Next Article in Journal
Synthesis of Indoloquinolines: An Intramolecular Cyclization Leading to Advanced Perophoramidine-Relevant Intermediates
Next Article in Special Issue
Novel Thiosemicarbazone Derivatives: In Vitro and In Silico Evaluation as Potential MAO-B Inhibitors
Previous Article in Journal
New Charge Transfer Complexes of K+-Channel-Blocker Drug (Amifampridine; AMFP) for Sensitive Detection; Solution Investigations and DFT Studies
Previous Article in Special Issue
Cannabidiol Attenuates MK-801-Induced Cognitive Symptoms of Schizophrenia in the Passive Avoidance Test in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Common Factors of Alzheimer’s Disease and Rheumatoid Arthritis—Pathomechanism and Treatment

Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland
*
Author to whom correspondence should be addressed.
Molecules 2021, 26(19), 6038; https://doi.org/10.3390/molecules26196038
Submission received: 17 August 2021 / Revised: 20 September 2021 / Accepted: 29 September 2021 / Published: 5 October 2021
(This article belongs to the Special Issue Novel Compounds in the Treatment of the CNS Disorders)

Abstract

:
The accumulation of amyloid plaques, or misfolded fragments of proteins, leads to the development of a condition known as amyloidosis, which is clinically recognized as a systemic disease. Amyloidosis plays a special role in the pathogenesis of neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease, and rheumatoid arthritis (RA). The occurrence of amyloidosis correlates with the aging process of the organism, and since nowadays, old age is determined by the comfort of functioning and the elimination of unpleasant disease symptoms in the elderly, exposure to this subject is justified. In Alzheimer’s disease, amyloid plaques negatively affect glutaminergic and cholinergic transmission and loss of sympathetic protein, while in RA, amyloids stimulated by the activity of the immune system affect the degradation of the osteoarticular bond. The following monograph draws attention to the over-reactivity of the immune system in AD and RA, describes the functionality of the blood–brain barrier as an intermediary medium between RA and AD, and indicates the direction of research to date, focusing on determining the relationship and the cause–effect link between these disorders. The paper presents possible directions for the treatment of amyloidosis, with particular emphasis on innovative therapies.

Graphical Abstract

1. Amyloid Plaques, Structure, Importance, Factors Predisposing to Their Appearance

Amyloidosis is a disease associated with the extracellular accumulation of misfolded protein fragments [1]. Amyloid proteins are characterized as “chameleon proteins” due to their characteristic ability to adopt several conformations [2]. It should be noted that all amyloid proteins are unbranched and have a diameter of 70 to 120 Å [3]. The first studies on isolated amyloid fibrils concerned the knowledge of their structure. X-ray diffraction studies have shown that amyloid-like proteins have a cross-structure β [4,5]. Later studies using NMR analysis further confirmed this hypothesis [6]. Due to the ubiquitous presence of proteins in the body’s cells, amyloidosis is clinically considered a systemic disease [7]. For example, amyloid accumulation plays a special role in the pathogenesis of Alzheimer disease (AD), Parkinson’s disease, or transthyretin amyloidosis [8]. Protein abnormal folding, characteristic of amyloidosis, most often concerns the transthyretin protein (TRT) and the immunoglobulin light chain [9].
In amyloidosis, a protein acquires an abnormal structure through a variety of mechanisms. One of them is that the protein has a naturally intrinsic tendency to adopt a pathological structure that becomes visible with age. The present situation occurs in senile systemic amyloidosis [10]. A separate mechanism is that substitution results in replacing a single amino acid with another, which predisposes to the emergence of hereditary amyloidosis [11]. The last mechanism concerns the proteolytic remodeling of the precursor protein [12].
Amyloid is a structure that occurs in the form of insoluble and pathological deposits. The most common component of the deposits is the SAP (serum amyloid P) glycoprotein, belonging to the pentraxin family, which after radioactive labeling becomes a diagnostic tool for imaging the presence of pathological proteins [13]. The SAP glycoprotein is protected against proteolysis, and this property ensures its resistance to degradation [14]. Proteoglycans are also a component of amyloid that exhibit similar cell deposition kinetics as fibrillar proteins. Moreover, they are located in the vicinity of specific structures of the extracellular matrix such as perlekan, laminin, entactin, and collagen IV [15].
Amyloid accumulation is dependent on several factors. High local protein concentration, low pH in the cell, and the presence of filamentous seeds predispose to the accumulation of pathological deposits. Amyloid deposition is additionally influenced by the presence of specific receptors on the cell surface. RAGE receptors are such receptors—the receptor for advanced glycation end-products, which is the receptor for A-β [16].
The aim of the current research work is to show how amyloid contributes to tissue damage and organ dysfunction. The most common theory explores that the presence of amyloid conglomerates disrupts the proper functioning of structures in its vicinity. The pressure exerted by the amyloid becomes a direct cause of degeneration of the tissues and organs in the vicinity of the amyloid. It is worth mentioning that the accumulation of proteins with an abnormal structure leads to the disturbance of the organism’s homeostasis [17].
The initiation process of amyloid formation is related to the human biological clock (especially with old age), genetic mutations, modifications after protein synthesis, or it results from an increased concentration of amyloidogenic precursor [18]. Many scientific works emphasize the search for links between the aging process of the body and the occurrence of amyloidosis. It is known that the risk of developing amyloidosis increases with age. The aging of the organism predisposes to the appearance of amyloid in tissues and organs and is also a signal for the deposition of proteins with a pathological structure [19]. The above dependence is related to the importance of the protostatic process, which fluctuates with age. The main importance of this process lies in maintaining the proper concentration of proteins in cells, their spatial structure, and their subcellular localization [20]. It should be pointed out that the aging of the organism initiates the deposition of amyloid precursor proteins such as amyloid β (Aβ), tau, or α-synuclein, the presence of which is associated with the pathology of Alzheimer’s or Parkinson’s disease [21]. Furthermore, one must mention that for the fibrillation process, it is necessary to have a sufficient amyloidogenic potential of the protein and the achievement of a critical local concentration by the protein precursor. The initiation process can be enhanced by the above-mentioned factors and the interactions of the protein with extracellular matrices [22].
The most commonly deposited pathological proteins are transthyretin (TRT), islet amyloid polypeptide (IAPP), atrial natriuretic factor (ANF), apolipoprotein AI (ApoAI), and the recently discovered fibulin-like substance containing epidermal growth factor: extracellular matrix protein 1 (EFEMP1) in the extracellular matrix1 (EFEMP1) people, APrP (Prion protein, wild type), ACal ((Pro)calcitonin), ASem1, Semenogelin 1, and huntingtin [23,24]. The authors resigned from a detailed discussion of all pathological proteins. However, it should be noted that AD and RA discussed in the work are diseases with amyloid pathology, which required an introduction to this topic. Moreover, a lot of previous scientific works on amyloidosis have mainly focused on the pathology of amyloid in Alzheimer’s disease [25]. However, in the field of recent research, it has been noted that amyloidosis is a disorder involving many tissues and organs. In these studies, the existence of cause–effect relationships between amyloid accumulation and the occurrence of metabolic diseases, cardiovascular diseases, and skeletal system disorders has been reported [26] (Table 1). The main determinant of research on amyloid is the need to determine the relationship between the aging process of the body and the predisposition to the accumulation of pathological proteins. Exploring similar topics is justified, since life expectancy is longer, and nowadays, old age is determined by the comfort of functioning or the elimination of unpleasant symptoms of diseases accompanying the elderly [27].
Amyloid accumulation takes place throughout the body: in the central and peripheral nervous system and in many organs: the heart, kidneys, and the bone and joint system [29]. The literature describes many types of systemic amyloidosis, four of which are seen most frequently: AL (immunoglobulin light chain amyloidosis), AA (also known as secondary amyloidosis), ATTR (transthyretin amyloidosis), and Aβ2M (Beta-2 Microglobulin amyloidosis) [30]. These amyloidoses are described in Table 2.

