rheumatol 2011; 29: 850–7. [PubMed] [Google Scholar]
  • 269.Broadhurst D, Kell DB. Statistical strategies for avoiding false discoveries in metabolomics and related experiments. Metabolomics 2006; 2: 171–96. [Google Scholar]
  • 270.Vinet É, Kuriya B, Widdifield J, Bernatsky S. Rheumatoid arthritis disease severity indices in administrative databases: a systematic review. J Rheumatol 2011; 38: 2318–25. [DOI] [PubMed] [Google Scholar]
  • 271.Suissa S, Garbe E. Primer: administrative health databases in observational studies of drug effects–advantages and disadvantages. Nat Clin Pract Rheumatol 2007; 3: 725–32. [DOI] [PubMed] [Google Scholar]
  • 272.Arnett FC, Edworthy SM, Bloch DA, Mcshane DJ, Fries JF, Cooper NS, Healey LA, Kaplan SR, Liang MH, Luthra HS, Medsger TA, Mitchell DM, Neustadt DH, Pinals RS, Schaller JG, Sharp JT, Wilder RL, Hunder GG. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 1988; 31: 315–24. [DOI] [PubMed] [Google Scholar]
  • 273.Ward MM, Guthrie LC, Alba MI. Measures of arthritis activity associated with patient-reported improvement in rheumatoid arthritis when assessed prospectively versus retrospectively. Arthritis Care Res 2015; 67: 776–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Smedstad LM, Kvien TK, Moum T, Vaglum P. Correlates of patients' global assessment of arthritis impact. A 2-year study of 216 patients with RA. Scand J Rheumatol 1997; 26: 259–65. [DOI] [PubMed] [Google Scholar]
  • 275.Khan NA, Spencer HJ, Abda EA, Alten R, Pohl C, Ancuta C, Cazzato M, Geher P, Gossec L, Henrohn D, Hetland ML, Inanc N, Jacobs JW, Kerzberg E, Majdan M, Oyoo O, Peredo-Wende RA, Selim ZI, Skopouli FN, Sulli A, Horslev-Petersen K, Taylor PC, Sokka T, group Q-R. Patient's global assessment of disease activity and patient's assessment of general health for rheumatoid arthritis activity assessment: are they equivalent? Ann Rheum Dis 2012; 71: 1942–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Smolen JS, Landewé R, Breedveld FC, Buch M, Burmester G, Dougados M, Emery P, Gaujoux-Viala C, Gossec L, Nam J, Ramiro S, Winthrop K, de Wit M, Aletaha D, Betteridge N, Bijlsma JW, Boers M, Buttgereit F, Combe B, Cutolo M, Damjanov N, Hazes JM, Kouloumas M, Kvien TK, Mariette X, Pavelka K, van Riel PL, Rubbert-Roth A, Scholte-Voshaar M, Scott DL, Sokka-Isler T, Wong JB, van der Heijde D. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis 2014; 73: 492–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Nam JL, Ramiro S, Gaujoux-Viala C, Takase K, Leon-Garcia M, Emery P, Gossec L, Landewé R, Smolen JS, Buch MH. Efficacy of biological disease-modifying antirheumatic drugs: a systematic literature review informing the 2013 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2014; 73: 516–28. [DOI] [PubMed] [Google Scholar]
  • 278.Gaujoux-Viala C, Nam J, Ramiro S, Landewe R, Buch MH, Smolen JS, Gossec L. Efficacy of conventional synthetic disease-modifying antirheumatic drugs, glucocorticoids and tofacitinib: a systematic literature review informing the 2013 update of the EULAR recommendations for management of rheumatoid arthritis. Ann Rheum Dis 2014; 73: 510–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Colebatch AN, Edwards CJ, Østergaard M, van der Heijde D, Balint PV, D'Agostino MA, Forslind K, Grassi W, Haavardsholm EA, Haugeberg G, Jurik AG, Landewé RB, Naredo E, O'Connor PJ, Ostendorf B, Potočki K, Schmidt WA, Smolen JS, Sokolovic S, Watt I, Conaghan PG. EULAR recommendations for the use of imaging of the joints in the clinical management of rheumatoid arthritis. Ann Rheum Dis 2013; 72: 804–14. [DOI] [PubMed] [Google Scholar]
  • 280.Rains CP, Noble S, Faulds D. Sulfasalazine. A review of its pharmacological properties and therapeutic efficacy in the treatment of rheumatoid arthritis. Drugs 1995; 50: 137–56. [DOI] [PubMed] [Google Scholar]
  • 281.Plosker GL, Croom KF. Sulfasalazine: a review of its use in the management of rheumatoid arthritis. Drugs 2005; 65: 1825–49. [DOI] [PubMed] [Google Scholar]
  • 282.Kruszewska H, Zareba T, Tyski S. Antimicrobial activity of selected non-antibiotics – activity of methotrexate against Staphylococcus aureus strains. Acta Pol Pharm 2000;57(Suppl):117–9. [PubMed]
  • 283.Imwong M, Russell B, Suwanarusk R, Nzila A, Leimanis ML, Sriprawat K, Kaewpongsri S, Phyo AP, Snounou G, Nosten F, Renia L. Methotrexate is highly potent against pyrimethamine-resistant Plasmodium vivax. J Infect Dis 2011; 203: 207–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Smith CJ, Sayles H, Mikuls TR, Michaud K. Minocycline and doxycycline therapy in community patients with rheumatoid arthritis: prescribing patterns, patient-level determinants of use, and patient-reported side effects. Arthritis Res Ther 2011; 13: R168–R168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Mestres J, Gregori-Puigjané E. Conciliating binding efficiency and polypharmacology. Trends Pharmacol Sci 2009; 30: 470–4. [DOI] [PubMed] [Google Scholar]
  • 286.Kell DB, Dobson PD, Bilsland E, Oliver SG. The promiscuous binding of pharmaceutical drugs and their transporter-mediated uptake into cells: what we (need to) know and how we can do so. Drug Disc Today 2013; 18: 218–39. [DOI] [PubMed] [Google Scholar]
  • 287.Langevitz P, Bank I, Zemer D, Book M, Pras M. Treatment of resistant rheumatoid arthritis with minocycline: an open study. J Rheumatol 1992; 19: 1502–4. [PubMed] [Google Scholar]
  • 288.Kloppenburg M, Breedveld FC, Terwiel JP, Mallee C, Dijkmans BA. Minocycline in active rheumatoid arthritis. A double-blind, placebo-controlled trial. Arthritis Rheum 1994; 37: 629–36. [DOI] [PubMed] [Google Scholar]
  • 289.Tilley BC, Alarcón GS, Heyse SP, Trentham DE, Neuner R, Kaplan DA, Clegg DO, Leisen JC, Buckley L, Cooper SM, Duncan H, Pillemer SR, Tuttleman M, Fowler SE. Minocycline in rheumatoid arthritis. A 48-week, double-blind, placebo-controlled trial. MIRA Trial Group. Ann Intern Med 1995; 122: 81–9. [DOI] [PubMed] [Google Scholar]
  • 290.O'Dell JR, Haire CE, Palmer W, Drymalski W, Wees S, Blakely K, Churchill M, Eckhoff PJ, Weaver A, Doud D, Erikson N, Dietz F, Olson R, Maloley P, Klassen LW, Moore GF. Treatment of early rheumatoid arthritis with minocycline or placebo: results of a randomized, double-blind, placebo-controlled trial. Arthritis Rheum 1997; 40: 842–8. [DOI] [PubMed] [Google Scholar]
  • 291.O'Dell JR, Paulsen G, Haire CE, Blakely K, Palmer W, Wees S, Eckhoff PJ, Klassen LW, Churchill M, Doud D, Weaver A, Moore GF. Treatment of early seropositive rheumatoid arthritis with minocycline: four-year followup of a double-blind, placebo-controlled trial. Arthritis Rheum 1999; 42: 1691–5. [DOI] [PubMed] [Google Scholar]
  • 292.Langevitz P, Livneh A, Bank I, Pras M. Benefits and risks of minocycline in rheumatoid arthritis. Drug Saf 2000; 22: 405–14. [DOI] [PubMed] [Google Scholar]
  • 293.O'Dell JR, Elliott JR, Mallek JA, Mikuls TR, Weaver CA, Glickstein S, Blakely KM, Hausch R, Leff RD. Treatment of early seropositive rheumatoid arthritis: doxycycline plus methotrexate versus methotrexate alone. Arthritis Rheum 2006; 54: 621–7. [DOI] [PubMed] [Google Scholar]
  • 294.Alarcón GS. Early rheumatoid arthritis: combination therapy of doxycycline plus methotrexate versus methotrexate monotherapy. Nat Clin Pract Rheumatol 2006; 2: 296–7. [DOI] [PubMed] [Google Scholar]
  • 295.Morgan C, McBeth J, Cordingley L, Watson K, Hyrich KL, Symmons DP, Bruce IN. The influence of behavioural and psychological factors on medication adherence over time in rheumatoid arthritis patients: a study in the biologics era. Rheumatology 2015; 54: 1780–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Watson K, Symmons D, Griffiths I, Silman A. The British Society for Rheumatology biologics register. Ann Rheum Dis 2005;64(Suppl 4):iv42–3. [DOI] [PMC free article] [PubMed]