2. Amyloid Plaques in AD and RA

AD is a disorder of the nervous system, and according to the estimates of the World Report on Alzheimer’s disease, in 2050, its worldwide incidence will exceed 152 million people [35]. Alzheimer’s disease is a neurodegenerative disease [36]. According to the ICD-10 classification, it is defined as a progressive disorder characterized by the degradation of nerve cells [37]. Loss of neurons in memory regions of the brain causes dementia-like changes. Neurodegeneration correlates with the deterioration of cognitive functions, which, with the progression of the disease, significantly limits the efficient functioning of patients. Moreover, changes in the upper levels of the central nervous system (CNS) cause changes in the patient’s behavior or the emergence of psychiatric disorders [38].
In terms of neurophysiology, Alzheimer’s disease is associated with the appearance of two pathological structures in the brain tissue structures: extracellular amyloid plaques and intracellular neurofibrillary tangles (NFTs). The appearance of the above structures contributes to the occurrence of nerve cell atrophy [39,40].
The process of Aβ peptide formation, which is a disease marker, is related to the enzymatic cleavage of the amyloid precursor protein (APP) [41]. APP alternates in two ways, with two different cutting paths. In the non-amyloidogenic pathway, APP is cleaved by α- and γ-secretase to form Aβ17-40/42 peptide or Aβ1-16 peptide. On the other hand, in case of the amyloidogenic pathway, APP is cleaved sequentially by β- and γ-secretases, leading to the formation of full-length β peptide A (mainly Aβ1-14/42) [42]. Although Aβ1-40 is present in much greater amounts in the brain, Aβ1-42 is a less soluble form and is more prone to accumulate. The accumulation process leads to the formation of conglomerates, which are referred to in the literature as oligomers. The above structures are rearranged into protofibrils and filaments, having their organization in amyloid plaques [43]. One hypothesis is that soluble fibril precursors adopt a specific quaternary conformation that exhibits significant cytotoxicity that is largely unknown at present. Cellular toxicity is dictated by the stimulation of oxidative stress mechanisms and additionally assumes the activation of cellular apoptotic pathways. Moreover, the aforementioned hypothesis indicates that mature, fibrillar amyloid deposits are inactive reservoir structures in balance with less toxic syndromes [44].
The presence of pathological plaques affects the neurotransmitter systems, especially the glutamatergic system. In this system, the main neurotransmitter is glutamate, which plays a key role in the processes of creating memory engrams. The activity of glutamate comes down to its mediation in learning and memory processes. The activity of glutamate is related to the second type of messenger, calcium ions (Ca2+), which help to create the chemical environment necessary for information gathering [45]. Under pathological conditions, excess glutamate causes an excessive intracellular influx of calcium ions, which in turn leads to calcium overload. In the environment of excessive calcium presence, nerve cells die [46]. In Alzheimer’s disease, Aβ plaques cause extracellular accumulation of glutamate and intracellular deposition of calcium ions. Non-fibrillar oligomers, which are likely present in higher concentrations in the immediate vicinity of amyloid plaques, can also disrupt calcium homeostasis [47,48]. Therefore, it is worth noting that Aβ plaques increase the susceptibility of neurons to excitotoxicity and loss of synaptic protein [49].
In Alzheimer’s disease, the dysfunction of cholinergic transmission in the forebrain is also observed. In patients with Alzheimer’s disease, the depleted presynaptic presence of cholinergic markers has been detected in the areas of the cerebral cortex, and it has been shown that the Meynert basal nucleus (NBM) located in the basal forebrain undergoes neurodegeneration as the disease progresses [50,51]. The loss of neurons in the forebrain and limbic system leads to dysfunctional changes in nicotinic receptors with a decrease in their density in the cerebral cortex and influences the activity of muscarinic receptors in the cerebral cortex [52,53].
The cholinergic neurons of the forebrain are the cells with the greatest neurodegenerative potential and also the structures most susceptible to the formation of neurofibrillary tangles [54]. The impoverishment of cholinergic transmission is caused by the presence of amyloid, and this relationship correlates with the negative effect of senile plaques on choline acetyltransferase, which participates in the synthesis of acetylcholine [55]. Studies in animal models have shown that cholinergic loss results in increased accumulation of Aβ and tau protein [56]. On the basis of other studies, it is determined that disturbances of cholinergic transmission in the brains of rats induce pro-inflammatory mechanisms and influence the disclosure of cognitive disorders [57]. Acetylcholine, being a neurotransmitter of the cholinergic system, additionally affects the functionality of the blood–brain barrier. It has been argued that the loss of cholinergic transmission potentially contributes to abnormalities in the diffusion and transport of metabolites between the interstitial fluid and the cerebrospinal fluid. Impairment of substance exchange across the blood–brain barrier impairs the clearance of Aβ from the brain [58]. It has also been shown that defective cholinergic transmission affects the continuity of the blood–brain barrier and thus disrupts the perivascular clearance of Aβ [59].
It should be noted that the accumulation of amyloid beta begins in other parts of the brain. On the basis of the research of Palmqvist et al. [60], it is known that the accumulation of Aβ fibrils begins in certain regions of the brain, before they can be found throughout the neocortex, and before neurodegeneration is present. The researchers described that the early stages of amyloid deposition take place in the precuneus, posterior cingulate cortex, and orbitofrontal cortex. When examining subjects with even earlier signs of Aβ accumulation (CSF (cerebrospinal fluid)−/PET (positron emission tomography)− subjects who converted to CSF+/PET− within 2 years), a significantly increased Aβ fibril accumulation rate was again seen in the medial orbitofrontal and posterior cingulate cortex compared with stable CSF−/PET− subjects [60]. Braak [61] described the tau pathology progression from locus coeruleus through the transentorhinal region to cortical areas. These data suggest that tauopathy associated with sporadic Alzheimer disease may begin earlier than previously thought and possibly in the lower brainstem rather than in the transentorhinal region [61].
In recent years, scientific works examining the influence of peripheral processes on the pathomechanism of nervous system diseases have gained great value. In AD, it refers to the influence of the immune system on the onset of the disorder, which is the inflammatory basis of the disease. Thus, according to the literature, immunological mechanisms are responsible for the occurrence of dementia-like disorders [62]. The inflammatory reaction within the central nervous system is mediated by microglial cells. The activated microglia produce pro-inflammatory cytokines such as TNF-α (tumor necrosis factor-α), IL-1 (interleukin-1), IL-6 (interleukin-6), and chemokines. The above mediators of the inflammatory reaction initiate the process of chronic inflammation within the nervous system, leading to the death of neurons and the intensification of neurodegenerative changes [63,64,65]. The relationship of Aβ with the inflammatory theory is justified because senile plaques intensify the production of free radicals and initiate the cascade of inflammatory processes [66]. This is due to the presence of microglia near senile plaques and neurons containing neurofibrillary tangles, which, when mobilized, constitute an immunological line of defense [67,68]. Mobilized microglia can show a twofold phenotype depending on the environment. The protective function of microglia becomes apparent when Aβ fibers or neuronal debris are removed or when it is involved in synaptic remodeling and the release of growth factors. In contrast, the neurotoxic phenotype stimulates the activity of cytokines such as TNF-α and IL-1β (interleukin 1β). These immune system mediators cause tissue damage and disease progression [69].
The accumulation of Aβ starts in the preclinical phase of AD, leading to the progression of degenerative changes within ten years. In the prodromal phase of the disease, amyloid activity shows a potential for a plateau. At this time, mild cognitive impairment (MCI) becomes apparent [70]. In the prodromal phase of the disease, hyperphosphorylation of the tau protein occurs alongside its aggregation into neurofibrillary tangles. This response contributes to the direct neuronal dysfunction and contributes to the progression of dementia and progressive dementia [71].
RA is defined as a degenerative disease of the skeletal system. The disorder is characterized by the presence of a systemic inflammatory reaction that affects articular cartilage and bones [72]. Normally, the disease process involves immune cells that cascade to engage subsequent cells and mediators. Involvement of the immune system is associated with the activation of appropriate cells, resulting in the release of matrix metalloproteinases (MMPs) and inflammatory cytokines [73]. Ultimately, the ongoing processes result in the occurrence of painful joint swelling and impairment of their functions [74].
The factors that predispose to RA are not clearly explained. It has been suggested that the disease is related to genetic and exogenous factors. The first cells involved in the pathogenesis of RA are CD4+ lymphocytes. These cells recognize antigens in the synovial tissue and stimulate monocytes, macrophages, and synovial fibroblasts. The above-mentioned cells secrete metalloproteinases that are involved in the erosion of cartilage and bone. In addition to their degradative activity, these immune cells are involved in the production of interleukin (IL)-1, IL-6, and TNF-α, which are responsible for the key inflammatory response in RA. The entire cascade process eventually leads to synovitis, which becomes thickened and enlarged [75].
Appropriate biomarkers appear in the serum in RA patients, such as rheumatoid factor (RF), anti-cyclic citrullinated peptide (anti-CCP), C-reactive protein (CRP), amyloid A protein, and calgranulin [76]. The presence of amyloid structures is of particular interest, since the amyloidosis of the light chains of transthyretin and immunoglobulins contributes to amyloid deposition in soft tissues. The accumulation process is, in effect, a direct cause of orthopedic diseases [77]. One of the studies indicated the presence of amyloid deposits detected during orthopedic procedures, especially in people over 70 years of age [78]. Moreover, a study by Donelly et al. [79] indicates that 10% of men over 50 and women over 60 after biopsy have tendon sheath positive markers of amyloid tissue [79].
Protein amyloid A (SAA, serum amyloid A), which is produced by liver cells, deserves additional attention. Hepatocytes that are stimulated by the presence of pro-inflammatory cytokines (including TNF-α, IL-1, and IL-6) produce SAA in increased amounts. One study determined that the overproduction of SAA can occur in a number of diseases related to inflammatory pathology. Such diseases include long-term inflammation, including chronic infections, such as tuberculosis, osteomyelitis and rheumatoid arthritis, inflammatory bowel disease, hereditary diseases, and hematological and solid neoplasms [80]. It is worth noting that the overproduction of pro-inflammatory cytokines promotes increased angiogenesis, the processes of connective-tissue binder degradation in RA, and increased amyloid activity, which characterize cytokines as pleiotropic mediators [81].