  • 297.Tougaard P, Zervides KA, Skov S, Hansen AK, Pedersen AE. Biologics beyond TNF-alpha inhibitors and the effect of targeting the homologues TL1A-DR3 pathway in chronic inflammatory disorders. Immunopharmacol Immunotoxicol 2016; 38: 29–38. [DOI] [PubMed] [Google Scholar]
  • 298.Symmons DP, Silman AJ. The world of biologics. Lupus 2006; 15: 122–6. [DOI] [PubMed] [Google Scholar]
  • 299.Perez-Alvarez R, Pérez-de-Lis M, Ramos-Casals M. Biogeas study group. Biologics-induced autoimmune diseases. Curr Opin Rheumatol 2013; 25: 56–64. [DOI] [PubMed] [Google Scholar]
  • 300.Ramos-Casals M, Brito-Zerón P, Soto MJ, Cuadrado MJ, Khamashta MA. Autoimmune diseases induced by TNF-targeted therapies. Best Pract Res Clin Rheumatol 2008; 22: 847–61. [DOI] [PubMed] [Google Scholar]
  • 301.Ramos-Casals M, Roberto Perez A, Diaz-Lagares C, Cuadrado MJ, Khamashta MA. Biogeas Study Group. Autoimmune diseases induced by biological agents: a double-edged sword? Autoimmun Rev 2010; 9: 188–93. [DOI] [PubMed] [Google Scholar]
  • 302.Voest EE, Vreugdenhil G, Marx JJM. Iron-chelating agents in non-iron overload conditions. Ann Internal Med 1994; 120: 490–9. [DOI] [PubMed] [Google Scholar]
  • 303.Boyle AJ, Mosher RE, Mc CD. Some in vivo effects of chelation. I. Rheumatoid arthritis. J Chronic Dis 1963; 16: 325–8. [DOI] [PubMed] [Google Scholar]
  • 304.Davis P. Penicillamine metal chelates and their possible importance in rheumatoid arthritis – a brief review. Clin Invest Med 1984; 7: 41–4. [PubMed] [Google Scholar]
  • 305.Magaro' M, Zoli A, Altomonte L, Mirone L, Corvino G, Storti S, Marra R, Ricerca BM, Pagano L, Di Cesare L. Iron chelation in rheumatoid arthritis: clinical and laboratory evaluation. Ann Rheum Dis 1990; 49: 268–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Sohn YS, Breuer W, Munnich A, Cabantchik ZI. Redistribution of accumulated cell iron: a modality of chelation with therapeutic implications. Blood 2008; 111: 1690–9. [DOI] [PubMed] [Google Scholar]
  • 307.Baker JF, Ghio AJ. Iron homoeostasis in rheumatic disease. Rheumatology 2009; 48: 1339–44. [DOI] [PubMed] [Google Scholar]
  • 308.Kontoghiorghe CN, Kolnagou A, Kontoghiorghes GJ. Potential clinical applications of chelating drugs in diseases targeting transferrin-bound iron and other metals. Expert Opin Investig Drugs 2013; 22: 591–618. [DOI] [PubMed] [Google Scholar]
  • 309.Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY, Smith WL, Kalow W, Andrews DF. Intramuscular desferrioxamine in patients with Alzheimer's disease. Lancet 1991; 337: 1304–8. [DOI] [PubMed] [Google Scholar]
  • 310.Smith MA, Zhu X, Tabaton M, Liu G, McKeel DW, Jr., Cohen ML, Wang X, Siedlak SL, Dwyer BE, Hayashi T, Nakamura M, Nunomura A, Perry G. Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment. J Alzheimers Dis 2010; 19: 363–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Bush AI. Drug development based on the metals hypothesis of Alzheimer's disease. J Alzheimers Dis 2008; 15: 223–40. [DOI] [PubMed] [Google Scholar]
  • 312.Ganz T. Iron in innate immunity: starve the invaders. Curr Opin Immunol 2009; 21: 63–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Weinberg ED, Miklossy J. Iron withholding: a defense against disease. J Alzheimers Dis 2008; 13: 451–63. [DOI] [PubMed] [Google Scholar]
  • 314.Giakoumi X, Tsironi M, Floudas C, Polymeropoylos E, Papalambros E, Aessopos A. Rheumatoid arthritis in thalassemia intermedia: coincidence or association? Israel Med Assoc J 2005; 7: 667–9. [PubMed] [Google Scholar]
  • 315.Donnelly SC, Joshi NG, Thorburn D, Cooke A, Reid G, Neilson M, Capell H, Stanley AJ. Prevalence of genetic haemochromatosis and iron overload in patients attending rheumatology and joint replacement clinics. Scott Med J 2010; 55: 14–6. [DOI] [PubMed] [Google Scholar]
  • 316.Scher JU, Littman DR, Abramson SB. Microbiome in inflammatory arthritis and human rheumatic diseases. Arthritis Rheumatol 2016; 68: 35–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, Huttenhower C, Littman DR. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2013; 2: e01202–e01202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.He W, Qu T, Yu Q, Wang Z, Lv H, Zhang J, Zhao X, Wang P. LPS induces IL-8 expression through TLR4, MyD88, NF-kappaB and MAPK pathways in human dental pulp stem cells. Int Endod J 2013; 46: 128–36. [DOI] [PubMed] [Google Scholar]
  • 319.Dobashi K, Aihara M, Araki T, Shimizu Y, Utsugi M, Iizuka K, Murata Y, Hamuro J, Nakazawa T, Mori M. Regulation of LPS induced IL-12 production by IFN-gamma and IL-4 through intracellular glutathione status in human alveolar macrophages. Clin Exp Immunol 2001; 124: 290–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Mattei F, Schiavoni G, Belardelli F, Tough DF. IL-15 is expressed by dendritic cells in response to type I IFN, double-stranded RNA, or lipopolysaccharide and promotes dendritic cell activation. J Immunol 2001; 167: 1179–87. [DOI] [PubMed] [Google Scholar]
  • 321.Poltorak A, He XL, Smirnova I, Liu MY, Van Huffel C, Du X, Birdwell D, Alejos E, Silva M, Galanos C, Freudenberg M, Ricciardi-Castagnoli P, Layton B, Beutler B. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 1998; 282: 2085–8. [DOI] [PubMed] [Google Scholar]
  • 322.Poltorak A, Ricciardi-Castagnoli P, Citterio S, Beutler B. Physical contact between lipopolysaccharide and Toll-like receptor 4 revealed by genetic complementation. Proc Natl Acad Sci 2000; 97: 2163–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Beutler B, Du X, Poltorak A. Identification of Toll-like receptor 4 (Tlr4) as the sole conduit for LPS signal transduction: genetic and evolutionary studies. J Endotoxin Res 2001; 7: 277–80. [PubMed] [Google Scholar]
  • 324.Lu YC, Yeh WC, Ohashi PS. LPS/TLR4 signal transduction pathway. Cytokine 2008; 42: 145–51. [DOI] [PubMed] [Google Scholar]
  • 325.O'Neill LAJ, Golenbock D, Bowie AG. The history of Toll-like receptors – redefining innate immunity. Nat Rev Immunol 2013; 13: 453–60. [DOI] [PubMed] [Google Scholar]
  • 326.Verstrepen L, Bekaert T, Chau TL, Tavernier J, Chariot A, Beyaert R. TLR-4, IL-1R and TNF-R signaling to NF-kappa B: variations on a common theme. Cell Mol Life Sci 2008; 65: 2964–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.O'Neill LAJ, Bryant CE, Doyle SL. Therapeutic targeting of toll-like receptors for infectious and inflammatory diseases and cancer. Pharmacol Rev 2009; 61: 177–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011; 34: 637–50. [DOI] [PubMed] [Google Scholar]
  • 329.Noort AR, Tak PP, Tas SW. Non-canonical NF-kappaB signaling in rheumatoid arthritis: Dr Jekyll and Mr Hyde? Arthritis Res Ther 2015; 17: 15–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Vallabhapurapu S, Karin M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol 2009; 27: 693–733. [DOI] [PubMed] [Google Scholar]
  • 331.Kellum JA, Kong L, Fink MP, Weissfeld LA, Yealy DM, Pinsky MR, Fine J, Krichevsky A, Delude RL, Angus DC, Gen IMSI. Understanding the inflammatory cytokine response in pneumonia and sepsis: results of the Genetic and Inflammatory Markers of Sepsis (GenIMS) Study. Arch Intern Med 2007; 167: 1655–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Nelson DE, Ihekwaba AE, Elliott M, Johnson JR, Gibney CA, Foreman BE, Nelson G, See V, Horton CA, Spiller DG, Edwards SW, McDowell HP, Unitt JF, Sullivan E, Grimley R, Benson N, Broomhead D, Kell DB, White MR. Oscillations in NF-kappaB signaling control the dynamics of gene expression. Science 2004; 306: 704–8. [DOI] [PubMed] [Google Scholar]