3. The Activity of the Immune System in AD and RA

Chronic systemic peripheral inflammation influences the neurodegenerative processes that are characteristic of AD. The activity of inflammatory cytokines such as TNF-α, IL-6, IL-1β, transforming growth factor beta (TGF-β), IL-12, and IL-18 is clearly noticeable in AD patients compared to healthy controls [82]. The above-mentioned cytokines and their influence are being studied both in the pathogenesis of AD and in RA, because the over-reactivity of the immune system is a common feature of these disorders.
When determining the positive correlation between AD and RA, it should be noted that the incidence of AD is much higher in RA patients than in healthy people [83]. In a separate study, cognitive decline was observed later in life in people struggling with arthritic diseases, especially RA [84]. Work on the effects of systemic inflammation has been studied through the activity of anti-inflammatory drugs. Methotrexate and non-steroidal anti-inflammatory drugs (NSAIDs), which are common pharmaceuticals used in RA, reduce the risk of AD-related dementia, especially when these drugs are administered early in the disease [85,86].
It is known that overactivity of the immune system is the bridge between these diseases; however, further research is needed to determine the exact correlation between RA and AD. A common feature between RA and AD is the dysregulation of cell cycle suppression genes, which in turn contributes to the incidence of systemic inflammation. Studies show that inflammatory changes affect the appearance of pathological changes in both disorders, and cell cycle modifications are significantly age-regulated [87].
In the context of Alzheimer’s disease, it is claimed that systemic inflammation influences the appearance of neurodegenerative changes [82]. Activation of the immune system contributes to the development of dysfunction in the central nervous system. Imaging tests show a reduced brain volume and pathological changes in white matter [88]. Systemic inflammation also affects the limited distribution of blood in the blood vessels, which contributes to brain dysfunction due to restricted oxygen supply. Therefore, it can be assumed that changes in circulating blood flow increase the risk of developing dementia [89]. In patients with depression and AD, raised levels of pro-inflammatory cytokines, acute phase proteins (APPs), interferon gamma (IFN-γ), interleukin 1 (IL-1), IL-6, and TNF-α were observed [90].
Based on the literature, it is suggested that NSAIDs (non-steroidal anti-inflammatory drugs) inhibit the formation of fibrous Aβ by stimulating α-secretase. As a result of such activity, APP is transformed by a non-amloidogenic path, which leads to a reduction in the formation of amyloidogenic forms [91]. The legitimacy of the use of NSAIDs in AD has been determined in several studies. A meta-analysis was performed, which included 16 cohort studies, conducted between 1995 and 2016 and involving 236,022 participants [92]. It showed that the current or previous use of NSAIDs was significantly associated with a reduced risk of AD compared to those who did not use NSAIDs. The study also shows correlations related to where the patients live (eight studies looked at populations from North America, six studies were from European countries, and two studies focused on Asian populations). Noteworthy, in this study, the researchers have studied the influence of the environmental factor. The authors showed that NSAID exposure was associated with a reduced risk of AD in population-based studies. Such correlations were not noted in the study on the importance of the environmental factors. The study pointed to a significant impact of NSAID exposure in Europe. The borderline effect of NSAID exposure was observed in studies conducted in North America, and NSAID exposure was not associated with the reduced risk of AD in studies conducted in Asia. Current evidence suggests an effect of NSAIDs on a reduced risk of AD especially in large population-based cohort studies. The authors suggest that there is a need for a much larger study to justify the dose of the drug and the duration of NSAID exposure [92]. Among the similar meta-analyses cited by the authors of the study is a meta-analysis performed by Etminan et al. The authors of this study report a positive correlation between the use of NSAIDs and the onset of AD. Despite the fact that it includes nine observational studies, it is necessary to point out some limitations related, among other things, to the use of a specific NSAID and the reduction of the risk of AD [93].
One study evaluated the safety and efficacy of low doses of naproxen in preventing the progression of pre-asymptomatic AD among at-risk individuals with cognitive impairment. This two-year study involved 195 elderly people (mean age 63 years) who had a positive family history of AD. Patients underwent an imaging examination to exclude cognitive impairment. The researchers have divided the study participants into two groups. One of them received a placebo, while the second group was administrated with naproxen sodium at a dose of 220 mg twice daily. Multimodal, neurosensory, cognitive imaging, and evaluation of cerebrospinal fluid biomarkers was performed at the start of the study, 3, 12, and 24 months. People who took naproxen sodium experienced side effects. Naproxen did not reduce the progression of AD. Moreover, secondary analyses did not show any significant effect of treatment on individual biomarker indicators of cerebrospinal fluid nor cognitive or neurosensory progressive pre-symptomatic AD [94]. The effectiveness of NSAIDs in AD was studied on the example of naproxen and celecoxib. This was a large, multi-year randomized, placebo-controlled clinical trial. The researchers concluded that not every drug can delay the onset of AD in adults with a family history of dementia [95].
The implementation of appropriate treatment for the patient often depends on their economic status. Particularly noteworthy is the study conducted by Moilna et al [96]. The aim of the study was to determine the relationship between socioeconomic status (SES) and delays in treatment with disease-modifying antirheumatic drugs (DMARDs) in RA patients. A total of 1209 RA patients were recruited for the study, of whom 1159 received DMARD treatment. Researchers have assessed SES based on education, occupation, and income. After a preliminary analysis, they divided the patients into terciles. The researchers assessed the severity of the disease, which was determined by the rate of disease activity in 28 joints. They determined joint damage based on manual radiographs according to the Sharp scale and physical disability on the basis of a modified Health Assessment Questionnaire (M-HAQ). The researchers conclude that patients with lower SES experience longer delays in starting DMARD treatment. Additionally, patients who experience a greater delay in initiating DMARD treatment do experience a clinically significant increase in joint damage [96].
The relationship between AD and RA was investigated by Chou et al. [97]. The authors of the study analyzed medical and pharmaceutical claims. The analysis took into account data from adults who were commercially insured between 2000 and 2007. After analyzing the data, 325 people showed symptoms of AD and were diagnosed with RA. The study took into account the presence of concomitant diseases in patients such as coronary artery disease, diabetes, hyperlipidemia, hypertension, and peripheral vascular disease. It was determined that the overall incidence of AD among people with RA was 0.79%, while the incidence of AD among those without RA was 0.11%. In addition, it was shown that chronic conditions such as coronary artery disease, diabetes, and vascular disease significantly increased the relative risk of AD among patients with RA. The authors of the study additionally analyzed patients suffering from RA. For this purpose, the prevalence of AD among patients with RA after exposure to various RA therapies was determined. The drugs used in RA were methotrexate, prednisone, sulfasalazine, three anti-TNF drugs (adalimumab, etanercept, and infliximab) and an anti-CD20 drug (rituximab). The researchers concluded that anti-TNF therapy in RA lowered the risk of AD among these patients. Although the researchers have shown that inflammation is probably important in the pathology of AD, more research should be carried out in this direction [97]. Ungprasert et al. [98] drew similar conclusions and indicated an increased risk of dementia in patients suffering from RA [98]. The authors of the study suggest that the correlation between AD and RA should be linked to blood vessel disease. In the above monograph, the authors indicate that atherosclerosis caused by RA is a possible explanation for such a correlation. Atherosclerotic cardiovascular disease is a well-known risk factor for dementia [99]. In addition, the predisposition to the occurrence of atherosclerosis in patients with RA results from the negative impact of intract cytokines on vascular endothelial cells [100]. It is worth noting that the occurrence of cardiovascular diseases is more common among patients with autoimmune inflammatory diseases [101].
Mendel’s randomized controlled trials assessing the potential relationship between AD and RA are of particular interest. The authors of the study used publicly available collections of studies consisting of three-stage trans-ethnic, European-specific, and Asian-specific genome-wide association study (GWAS) meta-analyses, which evaluated 10 million single nucleotide polymorphisms (SNPs) in 29,880 RA cases and 73,758 controls assigned as exposures. The researchers used Mendelian randomization (MR), which uses genetic variants as instrumental variables. Its use assesses the relationship between the risk factor or exposure and whether the outcome is consistent with the causal effect. The analysis is dedicated to studies in which it is difficult to measure exposure and outcome in the same group of people. The aim of the study was to determine whether RA is causally related to AD using MR analysis of two trials. In this study, Bae and Lee [102] indicated a significant causal relationship between RA and AD. The researchers suggest that the study has some limitations. First of all, genetic variants have little effect on RA, and there is a need to experiment on a large population of people. In addition, the work was carried out on the European population. This is an inaccurate fact, as the relationship between AD and RA may be related to ethnicity. Therefore, the authors suggest that there is a need to conduct a study on a diverse study group. [102].
Other researchers have noted that drug therapy with anti-rheumatic drugs reduces the risk of dementia [103]. Researchers observed a reduced risk of dementia in cDMARD users compared with those not taking such medications. In addition, after the use of drugs, there was a reduction in the risk of dementia in those using cDMARD. The strongest effect was achieved with the use of MTX (metotrexat). The risk of dementia was 0.5% compared to 1.6% at 5 years and 1.5% at 3.0% at 15 years. The results obtained by the researchers are consistent with the estimated incidence of dementia in the UK in 2013, which was 1.3%. Despite the results, the authors of the study point to some limitations. The study involved people who suffered from comorbidities, including cardiovascular disease, myocardial infarction, congeal heart failure, peripheral vascular disease, interstitial lung disease, anemia, and osteoporosis. Before starting RA therapy, patients took antihypertensive drugs, analgesics, statins, or proton pump inhibitors. [103].
One of the newest studies conducted in 2020 confirms the above-mentioned findings and additionally indicates the potential protective effect of anti-TNF drugs in patients with RA [104]. This study was conducted in 56 million adults in the United States and showed a positive correlation between AD and RA and a lower risk of AD in patients taking etanaercept, adalimumab, or infliximab. The authors of the study indicate that the above-mentioned biological drugs have poor distribution to the brain, and by acting systemically, they prevent TNF-α from entering the brain and thus prevent the risk of dementia. Thanks to this research, it is estimated that treatment of RA with TNF inhibitors can help prevent about 4.0% of AD cases (0.21 million) [104].
The correlations between AD and RA were investigated by McGeer and others [105]. Seventeen epidemiological studies from nine countries were used for the review. All these studies concluded that arthritis is a predisposing factor to the appearance of AD. The analysis also included studies that concluded about the positive importance of anti-inflammatory drug therapy in AD. Using statistical meta-analysis methods, the researchers estimated the risk of developing AD in RA patients and anti-inflammatories compared to the general population. Seven case-control studies with arthritis as a risk factor yielded a total odds ratio of 0.556 (p < 0001), while four case-control studies with steroids yielded an odds ratio of 0.656 (p = 0.049). Three case-control studies with NSAIDs gave an odds ratio of 0.496 (p = 0.0002). When NSAIDs and steroids were combined into one category of anti-inflammatory drugs, the odds ratio was 0.556 (p < 0.0001). Therefore, the study confirmed the validity of the hypothesis of the protective effect of anti-inflammatory drugs in AD. Still, the researchers suggest that there is a need for clinical trials to thoroughly verify this hypothesis [105].
In turn, Kao et al. [106], in a case-control study on 2271 patients with AD and 6813 patients in the control group without AD, indicated an inverse relationship between previous AD and RA. The mean age of the study group was 76.5 ± 7.9 years, while the mean age of the control group was 76.5 ± 8.3 years. After data analysis, significant differences were identified in geographic region (p < 0.001), level of urbanization (p = 0.002), hypertension (p < 0.001), diabetes (p = 0.027), hyperlipidemia (p < 0.001), stroke (p < 0.001), and coronary heart disease (p < 0.001) between cases and the control group after matching the sex, age group, and year of the index date. There was no significant difference in monthly income between the cases and the control group. The study showed that the earlier onset of RA was negatively associated with AD even in people suffering from comorbidities. Despite this correlation, the authors note that the study does not take into account several factors. First, the database used by the researchers does not contain information on the presence of inflammatory markers. What is more, the database lacked family interviews and indications of possible genetic predisposition. The researchers note that such factors can affect cognitive function and falsify the result of the study. The database also has no information on biochemical tests or medical imaging. In addition, the majority of the Chinese population was recruited for the study, which does not allow generalizing the results and relating them to the general public [106].
Similar conclusions were reached by Policicchio et al. [107], who determined that RA was associated with a lower incidence of AD. Researchers have shown that there is no correlation between the use of NSAIDs and AD. The indicated meta-analysis included eight case-control studies and two population studies. The causal relationship was determined on the basis of the MR discussed earlier. Previous analysis of the literature showed that RA was associated with a lower incidence of AD. The authors of the study question this hypothesis because MR analysis showed no correlation between AD and RA. Although there is epidemiological evidence to support the validity of the claim, the authors recognize that there are no causal relationships between these disease entities. Researchers suggest that a large impact on the validity of the thesis is influenced by factors interfering with research such as selection error or differential diagnosis of RA [107].
Therefore, the literature indicates that inflammation is a common feature of both RA and dementia, and this is confirmed by common inflammatory biomarkers found in both diseases (e.g., interleukin-6, interleukin-12, C-reactive protein, penxine 3, endotheline-1, resistin, and receptors for the end products of advanced glycation) [108]. In addition, by examining the broad relationships between the occurrence of AD, significant relationships between the occurrence of AD and the over-reactivity of the immune system were indicated. These studies have indicated the genetic overlap between AD and immune-mediated diseases [109]. Despite the presence of common markers of inflammation, this mention should be treated with great caution. Perhaps the presence of markers of inflammation does not indicate a cause-and-effect correlation between these disorders. Perhaps AD and RA are governed by other mechanisms of the formation of an inflammatory reaction, and the only feature that unites them is the presence of indicated markers. This topic requires further work to explain the possible mechanisms of the formation of an immune cascade both in AD in RA and in both disorders.

4. The Role of the Blood–Brain Barrier in AD and RA

The effects of systemic inflammation (infection, pathological conditions, sepsis) affect tissues and organs; however, it should be noted that the blood–brain barrier (BBB) becomes the mediating factor between RA and AD. The hyperactivity of the immune system, expressed by an increased concentration of inflammatory mediators, negatively affects the structure and permeability of the barrier [110]. Based on scientific studies, barrier permeability has been shown to be altered in RA patients. Barrier dysfunction is also associated with neurodegenerative diseases, including AD [111]. Among the mediators of the immune system, inflammatory cytokines deserve special attention, as they show the ability to easily exceed the BBB [112]. The barrier is crossed with the use of various structures, that is: by passing through the periventricular organs [113], by stimulating the vagus nerve [114], and by direct binding to the endothelium, which results in the opening of tight junctions, penetration of the cytokine, and its activity within the brain tissue [82].
The relationship between RA and AD has been studied in rats with collagen-induced arthritis (CIA) [115]. This model is characterized by a clinical picture of RA. In addition to immune system activity in the disease, the integrity of the BBB was also studied by measuring the expression of Aβ transport proteins in the blood vessels of the brain. The study showed an increase in astrogliosis, the level of peripheral and brain cytokines, and activation of microglia in the brain in CIA rats compared to the control group. It should be mentioned here that astrogliosis is observed at the time of neuronal degradation in response to injury, infection, or the presence of a neurodegenerative disease. During the study, barrier permeability was measured using sodium fluorescein and found that it was significantly increased in CIA rats. The vascular changes observed were associated with increased expression of matrix metalloproteinase and decreased expression of tight junction proteins. Occludin is one such protein. Increased expression of the RAGE receptor in the hippocampus, which is involved in the influx of Aβ from the blood to the brain, was also observed during the study. To visualize the transport of Aβ via the BBB in CIA rats, animals were administered Aβ42 intravenously. It was shown that the concentrations of Aβ42 in the cortex and in the whole brain were compared between the CIA rats and the control group. However, it was shown that in the hippocampus, the Aβ level of the CIA rats was approximately 1.8 times higher than that of the control. This relationship indicates an increased influx of Aβ from the blood to the hippocampus of CIA rats [115].
It was also suggested that RA patients are a group predisposed to disturbances of homeostasis within the blood vessels, heart, and cerebral vessels. The studies were conducted in CIA rodents and investigated endothelial dysfunction induced by chronic inflammation in RA. The expression of strict proteins was determined by immunoblotting and occludin immunofluorescence. Reduced expression of the protein involved in the formation of tight junctions (occludins) has been reported. The study concludes that barrier integrity is impaired in the pathophysiology of RA [116].

5. Therapeutic Strategies Targeting to Aggregates or Oligomers, Which Are the Most Dangerous Amyloid Forms

The amount of discovered amyloidogenic proteins continues to increase, which makes it difficult to diagnose patients suffering from amyloidosis. In the first stages of treatment, it becomes necessary to identify the amyloidogenic protein in order to make a proper diagnosis and implement appropriate treatment. Correct diagnosis is based on the use of histochemical tests [117], biochemical tests [118], genetic analyses [119,120], and functional imaging studies. At the present stage, the most effective approach to the treatment of systemic amyloidosis is to stop or reduce the synthesis of the amyloid precursor [121]. It is indicated that disruption of the expression of the corresponding gene using antisense oligonucleotides and small interference RNA can significantly reduce the amount of amyloidogenic precursor. Such a solution had a positive effect in the form of reduction of the synthesis of amyloidogenic light chains [122]. Despite the promising effect, the application of the present method in clinical trials faces some difficulties, including modulation of the intracellular concentration of interfering RNA [123].
First innovation in the treatment of amyloidosis was achieved by inhibiting proteases, which are responsible for the generation of amyloidogenic fragments. It is estimated that this strategy would also find application in the treatment of Alzheimer’s disease. This is because the therapeutic target in AD is to inhibit β- and γ-secretases that produce the amyloidogenic peptide [120]. In addition, the new clinical approach points to the positive importance of lipid-lowering drugs from the statin group, which can prevent the progression of AD through a mechanism related to the modulation of the ability of secretases to cleave the amyloid precursor [124]. In addition, anti-inflammatory drugs used in the treatment of AD may have a direct influence on secretase activity [125]. The use of secretase inhibitors in therapy has some limitations. For example, one of the γ-secretase inhibitors, R-flurbiprofen (Flurizan TM), had a limited effect on patients with mild AD and had no effect on patients with moderate AD [126]. In addition, a significant limitation of such treatment is the fact that γ-secretases take part in many physiological functions in humans. Therefore, it is an enzyme not specific to APP, because it participates in the regulation of cell growth and in the transformation of proteins. This insufficiency contributes to the toxic effects in humans [127]. This adverse effect creates harmful defects in memory processing, myelination, and motor coordination [128]. The limitation of using secretase inhibitors is also the BBB, because the endothelial cells of this barrier restrict the diffusion of large or hydrophilic molecules. The secretase inhibitor needs to cross the BBB and neuronal membranes, because the process of modification of APP runs in the endosomes of neurons within the brain. It is also known that the largest molecular size that can cross the BBB is approximately 550 DA, so in the development of new drugs, the size of the molecule is important [126]. It is also considered that for AD therapies, it is necessary to treat individuals in the “preclinical” (presymptomatic) stage [129].
Progress in science was brought by the recently described therapy with the use of mesenchymal stem cells (MSC) [130]. These cells are isolated from bone marrow, adipose tissue, and the umbilical cord [131]. They are pluripotent in nature and have the ability to transform into any type of cell: osteoblasts, chondrocytes, and adipocytes. MSCs exhibit a wide range of activities and influence the function and activity of the immune system by modulating the proliferation of key mediators of innate and acquired immunity. The injection of MSC into the brain in AD and intra-articular in RA is beneficial because inflammation is reduced, cell proliferation is stimulated, and behavioral indicators are improved [132].
Currently, great hopes are placed on aducanumab, which in amyloid PET imaging limited the pathology of amyloid and was effective at reducing dementia in patients in phase III clinical trials [133]. If subsequent assumptions are met, aducanumab may become the first drug modifying the course of the disease, thus confirming the validity of the anti-amyloid strategy [133]. Aducanumab therapy is associated with adverse reactions known as Amyloid Related Imaging Abnormalities (ARIA). During therapy, in a mice model, bleeding and swelling of the brain were observed, which consequently causes headaches, confusion, or convulsions. For comparison, Xiong et al. [134] in 2021 did not notice these adverse effects after therapy with a new antibody targeting APOE. This antibody is determined as HAE-4. In studies in mice treated with HAE-4, amyloid plaques were removed from brain tissue and blood vessels without increasing the risk of bleeding into the brain. What is more, the researchers pointed out that the brain’s blood vessels showed a greater ability to expand and narrow on demand, and inflammation within the brain was significantly reduced [134]. A monoclonal antibody targeting amyloid is also a candidate for treatment—it is BAN240, developed by Eisai and Biogen [135]. However, it is highly probable that when used alone, these agents will not be able to limit or cure such a complex pathology of amyloidosis. There is great hope in the use of combination therapy [135].

6. Conclusions

Systemic inflammation affects the appearance of neurodegenerative changes. The similarity of AD and RA pathomechanisms is visible in the activity of the immune system, which, under the influence of appropriate factors, produces inflammatory biomarkers (e.g., interleukin-6, interleukin-12, C-reactive protein, penxine 3, endotheline-1, resistin, and receptors for the end products of advanced glycation). Cytokines mediate many processes in the body and affect the tightness of the blood–brain barrier, including its integrity, and strictly reduce the expression of occluding-forming junctions. In addition, AD and RA are disorders associated with the pathology of amyloid. In AD, Aβ plaques increase the susceptibility of neurons to excitotoxicity, loss of synaptic protein, and cholinergic transmission, while in RA, cytokine-stimulated amyloid contributes to the degradation of the bone–joint bond. The incidence of AD is much higher in RA patients than in healthy people. The existence of a complex network of connections is explained by demonstrating the correlation between the nervous, skeletal, and immune systems as well as the aging mechanisms of the organism. Despite the presence of common factors, this mention should be treated with great caution. The presence of inflammatory markers may not clearly indicate a cause-and-effect relationship between these disorders. It is possible that AD and RA have different inflammatory mechanisms and the only trait that they have in common is the presence of the markers indicated. This topic requires further work to elucidate possible mechanisms of the immune cascade in both AD in RA and in both diseases simultaneously. Currently, the treatment of systemic amyloidoses involves stopping or reducing the amyloid precursor synthesis, inter alia, by disrupting the expression of the relevant gene using antisense oligonucleotides and small interfering RNA. Innovation in the treatment of amyloidosis has also been achieved by inhibiting proteases, which may also be effective in the treatment of AD. Innovative therapies using mesenchymal stem cells and monoclonal antibodies targeting amyloid are of particular importance. As these treatments are unsatisfactory and have numerous side effects, the search for therapeutic strategies with greater efficacy and a higher safety profile is still ongoing.

Author Contributions

Writing: P.T. and M.H., review and editing: M.H. and J.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siddiqi, O.K.; Ruberg, F.L. Cardiac amyloidosis: An update on pathophysiology, diagnosis, and treatment. Trends Cardiovasc. Med. 2018, 28, 10–21. [Google Scholar] [CrossRef] [PubMed]
  2. Perutz, M.F. Amyloid fibrils: Mutations make enzyme polymerize. Nature. 1997, 385, 773–775. [Google Scholar] [CrossRef]
  3. Weatherall, D.J.; Ledingham, J.G.G.; Warell, D.A. Amyloidosi. In The Oxford Textbook of Medicine, 3rd ed.; Oxford University Press: Oxford, UK, 1996; pp. 1512–1524. [Google Scholar]
  4. Pauling, L.; Corey, R. Configuration of polypeptide chains with favoured orientation around single bonds: Two new pleated sheets. Proc. Natl. Acad. Sci. USA 1951, 37, 729–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sawaya, M.R.; Sambashivan, S.; Nelson, R.; Ivanova, M.I.; Sievers, S.A.; Apostol, M.I.; Thompson, M.J.; Balbirnie, M.; Wiltzius, J.J.; McFarlane, H.T.; et al. Atomic structures of amyloid cross-beta spines reveal varied steric zippers. Nature 2007, 447, 453–457. [Google Scholar] [CrossRef]
  6. Sunde, M.; Serpell, L.C.; Bartlam, M.; Fraser, P.E.; Pepys, M.B.; Blake, C.C. Common core structure of amyloid fibrils by synchrotron X-ray diffraction. J. Mol. Biol. 1997, 273, 729–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Wininger, A.E.; Phelps, B.M.; Le, J.T.; Harris, J.D.; Trachtenberg, B.H.; Liberman, S.R. Musculoskeletal pathology as an early warning sign of systemic amyloidosis: A systematic review of amyloid deposition and orthopedic surgery. BMC Musculoskelet. Disorders 2021, 22, 51. [Google Scholar] [CrossRef] [PubMed]
  8. Reitz, C.; Brayne, C.; Mayeux, R.; Kelly, J.W.; Maurer, M.S. Epidemiology of Alzheimer disease. Nat. Rev. Neurol. 2011, 7, 137–152. [Google Scholar] [CrossRef] [PubMed]
  9. Sperry, B.W.; Vranian, M.N.; Hachamovitch, R.; Joshi, H.; Ikram, A.; Phelan, D.; Hanna, M. Subtype-specific interactions and prognosis in cardiac amyloidosis. J. Am. Heart Assoc. 2016, 5, 002877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Saraiva, M.J. Transthyretin amyloidosis: A tale of weak interactions. FEBS Lett. 2001, 498, 201–203. [Google Scholar] [CrossRef] [Green Version]
  11. Kozak, S.; Ulbrich, K.; Migacz, M.; Szydło, K.; Mizia-Stec, K.; Holecki, M. Cardiac Amyloidosis-Challenging Diagnosis and Unclear Clinical Picture. Medicina 2021, 57, 450. [Google Scholar] [CrossRef]
  12. Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science. 2002, 297, 353–356. [Google Scholar] [CrossRef] [Green Version]
  13. Hawkins, P.N.; Myers, M.J.; Lavender, J.P.; Pepys, M.B. Diagnostic radionuclide imaging of amyloid: Biological targeting by circulating human serum amyloid P component. Lancet 1988, 1, 1413–1418. [Google Scholar] [CrossRef]
  14. Tennent, G.A.; Lovat, L.B.; Pepys, M.B. Serum amyloid P component prevents proteolysis of the amyloid fibrils of Alzheimer disease and systemic amyloidosis. Proc. Natl. Acad. Sci. USA 1995, 92, 4299–4303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Snow, A.D.; Kisilevsky, R.; Stephens, C.; Anastassiades, T. Characterization of tissue and plasma glycosaminoglycans during experimental AA amyloidosis and acute inflammation: Qualitative and quantitative analysis. Lab. Investig. J. Tech. Methods Pathol. 1987, 56, 665–675. [Google Scholar]
  16. Stevens, F.J.; Kisilevsky, R. Immunoglobulin light chains, glycosaminoglycans, and amyloid. Cell Mol. Life Sci. 2000, 57, 441–449. [Google Scholar] [CrossRef]
  17. Sipe, J.D.; Benson, M.D.; Buxbaum, J.N.; Ikeda, S.I.; Merlini, G.; Saraiva, M.J.; Westermark, P. Nomenclature 2014: Amyloid fibril proteins and clinical classification of the amyloidosis. Amyloid 2014, 21, 221–224. [Google Scholar] [CrossRef] [PubMed]
  18. Bellotti, V.; Mangione, P.; Merlini, G. Review: Immunoglobulin light chain amyloidosis--the archetype of structural and pathogenic variability. J Struct Biol. 2000, 130, 280–289. [Google Scholar] [CrossRef]
  19. Merlini, G.; Westermark, P. The systemic amyloidoses: Clearer understanding of the molecular mechanisms offers hope for more effective therapies. J. Intern. Med. 2004, 255, 159–178. [Google Scholar] [CrossRef]
  20. Buxbaum, J.N.; Tagoe, C.E. The genetics of the amyloidoses. Annu. Rev. Med. 2000, 51, 543–569. [Google Scholar] [CrossRef]
  21. Westermark, P.; Picken, M.; Herrera, G.; Dogan, A. Localized amyloidoses and amyloidoses associated with aging outside the central nervous system. Amyloid Relat. Disord. Curr. Clin. Pathol. 2020, 143, 322–334. [Google Scholar]
  22. McLaurin, J.; Yang, D.; Yip, C.M.; Fraser, P.E. Modulating factors in amyloid-beta fibril formation. J. Struct. Biol. 2000, 130, 259–270. [Google Scholar] [CrossRef]
  23. Benson, M.D.; Buxbaum, J.N.; Eisenberg, D.S.; Merlini, G.; Saraiva, M.J.M.; Sekijima, Y.; Sipe, J.D.; Westermark, P. Amyloid nomenclature 2020: Update and recommendations by the International Society of Amyloidosis (ISA) nomenclature committee. Amyloid 2020, 27, 217–222. [Google Scholar] [CrossRef]
  24. Sipe, J.D.; Benson, M.D.; Buxbaum, J.N.; Ikeda, S.; Merlini, G.; Saraiva, M.J.M.; Westermark, P. Amyloid fibril protein nomenclature: 2012 recommendations from the Nomenclature Committee of the International Society of Amyloidosis. Amyloid 2012, 19, 167–170. [Google Scholar] [CrossRef]
  25. Ruberg, F.L.; Grogan, M.; Hanna, J.W.; Kelly, M.S. Maurer Transthyretin amyloid cardiomyopathy: JACC state-of-the-art review. J. Am. Coll. Cardiol. 2019, 73, 2872–2891. [Google Scholar] [CrossRef]
  26. Prokaeva, T.; Spencer, B.; Kaut, M.; Ozonoff, A.; Doros, G.; Connors, L.H.; Skinner, M.; Seldin, C.D. Soft tissue, joint, and bone manifestations of AL amyloidosis: Clinical presentation, molecular features, and survival. Arthritis Rheum. 2007, 56, 3858–3868. [Google Scholar] [CrossRef]
  27. Grogan, M.; Dispenzieri, A.; Gertz, M.A. Light-chain cardiac amyloidosis: Strategies to promote early diagnosis and cardiac response. Heart 2017, 103, 1065–1072. [Google Scholar] [CrossRef] [PubMed]
  28. Tasaki, M.; Lavatelli, F.; Obici, L.; Obayashi, K.; Miyamoto, T.; Merlini, G.; Palladini, G.; Ando, Y.; Ueda, M. Age-related amyloidosis outside the brain: A state-of-the-art review. Ageing Res. Rev. 2021, 70, 181–195. [Google Scholar] [CrossRef] [PubMed]
  29. Maurer, M.S.; Elliott, P.; Comenzo, R.; Semigran, M.; Rappezi, C. Addressing common questions encountered in the diagnosis and Management of Cardiac Amyloidosis. Circulation 2017, 135, 1357–1377. [Google Scholar] [CrossRef] [Green Version]
  30. Falk, R.H.; Comenzo, R.L.; Skinner, M. The systemic amyloidoses. N. Engl. J. Med. 1997, 337, 898–909. [Google Scholar] [CrossRef] [PubMed]
  31. Kyle, R.A.; Gertz, M.A. Primary systemic amyloidosis: Clinical and laboratory features in 474 cases. Semin. Hematol. 1995, 32, 45–59. [Google Scholar] [PubMed]
  32. Lachmann, H.J.; Goodman, H.J.; Gilbertson, J.A.; Gallimore, J.R.; Sabin, C.A.; Gallimore, J.D.; Hawkins, P.N. Natural history and outcome in systemic AA amyloidosis. N. Engl. J. Med. 2007, 356, 2361–2371. [Google Scholar] [CrossRef] [Green Version]
  33. Dungu, J.N.; Anderson, L.J.; Whelan, C.J.; Hawkins, P.N. Cardiac transthyretin amyloidosis. Heart 2012, 98, 1546–1554. [Google Scholar] [CrossRef]
  34. Drüeke, T.B.; Massy, Z.A. Beta2-microglobulin. Semin. Dial. 2009, 22, 378–380. [Google Scholar] [CrossRef] [PubMed]
  35. Patterson, C.; World Report on Alzheimer’s Disease 2018. ADI, London 1–48. Available online: www.alz.co.uk/research/WorldAlzheimerReport2018.pdfGoogleScholar (accessed on 8 November 2018).
  36. Long, J.M.; Holtzman, D.M. Alzheimer disease: An update on pathobiology and treatment strategies. Cell 2019, 179, 312–339. [Google Scholar] [CrossRef] [PubMed]
  37. Póżyński, S. Classification of Mental and Behavioral Disorders in ICD-10. Descriptions and Recognition; UWM “Vesalius” Institute of Psychiatry and Neurology Krakow: Warsaw, Poland, 2000. [Google Scholar]
  38. American Psychiatric Association. American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; American Psychiatric Association Publishing: Washington, DC, USA, 2013. [Google Scholar]
  39. Goedert, M.; Spillantini, M.G. A century of Alzheimer’s disease. Science 2006, 314, 777–781. [Google Scholar] [CrossRef] [Green Version]
  40. Querfurth, H.W.; LaFerla, F.M. Alzheimer’s Disease. N. Engl. J. Med. 2010, 362, 329–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Kamat, P.K.; Tota, S.; Shukla, R.; Ali, S.; Najmi, A.K.; Nath, C. Mitochondrial dysfunction: A key event in okadaic acid (ICV) induced memory disorder and apoptotic cell death in the rat brain. Pharm. Biochem. Behav. 2012, 100, 311–319. [Google Scholar] [CrossRef]
  42. Kamat, P.K.; Rai, S.; Swarnkar, S.; Shukla, R.; Nath, C. Mechanism of synapse redox stress in okadaic acid (ICV) induced memory disorder: Role of NMDA receptor. Neurochem Int. 2014, 20, 10–15. [Google Scholar]
  43. Rai, S.; Kamat, P.K.; Nath, C.; Shukla, R. A study on neuroinflammation and NMDA receptor function in STZ (ICV) induced memory lord. J. Neuroimmunol. 2013, 254, 1–9. [Google Scholar] [CrossRef]
  44. Davies, P.; Maloney, A.J. Selective loss of central cholin- 806 ergic neurons in Alzheimer’s disease. Lancet. 1976, 2, 1403. [Google Scholar] [CrossRef]
  45. Kamat, P.K.; Kalani, A.; Rai, S.; Swarnkar, S.; Tota, S.; Nath, C.; Tyagi, N. Mechanism of oxidative stress and synapse dysfunction in the pathogenesis of Alzheimer’s disease: Therapeutic strategies. Mol. Neurobiol. 2016, 53, 648–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kamat, P.K.; Tota, S.; Saxena, G.; Shukla, R.; Nath, C. Okadaic acid (ICV) induced memory disorder in rats. Brain Res. 2010, 1309, 66–74. [Google Scholar] [CrossRef] [PubMed]
  47. Sciacca, M.F.; Kotler, S.A.; Brender, J.R.; Chen, J.; Lee, D.K.; Ramamoorthy, A. Two-step mechanism of membrane disruption by Abeta through membrane fragmentation and pore formation. Biophys. J. 2012, 103, 702–710. [Google Scholar] [CrossRef] [Green Version]
  48. Capone, R.; Quiroz, F.G.; Prangkio, P.; Saluja, I.; Sauer, A.M.; Bautista, M.R.; Turner, R.S.; Yang, J.; Mayer, M. Amyloid-beta-induced ion flux in artificial lipid bilayers and neuronal cells: Resolving a controversy. Neurotox. Res. 2009, 16, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Kuchibhotla, K.V.; Goldman, S.T.; Lattarulo, C.R.; Wu, H.Y.; Hyman, B.T.; Bacskai, B.J. Abeta plaques lead to incorrect regulation of calcium homeostasis in vivo, causing structural and functional disruption of neural networks. Neuron 2008, 59, 214–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Bowen, D.M.; Smith, C.B.; White, P.; Davison, A.N. Neurotransmitter-related enzymes and indices of hypoxia in senile dementia and other abiotrophie. Brain 1976, 99, 459–496. [Google Scholar] [CrossRef] [Green Version]
  51. Mesulam, M. A horseradish peroxidase method for the identification of the efferents of acetyl cholinesterase-containing neurons. J. Histochem. Cytochem. 1976, 24, 1281–1285. [Google Scholar] [CrossRef]
  52. Nordberg, A.; Winblad, B. Reduced number of [3H]nicotine and [3H]acetylcholine binding sites in the frontal cortex of Alzheimer brains. Neurosci. Lett. 1986, 72, 115–119. [Google Scholar] [CrossRef]
  53. Schroder, H.; Giacobini, E.; Struble, R.G.; Zilles, K.; Maelicke, A. Nicotinic cholinoceptive neurons of the frontal cortex are reduced in Alzheimer’s disease. Neurobiol. Aging 1991, 12, 259–262. [Google Scholar] [CrossRef]
  54. Mesulam, M.M. Cholinergic circuitry of the human nucleus basalis and its fate in Alzheimer’s disease. J. Comp. Neurol. 2013, 521, 4124–4144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Perry, E.K.; Perry, R.H.; Blessed, G.; Tomlinson, B.E. Necropsy evidence of central cholinergic deficits in senile dementia. Lancet 1977, 1, 189. [Google Scholar] [CrossRef]
  56. Ramos-Rodriguez, J.J.; Pacheco-Herrero, M.; Thyssen, D.; Murillo-Carretero, M.I.; Berrocoso, E.; Spires-Jones, T.L.; Garcia-Alloza, M. Rapid β-amyloid deposition and cognitive impairment after cholinergic denervation in APP/PS1 mice. J. Neuropath Exp. Neurol. 2013, 72, 272–285. [Google Scholar] [CrossRef] [PubMed]
  57. Field, R.H.; Gossen, A.; Cunningham, C. Prior pathology in the basal forebrain cholinergic system predisposes to inflammation-induced working memory deficits: Reconciling inflammatory and cholinergic hypotheses of delirium. J. Neurosci. 2012, 32, 6288–6294. [Google Scholar] [CrossRef] [PubMed]
  58. Hunter, J.M.; Kwan, J.; Malek-Ahmadi, M.; Maarouf, C.L.; Kokjohn, T.A.; Belden, C.; Bacskai, B.J.; Garcia- Alloza, M. Morphological and pathological evolution of the brain microcirculation in aging and Alzheimer’s disease. PLoS ONE 2012, 7, e36893. [Google Scholar] [CrossRef] [PubMed]
  59. Engelhardt, B.; Carare, R.O.; Bechmann, I.; Flugel, A.; Laman, J.D.; Weller, R.O. Vascular, glial, and lymphatic immune gateways of the central nervous system. Acta Neuropathol. 2016, 132, 317–338. [Google Scholar] [CrossRef] [Green Version]
  60. Palmqvist, S.; Schöll, M.; Strandberg, O.; Mattsson, N.; Stomrud, E.; Zetterberg, H.; Blennow, K.; Landau, S.; Jagust, W.; Hansson, O. Earliest accumulation of β-amyloid occurs within the default-mode network and concurrently affects brain connectivity. Nat. Commun. 2017, 8, 1214. [Google Scholar] [CrossRef] [Green Version]
  61. Braak, H.; Thal, D.R.; Ghebremedhin, E.; Del Tredici, K. Stages of the pathologic process in Alzheimer disease: Age categories from 1 to 100 years. J. Neuropathol. Exp. Neurol. 2011, 70, 960–969. [Google Scholar] [CrossRef] [PubMed]
  62. Halliday, G.; Robinson, S.R.; Shepherd, C.; Kril, J. Alzheimer’s Disease and Inflammation: A National and Country Therapeutic ReviewClin. Exp. Pharmacol. Physiol. 2000, 27, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Wyss-Coray, T. Inflammation in Alzheimer’s Disease: Driving Force, Witness, or Positive Response? Nat. Med. 2006, 12, 1005–1015. [Google Scholar]
  64. Giulian, D.; Li, J.; Li, X.; George, J.; Rutecki, P. Effect of microglia derived cytokines on gliosis in CNS Dev. Neurosci. 1994, 16, 128–136. [Google Scholar]
  65. McGeer, E.G.; McGeer, P.L. Inflammatory processes in Alzheimer’s disease. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2003, 27, 741–749. [Google Scholar] [CrossRef]
  66. Sarter, J.P. Developmental origins of the age-related decline in cortical cholinergic function and associated cognitive abilities. Neurobiol. Aging 2004, 25, 1127–1139. [Google Scholar] [CrossRef] [PubMed]
  67. Nagele, R.G.; Wegiel, J.; Venkataraman, V.; Imaki, H.; Wang, K.C.; Wegiel, J. Contribution of glial cells to the development of amyloid plaques in Alzheimer’s disease. Neurobiol. Aging 2004, 25, 663–674. [Google Scholar] [CrossRef] [PubMed]
  68. Serrano-Pozo, A.; Mielke, M.L.; Gómez-Isla, T.; Betensky, R.A.; Growdon, J.H.; Frosch, M.P.; Blennow, K.; Zetterberg, H. Reactive glia not only associates with plaques but also parallels tangles in Alzheimer’s disease. Am. J. Pathol. 2014, 179, 1373–1384. [Google Scholar] [CrossRef] [PubMed]
  69. Lyman, M.; Lloyd, D.G.; Ji, X.; Vizcaychipi, M.P.; Ma, D. Neuroinflammation: The role and consequences. Neurosci. Res. 2014, 79, 1–12. [Google Scholar] [CrossRef]
  70. Villemagne, V.L.; Burnham, S.; Bourgeat, P.; Brown, B.; Ellis, K.A.; Salvado, O.; Maruff, P.; Ames, D.; Rowe, C.C. Amyloid β deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: A prospective cohort study. Lancet Neurol. 2013, 12, 357–367. [Google Scholar] [CrossRef]
  71. Jack, C.R.; Wiste, H.J.; Lesnick, T.G.; Weigand, S.D.; Knopman, D.S.; Vemuri, P.; Mielke, M.M.; Lowe, V.J. Brain β-amyloid load approaches a plateau. Neurology 2013, 80, 890–896. [Google Scholar] [CrossRef] [Green Version]
  72. Doan, T.; Massarotti, E. Rheumatoid Arthritis: An Overview of New and Emerging Therapies. J. Clin. Pharmacol. 2005, 45, 751. [Google Scholar] [CrossRef]
  73. Choy, E.H.S.; Panayi, G.S. Cytokine pathways and joint inflamation in rheumatoid arthritis. N. Engl. J. Med. 2001, 344, 907–916. [Google Scholar] [CrossRef]
  74. Ghoryani, M.; Shariati-Sarabi, Z.; Tavakkol-Afshari, J.; Ghasemi, A.; Poursamimi, J.; Mohammadi, M. Amelioration of clinical symptoms of patients with refractory rheumatoid arthritis following treatment with autologous bone marrow-derived mesenchymal stem cells: A successful clinical trial in Iran. Biomed. Pharmacother. 2019, 109, 1834–1840. [Google Scholar] [CrossRef]
  75. Feldman, M.; Maini, R.N. The role of cytokines in the pathogenesis of rheumatoid arthritis. Rheumatology 1999, 38, 3–7. [Google Scholar]
  76. Zheng, X.; Wu, S.; Hincapie, M.; Hancock, W.D. Study of the human plasma proteome of rheumatoid arthritis. J. Chromatography 2009, 1216, 3538–3545. [Google Scholar] [CrossRef]
  77. Boldi, K.; Prokaeva, T.; Spencer, B.; Eberhard, M.; Connors, R.H.; Seldin, D.C. AL-Base a visual platform analysis for the study of amyloidogenic immunoglobulin light chain sequences. Amyloid: J. Protein Fold. Disord. 2009, 16, 1–8. [Google Scholar]
  78. Yuan, J.; Meloni, B.P.; Shi, T.; Bonser, A.; Papadimitriou, J.M.; Mastaglia, F.L.; Zhang, C.; Zheng, M.; Gao, J. The potential influence of bone-derived modulators on the progression of Alzheimer’s disease. J. Alzheimer’s Dis. 2019, 69, 59–70. [Google Scholar] [CrossRef]
  79. Donnelly, J.P.; Hanna, M.; Sperry, B.W.; Seitz, W.H. Carpal tunnel syndrome: A potential early, red-flag sign of amyloidosis. J. Hand Surg. Am. 2019, 44, 868–876. [Google Scholar] [CrossRef]
  80. Brunger, A.F.; Nienhuis, H.L.A.; Bijzet, J.; Hazenberg, B.P.C. Causes of AA amyloidosis: A systematic review. Amyloid 2020, 27, 1–12. [Google Scholar] [CrossRef]
  81. Hart, P.H.; Hunt, E.K.; Bonder, C.S.; Watson, C.J.; Finlay-Jones, J.J. Regulation of surface and soluble TNF receptor expression on human monocytes and synovial fluid macrophages by IL-4 and IL-10. J. Immunol. 1996, 157, 3672–3680. [Google Scholar]
  82. Walker, K.A.; Gottesman, R.F.; Wu, A.; Knopman, D.S.; Gross, A.L.; Mosley, T.H.; Selvin, E.; Jack, C.R.; Gottesman, R.F. Systemic inflammation during midlife and cognitive change over 20 years: The ARIC study. Neurology 2019, 92, 1256–1267. [Google Scholar]
  83. Lin, T.M.; Chen, W.S.; Sheu, J.J.; Chen, Y.H.; Chen, J.H.; Chang, C.C. Autoimmune rheumatic diseases increase dementia risk in middle-aged patients: A nationwide cohort study. PLoS ONE 2018, 13, e018675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Wallin, K.; Solomon, A.; Kreholt, I.; Tuomilehto, J.; Soininen, H.; Walin, K. Midlife rheumatoid arthritis increases the risk of cognitive impairment two decades later: A population-based study. J. Alzheimer’s Dis. 2012, 31, 669–676. [Google Scholar] [CrossRef] [PubMed]
  85. Sanchez, S.; Judge, A.; Batra, R.N.; Price, A.J. Development and validation of a clinical prediction model for patient-reported pain and function after primary total knee replacement surgey. Scientific Reports. 2018, 3381, 25–30. [Google Scholar]
  86. Zandi, P.P.; Breitner, J.C.S. Do NSAIDs prevent Alzheimer’s disease? And, if so, why? The epidemiological evidence. Neurobiol. Aging 2001, 22, 811–817. [Google Scholar] [CrossRef]
  87. Lebrasseur, N.K.; Tchkonia, T.; Kirkland, J.L. Cellular senescence and the biology of aging, disease, and frailty. Nestle Nutr. Inst. Workshop Ser. 2015, 83, 11–18. [Google Scholar] [PubMed] [Green Version]
  88. Walker, K.A.; Windham, B.G.; Power, M.C.; Hoogeveen, R.C.; Folsom, A.R.; Ballantyne, C.M.; Konopman, D.S.; Selvin, E.; Jack, C.R.; Gottesman, R.F. The association of mid to late-life systemic inflammation with white matter structure in older adults: The ARIC study. Neurobiol. Aging 2018, 68, 26–33. [Google Scholar] [CrossRef]
  89. Sangha, P.S.; Thakur, M.; Akhtar, Z.; Ramani, S.; Rubby, S. The Link Between Rheumatoid Arthritis and Dementia. Rev. Cureus. 2020, 12, e7855. [Google Scholar] [CrossRef]
  90. Juszczyk, G.; Mikulska, J.; Kasperek, K.; Pietrzak, D.; Mrozek, W.; Herbet, M. Chronic Stress and Oxidative Stress as Common Factors of the Pathogenesis of Depression and Alzheimer’s Disease: The Role of Antioxidants in Prevention and Treatment. Antioxidants 2021, 10, 1439. [Google Scholar] [CrossRef]
  91. Kukar, T.; Golde, T. Possible mechanisms of action of NSAIDs and related compounds that modulate gamma-secretase cleavage. Curr. Top. Med. Chem. 2008, 8, 47–53. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, C.; Wang, Y.; Wang, D.; Zhang, J.; Zhang, F. NSAID Exposure and Risk of Alzheimer’s Disease: An Updated Meta-Analysis from Cohort Studies. Front. Aging Neurosci. 2018, 10, 83. [Google Scholar] [CrossRef]
  93. Etminan, M.; Gill, S.; Samii, A. Effect of non-steroidal anti-inflammatory drugs on risk of Alzheimer’s disease: Systematic review and meta-analysis of observational studies. BMJ 2003, 327, 128. [Google Scholar] [CrossRef] [Green Version]
  94. Meyer, P.F.; Tremblay-Mercier, J.; Leoutsakos, J.; Madjar, C.; Lafaille-Maignan, M.E.; Savard, M.; Rosa-Neto, P.; Poirier, J.; Etienne, P.; Breitner, J.; et al. INTREPAD: A randomized trial of naproxen to slow progress of presymptomatic Alzheimer disease. Neurology 2019, 92, e2070–e2080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Hershey, L.A.; Lipton, R.B. Naproxen for presymptomatic Alzheimer disease: Is this the end, or shall we try again? Neurology 2019, 92, 829–830. [Google Scholar] [CrossRef]
  96. Molina, E.; Del Rincon, I.; Restrepo, J.F.; Battafarano, D.F.; Escalante, A. Association of socioeconomic status with treatment delays, disease activity, joint damage, and disability in rheumatoid arthritis. Arthritis Care Res. 2015, 67, 940–946. [Google Scholar] [CrossRef]
  97. Chou, R.C.; Kane, M.; Ghimire, S.; Gautam, S.; Gui, J. Treatment for rheumatoid arthritis and risk of Alzheimer’s disease: A nested case-control analysis. CNS Drugs 2016, 30, 1111–1120. [Google Scholar] [CrossRef] [Green Version]
  98. Ungprasert, P.; Wijarnpreecha, K.; Thongprayoon, C. Rheumatoid arthritis and the risk of dementia: A systematic review and meta-analysis. Neurol. India 2016, 64, 56–61. [Google Scholar] [CrossRef] [PubMed]
  99. Dufouil, C.; Seshadri, S.; Chêne, G. Cardiovascular risk profile in women and dementia. J. Alzheimers Dis. 2014, 42 (Suppl. S4), 353–363. [Google Scholar] [CrossRef] [PubMed]
  100. Scott, D.L.; Wolfe, F.; Huizinga, T.W. Rheumatoid arthritis. Lancet 2010, 376, 1094–1108. [Google Scholar] [CrossRef]
  101. Turesson, C.; O’Fallon, W.M.; Crowson, C.S.; Gabriel, S.E.; Matteson, E.L. Extra-articular disease manifestations in rheumatoid arthritis: Incidence trends and risk factors over 46 years. Ann. Rheum Dis. 2003, 62, 722–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Bae, S.C.; Lee, Y.H. Causal association between rheumatoid arthritis and a decreased risk of Alzheimer’s disease: A Mendelian randomization study. Z. Rheumatol. 2019, 78, 359–364. [Google Scholar] [CrossRef]
  103. Judge, A.; Garriga, C.; Arden, N.K.; Lovestone, S.; Prieto-Alhambra, D.; Cooper, C.; Edwards, C.J. Protective effect of antirheumatic drugs on dementia in rheumatoid arthritis patients. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2017, 3, 612–621. [Google Scholar] [CrossRef]
  104. Zhou, M.; Xu, R.; Kaelber, D.C.; Gurney, M.E. Tumor Necrosis Factor (TNF) blocking agents are associated with lower risk for Alzheimer’s disease in patients with rheumatoid arthritis and psoriasis. PLoS ONE 2020, 15, e0229819. [Google Scholar] [CrossRef] [Green Version]
  105. McGeer, P.L.; Schulzer, M.; McGeer, E.G. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer’s disease: A review of 17 epidemiologic studies. Neurology 1996, 47, 425–432. [Google Scholar] [CrossRef] [PubMed]
  106. Kao, L.T.; Kang, J.H.; Lin, H.C.; Huang, C.C.; Lee, H.C.; Chung, S.D. Rheumatoid arthritis was negatively associated with Alzheimer’s disease: A population-based case-control study. PLoS ONE 2016, 11, e0168106. [Google Scholar] [CrossRef] [Green Version]
  107. Policicchio, S.; Ahmad, A.N.; Powell, J.F.; Proitsi, P. Rheumatoid arthritis and risk for Alzheimer’s disease: A systematic review and meta-analysis and a Mendelian Randomization study. Sci. Rep. 2017, 7, 12861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Gorelick, P.B. Role of inflammation in cognitive impairment: Results of observational epidemiological studies and clinical trials. Ann. NY Acad Sci. 2010, 1207, 155–162. [Google Scholar] [CrossRef] [PubMed]
  109. Jiang, Q.; Jin, S.; Jiang, Y.; Liao, M.; Feng, R.; Zhang, L.; Lui, B.; Cai, Q. Alzheimer’s disease variants with the genome-wide significance are significantly enriched in immune pathways and active in immune cells. Mol. Neurobiol. 2017, 54, 594–600. [Google Scholar] [CrossRef]
  110. Le Page, A.; Dupuis, G.; Frost, E.H.; Larbi, A.; Pawelec, G.; Witkowski, J.M.; Fulop, T. Role of the peripheral innate immune system in the development of Alzheimer’s disease. Exp. Gerontol. 2018, 107, 59–66. [Google Scholar] [CrossRef]
  111. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef]
  112. Banks, W.A.; Kastin, A.J.; Broadwell, R.D. Passage of cytokines across the blood-brain barrier. Neuroimmunomodulation 1995, 2, 241–248. [Google Scholar] [CrossRef]
  113. Roth, J.; Harre, E.M.; Rummel, C.; Gerstberger, R.; Hubschle, T. Signaling the brain in systemic inflammation: Role of sensory circumventricular organs. Front. Biosci. 2004, 9, 290–300. [Google Scholar] [CrossRef] [Green Version]
  114. Das, U.N. Vagus nerve stimulation, depression, and inflammation. Neuropsychopharmacology 2007, 32, 2053–2054. [Google Scholar] [CrossRef] [PubMed]
  115. Lai, P.; Wang, T.; Zhang, N.; Wu, K.; Yao, C.J.; Lin, C. Changes of blood-brain-barrier function and transfer of amyloid beta in rats with collagen-induced arthritis. J. Neuroinflamm. 2021, 18, 174–182. [Google Scholar] [CrossRef]
  116. Nishioku, T.; Yamauchi, A.; Takata, F.; Watanabe, T.; Furusho, K.; Shuto, H.; Dohgu, S.; Kataoka, Y. Disruption of the blood–brain barrier in collagen-induced arthritic mice. Neurosci. Lett. 2010, 482, 208–211. [Google Scholar] [CrossRef]
  117. Arbustini, E.; Morbini, P.; Verga, L.; Anesi, E.; Merlini, G. Light and electron microscopy immunohistochemical characterization of amyloid deposits. Amyloid 1997, 4, 157–170. [Google Scholar] [CrossRef]
  118. Murphy, C.L.; Eulitz, M.; Hrncic, R.; Wal, C.; Weis, L.; Deborah, T. Chemical typing of amyloid protein contained in formalin-fixed paraffin-embedded biopsy specimens. Am. J. Clin. Pathol. 2001, 116, 135–142. [Google Scholar] [CrossRef] [Green Version]
  119. Lachmann, H.J.; Booth, D.R.; Booth, S.E.; Pepys, M.B.; Hawkins, P.N. Misdiagnosis of hereditary amyloidosis as AL (primary) amyloidosis. N. Engl. J. Med. 2002, 346, 1786–1791. [Google Scholar] [CrossRef] [Green Version]
  120. Saraiva, M.J. Sporadic cases of hereditary systemic amyloidosis. N. Engl. J. Med. 2002, 346, 1818–1819. [Google Scholar] [CrossRef]
  121. Merlini, G.; Bellotti, V. Molecular Mechanisms of Amyloidosis. N. Engl. J. Med. 2003, 349, 583–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Ohno, S.; Yoshimoto, M.; Honda, S.; Myiachi, S. The antisense approach in amyloid light chain amyloidosis: Identification of monoclonal Ig and inhibition of its production by antisense oligonucleotides in in vitro and in vivo models. J. Immunol. 2002, 169, 4039–4045. [Google Scholar] [CrossRef] [Green Version]
  123. Xia, H.; Mao, Q.; Paulson, H.L.; Davidson, B.L. siRNA-mediated gene silencing in vitro and in vivo. Nat. Biotechnol. 2002, 20, 1006–1010. [Google Scholar] [CrossRef] [PubMed]
  124. Jick, H.; Zornberg, G.L.; Jick, S.S.; Seshadri, S.; Drachman, D.A. Statins and the risk of dementia. Lancet 2000, 356, 1627–1631. [Google Scholar] [CrossRef]
  125. Weggen, S.; Eriksen, J.L.; Das, P.; Sagi, S.A.; Wang, R.; Pietrzik, C.U.; Findlav, K.A.; Smith, T.E.; Murphy, M.P.; Bulter, T.; et al. A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature 2001, 414, 212–216. [Google Scholar] [CrossRef]
  126. Eriksen, J.L.; Sagi, S.A.; Smith, T.E.; Weggen, S.; Das, P.; McLendon, D.C.; Ozols, V.V.; Jessing, K.W.; Zavitz, K.H.; Koo, E.H.; et al. NSAIDs and enantiomers of flurbiprofen target γ-secretase and lower Aβ42 in vivo. J. Clin. Investig. 2003, 112, 440–449. [Google Scholar] [CrossRef] [Green Version]
  127. Ghosh, A.K.; Shin, D.; Downs, D.; Koelsch, G.; Lin, X.; Ermolieff, J.; Tang, I. Design of potent inhibitors for human brain memapsin 2 (β-secretase). J. Am. Chem. Soc. 2000, 122, 3522–3523. [Google Scholar] [CrossRef]
  128. Green, R.C.; Schneider, L.S.; Amato, D.A.; Beelen, A.P.; Wilcock, G.; Swabb, E.A.; Zawitz, K.H. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: A randomized controlled trial. JAMA. 2009, 302, 2557–2564. [Google Scholar] [CrossRef] [Green Version]
  129. Walker, L.C.; Rosen, R.F. Alzheimer therapeutics—What after the cholinesterase inhibitors? Age Ageing 2006, 35, 332–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Jevotovsky, D.S.; Alfonso, A.R.; Einhorn, T.A.; Chiu, E.S. Osteoarthritis and stem cell therapy in humans: A systematic review. Osteoarthr. Cartil. 2018, 26, 711–729. [Google Scholar] [CrossRef] [Green Version]
  131. Xu, Z.; Nan, W.; Zhang, X.; Sun, Y.; Yang, J.; Lu, K.; Zhao, Y. Umbilical cord mesenchymal stem cells conditioned medium promoted AB25-35 phagocytosis by modulating autophagy and AB-degrading enzymes in BV2 cells. J. Mol. Neurosci. 2018, 65, 222–233. [Google Scholar] [CrossRef] [PubMed]
  132. Duncan, T.; Valenzuela, M. Alzheimer’s disease, dementia, and stem cell therapy. Stem Cell Res. 2017, 8, 111–115. [Google Scholar] [CrossRef] [PubMed]
  133. Biogen. Biogen Plans to Submit a Regulatory Application for Aducanumab in Alzheimer’s Disease Based on a New Analysis of a Larger Dataset from Phase 3 Trials. 2019. Available online: http://investors.biogen.com/news-releases/news-release-details/biogen-plans-regulatory-filing-aducanumab-alzheimers-disease (accessed on 10 November 2020).
  134. Xiong, M.; Jiang, H.; Serrano, J.R.; Gonzales, E.R.; Wang, C.; Gratuze, M.; Hoyle, R.; Bien-Ly, N.; Silverman, A.P.; Sullivan, P.M.; et al. APOE immunotherapy reduces cerebral amyloid angiopathy and amyloid plaques while improving cerebrovascular function. Sci. Transl. Med. 2021, 13, eabd7522. [Google Scholar] [CrossRef] [PubMed]
  135. NIH US National Library of Medicine. Study to Confirm the Safety and Efficacy of BAN2401 in Subjects with Early Alzheimer’s Disease (clarity AD) (2019). Available online: https://clinicaltrials.gov/ct2/show/NCT03887455 (accessed on 20 November 2020).
Table 1. Age-dependent amyloid distribution in selected organs [28].
Table 1. Age-dependent amyloid distribution in selected organs [28].
Systemic OrgansAmyloid
Brain
α-Synuclein
Tau
LungTransthyretin
HeartTransthyretin
Atrial natriuretic factor
AortaApolipoprotein AI
VeinEGF-containing fibulin-like extracellular matrix protein 1
Tendons and LigamentsTransthyretin
Apolipoprotein AI
Knee jointTransthyretin
Apolipoprotein AI
Table 2. Basic amyloidosis.
Table 2. Basic amyloidosis.
Kind of
Amyloidosis
CausePrecursorClinical
Manifestation
Literature
ALclonal plasma cell dyscrasia
multiple myeloma
non-Hodgkin lymphoma
Waldenström disease
lambda
or
kappa immunoglobulin free light chain
-cardiomyopathy
-nephrotic syndrome
-renal failure
-hepatomegaly
-splenomegaly
-diarrhea
-arthropathy
-bleeding
-adrenal dysfunction
[31]
AAlongstanding inflammation (rheumatoid arthritis,
inflammatory bowel
disease)
chronic infections (e.g.,
tuberculosis, osteomyelitis, leprosy)
hereditary
autoinflammatory diseases
(e.g., familial Mediterranean fever, which is called FMF)
HDL3-associated
apolipoprotein
SAA (serum amyloid A protein)
cute phase
reactant
-proteinuria (progressing to nephrotic syndrome)
-loss of renal function (progressing to renal failure)
-bowel involvement
-splenomegaly
-hepatomegaly
-cardiomyopathy
[32]
ATTRautosomal dominantly
inherited point mutations
of the precursor protein
transthyretin (TTR)
TTR-peripheral
-autonomic neuropathy
-cardiomyopathy
-renal failure
-eye involvement
(vitreous opacities)
[33]
Aβ2Mend-stage renal disease in
which highly increased
serum levels of β2-microglobulin
β2-microglobulin-shoulder pains as a first manifestation
- autonomic
neuropathy
-gastrointestinal
symptoms
[34]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Trzeciak, P.; Herbet, M.; Dudka, J. Common Factors of Alzheimer’s Disease and Rheumatoid Arthritis—Pathomechanism and Treatment. Molecules 2021, 26, 6038. https://doi.org/10.3390/molecules26196038

AMA Style

Trzeciak P, Herbet M, Dudka J. Common Factors of Alzheimer’s Disease and Rheumatoid Arthritis—Pathomechanism and Treatment. Molecules. 2021; 26(19):6038. https://doi.org/10.3390/molecules26196038

Chicago/Turabian Style

Trzeciak, Paulina, Mariola Herbet, and Jarosław Dudka. 2021. "Common Factors of Alzheimer’s Disease and Rheumatoid Arthritis—Pathomechanism and Treatment" Molecules 26, no. 19: 6038. https://doi.org/10.3390/molecules26196038

Article Metrics

Back to TopTop