  • 333.Ashall L, Horton CA, Nelson DE, Paszek P, Ryan S, Sillitoe K, Harper CV, Spiller DG, Unitt JF, Broomhead DS, Kell DB, Rand D, Sée V, White MRH. Pulsatile stimulation determines timing and specificity of NFkappa-B-dependent transcription. Science 2009; 324: 242–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Denes A, Coutts G, Lénárt N, Cruickshank SM, Pelegrin P, Skinner J, Rothwell N, Allan SM, Brough D. AIM2 and NLRC4 inflammasomes contribute with ASC to acute brain injury independently of NLRP3. Proc Natl Acad Sci USA 2015; 112: 4050–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Płóciennikowska A, Hromada-Judycka A, Borzęcka K, Kwiatkowska K. Co-operation of TLR4 and raft proteins in LPS-induced pro-inflammatory signaling. Cell Mol Life Sci 2015; 72: 557–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, Hu L, Shao F. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 2014; 514: 187–92. [DOI] [PubMed] [Google Scholar]
  • 337.Hagar JA, Powell DA, Aachoui Y, Ernst RK, Miao EA. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science 2013; 341: 1250–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol 2013; 13: 397–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Dobson PD, Kell DB. Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule? Nat Rev Drug Disc 2008; 7: 205–20. [DOI] [PubMed] [Google Scholar]
  • 340.Kell DB, Dobson PD, Oliver SG. Pharmaceutical drug transport: the issues and the implications that it is essentially carrier-mediated only. Drug Disc Today 2011; 16: 704–14. [DOI] [PubMed] [Google Scholar]
  • 341.Kell DB. Finding novel pharmaceuticals in the systems biology era using multiple effective drug targets, phenotypic screening, and knowledge of transporters: where drug discovery went wrong and how to fix it. FEBS J 2013; 280: 5957–80. [DOI] [PubMed] [Google Scholar]
  • 342.Kell DB, Oliver SG. How drugs get into cells: tested and testable predictions to help discriminate between transporter-mediated uptake and lipoidal bilayer diffusion. Front Pharmacol 2014; 5: 231–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Kell DB. What would be the observable consequences if phospholipid bilayer diffusion of drugs into cells is negligible? Trends Pharmacol Sci 2015; 36: 15–21. [DOI] [PubMed] [Google Scholar]
  • 344.Proal AD, Albert PJ, Marshall T. Autoimmune disease in the era of the metagenome. Autoimmun Rev 2009; 8: 677–81. [DOI] [PubMed] [Google Scholar]
  • 345.Proal AD, Albert PJ, Marshall TG. The human microbiome and autoimmunity. Curr Opin Rheumatol 2013; 25: 234–40. [DOI] [PubMed] [Google Scholar]
  • 346.Mangin M, Sinha R, Fincher K. Inflammation and vitamin D: the infection connection. Inflamm Res 2014; 63: 803–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Proal AD, Albert PJ, Marshall TG. Inflammatory disease and the human microbiome. Discov Med 2014; 17: 257–65. [PubMed] [Google Scholar]
  • 348.Proal AD, Albert PJ, Marshall TG. Infection, autoimmunity, and vitamin D. In: Shoenfeld Y, Rose NR. (eds). Infection and autoimmunity, New York: Academic Press, 2015, pp. 163–82. [Google Scholar]
  • 349.Kell DB, Westerhoff HV. Metabolic control theory: its role in microbiology and biotechnology. FEMS Microbiol Rev 1986; 39: 305–20. [Google Scholar]
  • 350.Thagard P. Explanatory coherence. Behav Brain Sci 1989; 12: 435–502. [Google Scholar]
  • 351.Thagard P, Verbeurgt K. Coherence as constraint satisfaction. Cogn Sci 1998; 22: 1–24. [Google Scholar]
  • 352.Thagard P. How scientists explain disease, Princeton, NJ: Princeton University Press, 1999. [Google Scholar]
  • 353.Thagard P. Coherence, truth, and the development of scientific knowledge. Philos Sci 2007; 74: 28–47. [Google Scholar]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES