Next Article in Journal
Longitudinal Optical Coherence Tomography Imaging Reveals Hyperreflective Foci Characteristics in Relapsing–Remitting Multiple Sclerosis Patients
Previous Article in Journal
Vascular Complications in Patients with ECMO Support after Cardiac Surgery
Previous Article in Special Issue
Supplementing Best Care with Specialized Rehabilitation Treatment in Parkinson’s Disease: A Retrospective Study by Different Expert Centers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolic and Immune System Dysregulation: Unraveling the Connections between Alzheimer’s Disease, Diabetes, Inflammatory Bowel Diseases, and Rheumatoid Arthritis

1
Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
2
Dementia Disorders Centre, Medical University of Wroclaw, 50-425 Scinawa, Poland
3
Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(17), 5057; https://doi.org/10.3390/jcm13175057
Submission received: 9 August 2024 / Revised: 22 August 2024 / Accepted: 23 August 2024 / Published: 26 August 2024

Abstract

:
Alzheimer’s disease (AD), diabetes mellitus (DM), inflammatory bowel diseases (IBD), and rheumatoid arthritis (RA) are chronic conditions affecting millions globally. Despite differing clinical symptoms, these diseases share pathophysiological mechanisms involving metabolic and immune system dysregulation. This paper examines the intricate connections between these disorders, focusing on shared pathways such as insulin resistance, lipid metabolism dysregulation, oxidative stress, and chronic inflammation. An important aspect is the role of amyloid-beta plaques and tau protein tangles, which are hallmark features of AD. These protein aggregates are influenced by metabolic dysfunction and inflammatory processes similar to those seen in DM, RA, and IBD. This manuscript explores how amyloid and tau pathologies may be exacerbated by shared metabolic and immune dysfunction. Additionally, this work discusses the gut–brain axis and the influence of gut microbiota in mediating disease interactions. Understanding these commonalities opens new avenues for multi-targeted therapeutic approaches that address the root causes rather than merely the symptoms of these conditions. This integrative perspective could lead to more effective interventions and improved patient outcomes, emphasizing the importance of a unified approach in managing these interconnected diseases.

1. Introduction

About 60%–70% of occurrences of dementia in older adults are caused by Alzheimer’s disease (AD). Though substantial progress has been made in understanding genetic and environmental risk factors, as well as the pathology findings connected to this neurodegenerative illness, the exact cause of AD is still unknown. The aggregation and deposition of amyloid-β (Aβ) peptides on the extracellular surface of neuronal cells, which results in the production of Aβ oligomers and fibrils in the brain, is one of the primary characteristics of AD. Additionally, tau protein hyperphosphorylation in AD has an influence on the brain, building up in neuronal microtubules and creating neurofibrillary tangles. These occurrences encourage cytotoxic effects on neural cells that lead to a deterioration in cognition. Another hallmark gaining interest is neuroinflammation occurring during the course of the disease. Neuroinflammation is facilitated by microglia and astrocytes, as well as some immune cells from the periphery that penetrate the brain [1,2]. Even though neuroinflammatory processes at the beginning play significant roles in neuroprotection, chronic and increasing neuroinflammation can negatively impact brain function and cause neurodegeneration and neurological disability [3]. Both innate and adaptive immune responses to infections require microglial cells. Furthermore, it has been demonstrated that the type of microglia plays a critical role in controlling myelination and neurogenesis [1]. Microglial cells become activated and secrete different inflammatory substances under pathological situations. Similar to this, proinflammatory cytokines, pathogenic Aβ, and tau species can stimulate astrocytes, causing them to release these substances [4,5]. Chronically activated microglia and astrocytes can induce brain damage by promoting the formation of toxic protein aggregates and increasing the susceptibility of neurons to cell death through the continuous release of pro-inflammatory cytokines [6,7].
The “glymphatic” pathway facilitates the exchange of interstitial fluid (ISF) and para-arterial cerebrospinal fluid (CSF) in the brain parenchyma, which helps the central nervous system (CNS) clean itself of a variety of hazardous waste products, including amyloid beta [8]. In the elderly brain, neuroinflammation significantly disrupts this system [9,10], which may cause amyloid beta buildup and subsequent neuronal damage. Aquaporin 4 (AQP4) channels, which are highly expressed on the astrocytic endfeet abutting cerebral capillaries, are necessary for lymphatic clearance [11,12]. However, AQP4 polarization is reduced by reactive astrogliosis in response to inflammatory signaling. Furthermore, the aged brain exhibits a substantial upregulation of reactive astrocytes of the “A1” phenotype, which prominently expresses classical complement cascade genes and contributes to neuronal death in many age-associated neurodegenerative disorders [13,14,15].
Alzheimer’s Disease, a neurodegenerative disorder leading to progressive cognitive decline, has traditionally been viewed through the lens of neuropathology. However, growing evidence suggests that metabolic abnormalities, such as insulin resistance and dyslipidemia, play critical roles in its pathogenesis. Similarly, diabetes (DM), characterized by chronic hyperglycemia and metabolic instability, is now understood to have profound effects on the nervous system, potentially accelerating neurodegenerative processes. Rheumatoid arthritis (RA), an autoimmune condition that primarily targets synovial joints, shares common inflammatory pathways with AD, suggesting that chronic systemic inflammation may be a crucial link between these diseases. The inflammation observed in RA is not confined to the joints but can have widespread effects, implicating immune system dysregulation as a shared factor. Moreover, irritable bowel diseases (IBD), encompassing Crohn’s disease and ulcerative colitis, involve chronic inflammation of the gastrointestinal tract. These conditions also demonstrate significant overlap with both metabolic and immune dysfunctions seen in AD, DM, and RA. The gut–brain axis and gut–immune interactions highlight a complex network of bidirectional communication that can influence disease progression and manifestation across these conditions.
This paper aims to explore the converging metabolic and immunological pathways that link Alzheimer’s disease, diabetes, rheumatoid arthritis, and inflammatory bowel diseases. By unraveling these connections, we hope to uncover novel insights into their shared etiologies and identify potential therapeutic targets that could benefit multiple chronic diseases simultaneously. Understanding these commonalities may pave the way for innovative, holistic approaches to treatment and prevention, ultimately improving patient outcomes across these diverse yet interconnected disorders.

2. AD and Diabetes

Over the past few decades, there has been a rise in the prevalence of diabetes mellitus (DM). Insulin resistance is a defining feature of type 2 diabetes mellitus (T2DM), which accounts for over 90% of all occurrences of diabetes worldwide, according to the International Diabetes Federation [16]. One in ten persons worldwide have diabetes. It was discovered that T2DM is associated with an increased risk of dementia, similar to dementia itself; however, dementia is also associated with a higher risk of this metabolic abnormality [17]. While AD is a central nervous system issue and T2DM is predominantly a peripheral disease, both conditions are chronic and complex, and they have certain characteristics in common, such as protracted prodromal periods [18]. Furthermore, both demonstrate how inflammation and chronic oxidative stress contribute to the development of the disease. In addition to age, sedentary lifestyle, poor dietary habits, obesity, chronic stress, and genetics are common risk factors [17]. A meta-analysis of DM cases and the risk of all types of dementia, including vascular dementia and AD, revealed a 73% increase in the risk of all types of dementia and a 56% increase in the risk of AD in diabetic patients [19]. The relationship between type 2 diabetes and AD may be explained by a number of theories, including hyperglycemia, which causes glutamate-induced excitotoxicity in neuronal cells and insulin resistance in the brain, which may aid in the buildup of amyloid-β, tau phosphorylation, oxidative stress, the formation of advanced glycation end products (AGEs), and apoptosis [20]. An 11-year follow-up study of a Taiwanese population found that diabetic patients are more susceptible to AD compared to non-diabetic patients [21]. Nevertheless, the connection between AD and T2DM remains unclear. The similarities between AD and diabetes mellitus, as well as the other diseases described in this paper, are shown in Figure 1.
According to MRI studies, hippocampus size has been found to decline in older diabetic individuals. Diabetes (db/db) mice also exhibit age-dependent atrophies of the hippocampus and cortical regions [22]. Reduced neurogenesis and increased neuronal death may have caused these alterations. Throughout adulthood, new neurons are constantly being created in two significant locations of the brain: the subventricular zone and the dentate gyrus of the hippocampus [23]. The growth of neuroprogenitor cells (NPCs) and their eventual differentiation into neurons, astrocytes, and oligodendrocytes is known as neurogenesis. Animal models of obesity and diabetes have been shown to exhibit impaired neurogenesis. For instance, in the Goto-Kakizaki rat, a genetic model of type 2 diabetes, reduced NPC survival has also been noted [24]. In spontaneously obese diabetic mice, a model for type 1 diabetes, the dentate gyrus of newly produced NPCs exhibits a reduction in bromodeoxyuridine-BrdU labeling [25]. In both spontaneously diabetic mice and animals treated with streptozotocin, impaired NPC proliferation is linked to increased glucocorticoid levels and decreased brain-derived neurotrophic factor (BDNF) expression [26]. The stimulation of the NF-kB pathway in the mouse hypothalamus has been linked to the impairment of NPCs caused by a high-fat diet, just like in diabetic rats [27]. According to a recent study, neuronal markers drastically decrease, and NPCs differentiate into a glial phenotype when they are differentiated in the presence of cytokines such as IL-1β, TNF-α, and IL-6 [28]. The results of this study imply that increased levels of circulating cytokines in diabetes may obstruct brain neurogenesis.
Moreover, reduced scores on numerous cognitive function tests are linked to higher levels of glycosylated hemoglobin [29]. Its levels in postmenopausal osteoporotic women without diabetes have been linked to the likelihood of acquiring MCI and dementia [30]. According to the Hisayama study, dementia, from all causes, is linked to glucose intolerance [31]. Individuals with diabetic retinopathy also had low scores on cognitive function tests, according to the Edinburgh type 2 diabetes study [32]. Moreover, higher average blood glucose levels over the previous five years are associated with an increased risk of dementia in both participants with and without diabetes, according to adult changes in the research [33]. In a group of type 2 diabetic individuals, even acute hyperglycemia has been demonstrated to impact mood and cognitive performance [34]. The Maastricht Aging Study found that during a 12-year period, diabetic individuals experienced more cognitive decline than non-diabetic participants, as seen by reductions in word recall and information processing speed [35]. A summary of prospective observational research showed that diabetes raises the risk of cognitive deterioration by a factor of 1.2 to 1.7 [36]. Cognitive performance is correlated with both changes in glycemic control and even the trajectories of glycemic control [37,38]. Because of the risks involved with hypoglycemic episodes, the American Geriatrics Society does not advise reducing the A1c below 8% [39]. Consequently, glucose management must be started early as an interventional technique to stop cognitive impairment [40].
Patients with diabetes may have cognitive deterioration due to a variety of causes. For instance, MRI studies have demonstrated that chronic hyperglycemia-mediated hippocampal impairment and insulin resistance have been linked to AD-like reductions in cerebral glucose metabolism, even in prediabetic people with adequate cognitive function [41,42]. There are numerous insulin receptors located all over the brain. The majority of insulin receptors are found on neurons, where they are part of the postsynaptic density (PSD) and are strongly expressed at the presynaptic axon terminal of synapses. Because of the high concentration of insulin receptors in the entorhinal cortex, frontal cortex, and hippocampal regions of the brain, insulin is thought to be important for learning and memory [43]. Furthermore, improvements in memory following insulin injection have been reported, as well as alterations in insulin receptors in the hippocampal region as a result of spatial learning. Although studies on human and animal models also reported similar effects, it is unclear how specifically insulin affects cognition [44]. Insulin favors long-term depression (LTD) and long-term potentiation (LTP), which may contribute to changes in hippocampal synaptic plasticity, according to the results of animal research. Learning and memory, which are dependent on the hippocampus, are mediated by these chemical pathways [45,46,47]. The expression of the N-methyl-D-aspartate (NMDA) receptor in the membrane is one such mechanism that is controlled by insulin [48]; its expression is governed by the insulin-activated ERK1/2 or PI3-K pathways [49,50]. Additionally, synaptic remodeling—which is necessary for neural plasticity—is influenced by insulin signaling [51]. Further results imply that memory development requires intact insulin signaling [52]. Moreover, the hormone acts on neurons through the AKT and MAPK signaling pathways [53,54]. It is thought to promote neuronal outgrowth, alter catecholamine release and uptake, and control the expression and location of GABA, a key inhibitory neurotransmitter in mammalian brain synapses. It is well-recognized that GABA is involved in sleep, memory, learning, and reproductive system activity [55]. Furthermore, it has a major impact on how the frontal brain regulates neuronal activity, body weight, plasticity, and food intake [56]. Insulin additionally controls the expression of NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors, and it modifies activity-dependent synaptic plasticity through the AKT pathway and NMDA receptor signaling, which results in long-term depression and long-term potentiation [46,57].
Cognitive dysfunction is likely associated with abnormalities in the hypothalamic–pituitary–adrenal axis, inflammatory mediators, rheological variables, and hyperglycemia [58]. An epigenetic mechanism of cognitive impairment in elderly obese individuals with diabetes has been proposed as a result of higher histone deacetylases class IIa in the brains of diabetic people [59]. Insulin resistance causes Akt—a protein involved in several physiological functions, including glucose metabolism—to become less activated. It also inhibits GSK3β, one of the kinases that phosphorylate tau. Consequently, elevated GSK3β activation during insulin resistance may result in tau hyperphosphorylation, which is a crucial element of the neurofibrillary tangles that are seen in AD patients’ brains. Insulin and Aβ peptide are broken down by an enzyme that cleaves insulin (Insulin Degrading Enzyme [IDE]). Consequently, IDE is sequestered from Aβ by hyperinsulinemia, which promotes its accumulation [60,61]. Among the shared pathways found in the brain tissues of AD and diabetes patients are protein misfolding, oxidative stress, and inflammation [40,62]. The accumulation of advanced glycation end products, a significant contributor to diabetic problems, has been discovered in the brain.
In type 2 diabetes, amyloid polypeptide (AP), a protein co-expressed and released with insulin, causes cell death in the islet of Langerhans. In an aquatic environment, AP spontaneously produces amyloid clumps, much like the Aβ protein in AD. The structural characteristics of these proteins showed a significant overlap (90%) according to the alignment analysis. A locally produced amyloid protein is deposited in both disorders, along with a slow reduction in cell count. Among these harmful processes is the chaperone protein pathway, a protein trafficking system that binds developing proteins and helps move them around the cell [63]. Interestingly, patients with AD showed a higher frequency of islet amyloid; nevertheless, brain amyloid did not increase in T2DM patients compared to the normoglycemic group; instead, the density of neuritic plaques was linked to the length of diabetes [63].
The microtubule-associated protein tau, which shares physicochemical characteristics with the condition in the brain, is also found in human islets of Langerhans, despite tauopathies being linked to degenerative conditions such as AD. According to Maj et al., beta-cell-derived mouse models with dramatically reduced insulin transcription, translation, and secretion overexpress the tau protein, highlighting the significance of tau-phosphorylation and dephosphorylation balance for appropriate insulin function [64]. It is interesting to note that pancreatic β cells from AD patients have deposits of the proteins tau and Aβ in their cytoplasm, as reported by Martinez-Valbuena et al. in T2DM patients with normal neuropathological testing [65].
In addition to the metabolism of amyloid precursor protein and the phosphorylation of tau protein, oxidative stress, impaired energetics, mitochondrial dysfunction, inflammation, lipid metabolism, and membrane lipid deregulation are among the numerous biochemical processes that also have an impact on AD and T2DM [66]. Neurotoxicity in AD is linked to mitochondrial failure and the generation of reactive oxygen species, both of which are observed in the condition [67]. It is also known that decreased cerebral glucose uptake develops decades before cognitive loss in AD. As a result, AD may be viewed as a metabolic disease mediated by brain insulin resistance, as numerous studies have demonstrated that insulin resistance might contribute to its pathogenesis [68]. Furthermore, dietary habits, gut microbiota, and peripheral metabolism may have an effect on the metabolic impairment in type 2 diabetes and Alzheimer’s disease [66].
Another protein connected to diabetes, Alpha-2-macroglobulin (A2M), was one of the proteins shown to be elevated in both the AD and T2DM groups relative to the control group in studies [69]. Among the most prevalent proteins in the peripheral blood circulation, A2M is the main non-immunoglobulin molecule. It can induce the transcriptional activation of multiple genes linked to cell proliferation/hypertrophy and atherosclerosis, as well as inhibit a wide range of proteases and pro-inflammatory cytokines [70]. A correlation has been reported between salivary levels of A2M and blood levels of HbA1c, triglycerides, fasting, and postprandial blood sugar in T2DM subjects [71,72]. The relationship between plasma A2M and DM has long been known [73]. According to research by Seddighi et al., there is a significant correlation between plasma A2M levels and the concentrations of total tau and phosphorylated tau in CSF [74]. Additionally, men who are cognitively normal but have higher serum levels of A2M are at a higher risk of developing clinical AD. Due to its ability to promote the aggregation of misfolded Aβ peptide, they postulated that A2M, an acute phase protein and chaperone protein, may be involved in the inflammation and pathogenesis of preclinical AD [74,75]. As a result, α2M appears to be significant in the pathophysiology of both AD and T2DM [69].
Neuroinflammation has been identified as an important factor in the pathophysiology of AD [76,77]. The idea that the central nervous system is immune-privileged has been called into question due to the reciprocal interactions between peripheral and central inflammation [78]. “Inflammaging” is the term used to describe chronic low-grade inflammation that contributes to aging-associated morbidity and mortality [79]. Insulin resistance and the pathophysiology of type 2 diabetes are also significantly influenced by systemic inflammation [80]. In general, microglia respond to peripheral inflammation in an adaptable manner. After exposing mice to IL-1β and TNFα systemically, the hippocampus exhibits the production of cytokines and chemokines [81]. According to a different study, immune system stimulation in mice causes brain pathologies similar to AD, which include the accumulation of APP and its proteolytic fragments as well as changes in Tau phosphorylation [82]. Higher peripheral amounts of IL-6, TNF-α, IL-1β, TGF-β, IL-12, and IL-8 are associated with AD, according to a meta-analysis of forty studies measuring peripheral blood cytokine concentrations [83]. Patients with AD who are in the early stages of the disease have activated peripheral immune cells in circulation [84]. Peripheral inflammation and cognitive dysfunction have also been linked in a number of studies. Peripheral inflammatory indicators, for instance, have been seen in AD and MCI patients [85]. They are correlated with dementia risk, according to meta-analyses of prior research [86]. Increased peripheral TNF-α and IL-1β levels have been linked to an increased risk of AD [87]. When stimulated by phytohemagglutinin (PHA), PBMCs obtained from MCI patients produce higher levels of IL-6 and IL-8 than healthy elderly controls [88]. These proteins are correlated with post-operative cognitive impairment [89]. It has been demonstrated that proinflammatory cytokines can cross the BBB [90,91]. As a result of the aging process, microglia respond to peripheral inflammation more severely and persistently, acting as a priming stimulus for them [92]. Gene expression analysis has demonstrated that brain-specific inflammatory responses are triggered by peripheral inflammation [93]. An increase in central inflammation is also likely to result from harm to the blood–brain barrier and subsequent immune cell infiltration. It is unclear whether cerebral inflammation develops from peripheral inflammation or the other way around. To fully comprehend the molecular connections between diabetes, obesity, and AD in relation to inflammation, more study is required. What is worth noting is the connection between DM and other inflammatory diseases described in our work. Diabetes not only affects the brain but also has an impact on, for example, IBD. A recent meta-analysis showed that by raising the incidence of hospitalization and infections but not of IBD-related complications or mortality, DM may have a deleterious effect on the course of IBD [94].

3. AD and Inflammatory Bowel Diseases

The two primary types of inflammatory bowel disease are ulcerative colitis (UC) and Crohn’s disease (CD). These variants can be identified by distinctions in genetic predisposition, risk factors, and clinical, endoscopic, and histological findings, despite certain similar traits. The exact etiology of inflammatory bowel disease is uncertain, although, in genetically predisposed individuals, the response of the mucosa to commensal gut flora appears to be dysregulated, leading to inflammation of the colon [95,96]. In ulcerative colitis, inflammation is typically limited to the mucosal surface. Starting in the rectum, the condition usually spreads proximally and continuously throughout the colon; however, a caecal patch of inflammation may be present in certain patients with proctitis or left-sided colitis. The degree of colonic involvement determines the stratification of disease distribution, which ranges from proctitis to extensive colitis (pancolitis) or left-sided colitis [95,97]. Crohn’s disease involves a complicated interaction of genetic predisposition, environmental variables, and changed gut microbiota that may cause dysregulated innate and adaptive immune responses. In a typical clinical setting, a young patient may present with persistent diarrhea, exhaustion, weight loss, and abdominal pain [98]. It is presented with skip lesions and transmural inflammation that can involve the entire gastrointestinal tract, extending from the mouth to the anus.
Overall, there are some papers that have studied the connection between the probability of AD and IBD. In Zhang et al.’s paper, they showed that patients with IBD were diagnosed with dementia more often than controls. Moreover, the diagnosis was performed at a lower median age than for the control group [99]. This information was also confirmed by Zingel et al. [100]. Similar results were obtained by Sand et al. However, they linked the slightly increased incidence with IBD patients’ more frequent visits to the doctors, resulting in higher detectability of dementia [101]. On the other hand, Huang et al. described that their results, although showing a higher incidence of dementia in IBD patients, are not strong and might be affected by other diseases [102]. Therefore, there is a need to carefully look at the data because there might be a bias connected to other issues.
It is commonly known that IBD is linked to gut microbial dysbiosis. But as of yet, no specific bacterium or microbial environment has been identified as the cause [103,104]. When comparing the intestinal microbiota of IBD patients to that of healthy controls, there are both qualitative and quantitative differences. Short-chain fatty acid (SCFA)-producing bacteria are less prevalent, mucosa-associated bacteria are more prevalent, and total microbial diversity is lower [105,106]. The intestinal barrier’s integrity and the immune system of the host are both impacted by dysbiosis. In people who are genetically vulnerable, this may result in an incorrect mucosal immune response and intestinal inflammation, which could eventually contribute to the onset of IBD. Disorders of the mucosal epithelial barrier that lead to the translocation of gut bacteria stimulate the hyperactivation of the mucosal immune system and the generation of proinflammatory cytokines, which in turn fuel the inflammation that is seen in patients with IBD [103,107]. The gut–brain axis, which is the term for the two-way communication between the gut and the brain, is mostly regulated by gut bacteria, according to numerous recent research. Endocrine, immunological, neurological, and metabolic pathways are only a few of the intricate and mainly unknown pathways that allow gut microorganisms and the brain to interact [108]. Gut hormones carry endocrine messages, while cytokines carry immunological information [109]. The vagus nerve is a vital pathway for the neural link between the gut microbiota and the central nervous system. This communication is thought to be mediated by molecules such as neurotransmitters, microRNA (miRNA), SCFA, and short non-coding RNA (sncRNA) [110]. Gut microbiota may have an impact on all of these signaling pathways; however, it is hypothesized that the pathology of AD is caused by the gut microbiota’s influence on neurological and immunological pathways [109]. Therefore, changes to these two-way communications in the dysbiosis state may also play a role in the pathogenesis of intestinal disorders and CNS disorders, such as the most common neurodegenerative disease, Alzheimer’s disease [111]. In our previous work we have described this connection in the AD [112]. In patients with AD, there are visible changes in the gut microbiota, which contribute to changes in the patients’ brains. The pharmacology and viability of bioactive chemicals are increasingly being studied as a novel and valuable strategy to address a range of neurological degenerative human disorders. Hericium erinaceus has emerged as one of the most intriguing possibilities within the class of mushrooms known as medicinal mushrooms (MMs). Indeed, a number of diseased brain disorders, including Alzheimer’s disease, have been demonstrated to be ameliorated by certain bioactive chemicals that were derived from H. erinaceus. The effects of erinacines on the central nervous system (CNS) have been linked to a significant increase in the generation of neurotrophic factors in a substantial body of in vitro and in vivo preclinical research [113]. Notably, it has been shown that H. erinaceus has neuroprotective benefits via increasing NGF production, which in turn regulates cholinergic neurons and enhances working memory [114,115,116]. Moreover, it was described as a mushroom with the potential to inhibit the activity of AChE, which is another potential hallmark of AD [117]. However, these positive outcomes are also visible in IBD patients. It was shown that H. erinaceus extracts may enhance the host immunity in vivo IBD model and encourage the growth of beneficial gut bacteria, which demonstrates therapeutic potential in alleviating IBD by modulating gut microbiota and the immune system [118]. Therefore, we could postulate that consuming this mushroom could be beneficial to both diseases.
An increasing amount of research has demonstrated that neuroinflammation is one of the primary pathways linking microbiota to AD and that the abnormal gut microbiota-to-CNS route also causes β-amyloid deposition in AD [119,120]. Microglia activation is largely dependent on the gut microbiota, and it has been noted that altering the gut microbiome, especially with regard to bacteria that produce SCFA, may modify neuroimmune activation [119,121]. The fact that the blood–brain barrier and the intestinal wall barrier become more permeable with aging is also significant. Additionally, this may have an impact on how the intestinal microbiota communicates with CNS and expose the latter to more potentially hazardous particles [108,110].
It is well established that dysbiosis of the gut microbiota and systemic inflammation can also impair the intestinal epithelial and blood–brain barriers’ protective qualities, rendering them more permeable and opening the door for peripheral immune cells to enter the brain. The integrity of the BBB is dependent on the proper makeup of the gut microbiota; SCFAs are important metabolites in mediating this impact [122]. Gut microbiota metabolites may enter the central nervous system more easily as a result of disruptions to the blood–brain and intestinal epithelial barriers, as well as microbiome dysbiosis linked to both CD and UC [123]. Massive amounts of lipopolysaccharides (LPSs) and amyloids formed from bacteria may seep from the gastrointestinal tract, build up in the brain and systemic circulation, and so aid in the etiology of Alzheimer’s disease. It has been suggested that LPSs and amyloids could enter the body directly through a damaged barrier and then enter the body indirectly through cytokines produced by LPS and amyloids or other tiny proinflammatory molecules that are typically transited [120,124]. Furthermore, amyloids and LPSs formed from bacteria can intensify the activation of the NF-κB signaling pathway, worsen the leakiness of the gut, and raise the levels of cytokines like IL-17 and IL-22 that are implicated in the inflammatory process of both IBD and AD [106,124]. Recent research has demonstrated that the immune system may be primed, and the immune response to endogenous synthesis of neuronal amyloid in the brain may be enhanced by exposure to bacterial amyloid proteins in the gut. Amyloid proteins generated from the microbiota have the ability to change the structure of proteins (proteinopathy) and increase inflammation in the nervous system, which can cause or worsen brain disease. Amyloid proteins produced from microbiota have the ability to function as prion proteins through molecular mimicry. This process is known as cross-seeding, wherein one amyloidogenic protein (such as tau, Aβ, α-syn, curli, or prion) induces another to take on a pathological β-sheet structure [125,126].
Gut microbiota dysbiosis represents another potential route by which CD and UC may induce the development of AD. It has been shown that bacterial metabolites, particularly SCFA, and the gut microbiota regulate microglial activation continuously. Defects in the maturation, differentiation, and function of microglia may result from an absence of a complex host microbiota [121]. Numerous investigations have demonstrated that dysbiosis of the gut microbiota is linked to both types of IBD [127]. Reduced intestinal microbiota diversity, a decline in SCFA-producing bacteria (particularly F. prausnitzii), a drop in bacteria with anti-inflammatory properties, and an increase in pro-inflammatory bacteria are all linked to CD and UC [127]. Similar to IBD, changes in the gut microbiota have also been linked to the onset of AD through immunological, endocrine, and neurological pathways [108]. Both AD and PD patients have altered gut microbiota compositions, with reduced abundances of important SCFA-producing genera such as Butyrivibrio, Eubacterium, Clostridium, and species F. prausnitzii [128]. The increased abundance of Escherichia and Shigella and the decreased presence of F. prausnitzii and Eubacterium rectale appear to be closely associated with the brain buildup of ß-amyloid in AD [128,129]. Furthermore, compared to healthy controls, AD patients had reduced abundances of anti-inflammatory bacteria such as Eubacterium hallii, Bacillus fragilis, and Bacteroides fragilis and higher abundances of proinflammatory bacteria like Escherichia, Shigella, and Pseudomonas. Proinflammatory cytokine production and amyloid deposition in the brain were linked to the abundance of proinflammatory taxa and the inhibition of anti-inflammatory taxa [128,129].
Another possibility for the co-occurrence of CD and AD in this instance is that patients with both diseases have elevated tau protein expression in common. The pathogenic process in CD is not limited to the lining of the epithelium. All parts of the gastrointestinal wall are affected, nevertheless, including the enteric nervous system (ENS), whose composition and neurochemical characteristics are changed during CD [130]. Prigent et al. showed that patients with CD but not UC have higher submucosal and myenteric plexus levels of tau expression [131]. This elevation is most likely caused by the Nrf2/NDP52 pathway. Like their counterparts in the central nervous system, enteric neurons have also been shown to produce tau protein in other recent investigations [132,133]. This demonstrates that CD and AD have a close association. Remarkably, these results are comparable to those for α-synuclein, which is overexpressed in the ENS in CD [134,135].
Interestingly, Dong et al. discovered that patients with UC and AD share genes PPARG and NOS2. Knowing that these genes reflect distinct polarization directions in macrophages and microglia, they performed an additional analysis using APP/PS1 and DSS-induced mice to examine any possible connections between the two disorders. They verified a significant decrease in the expression level of Arg1—a biomarker of M2-type polarization—in colon macrophages, hippocampal microglia (DSS-induced mice), and hippocampal microglia (APP/PS1 mice), while a significant increase was observed in the expression level of iNOS, a biomarker of M1-type polarization [136]. A summary of the aspects of IBD and AD is presented in Table 1.
Pro-opiomelanocortin (POMC), a common protein precursor, is the source of the complex and phylogenetically old melanocortin system, which includes α, β, and γ-melanocyte-stimulating hormone (MSH) and adrenocorticotropic hormone (ACTH). Post-translational modification of POMC yields precursor peptides (γ-MSH, ACTH, and β-lipotropins) that can be further changed by acetylation, amidation, and glycosylation to produce active mediators. Specifically, the result of the precursor convertase 2 enzyme’s activity on ACTH, an enzyme substrate, is the α-melanotropin group [137,138]. The processes involved with melanocortins include the regulation of melanogenesis, steroidogenesis, neuromodulation, and the modulation of inflammatory processes. In recent years, the system has come to be recognized as significant in cases of persistent inflammatory illnesses of the digestive tract, such as inflammatory bowel diseases, for which a wealth of data has been gathered using colitis-modeling mice. In fact, details about how this kind of mechanism might mitigate the inflammation caused by colitis and impact the intricate cytokine imbalance in the intestinal milieu damaged by persistent inflammation have surfaced. The central nervous system’s main melanocortin receptor is called MC4R [139]. The brainstem, spinal cord, hypothalamus, and cerebral cortex are where it is primarily expressed. Its distribution is mostly nonoverlapping and extends farther throughout the CNS than MC3R. However, the coexpression of MC3R and MC4R is seen in the posterior hypothalamus nucleus, as well as in the ventral periventricular and premammillary nuclei [140]. Because melanocortins are located in the brainstem, they can reduce the expression of proinflammatory cytokines in endotoxemia, sepsis, and other inflammatory diseases by activating various anti-inflammatory pathways via the efferent vagal pathway [137,141,142]. However, preliminary immunohistochemistry data showed that compared to healthy mucosa, disease-affected CD/UC tracts had significantly greater levels of MC3R expression (together with MC4R expression). Thus, the study demonstrated that these receptors are expressed at the intestinal level in IBD patients and that their expression varies according to the severity of the disease [143]. In fact, MC4R’s function and peripheral localizations are becoming more apparent despite its greater central localization. The expression of MC4R has been demonstrated by Panaro et al. in many gastrointestinal tract segments, including the stomach, small intestine, and descending colon. The mRNA encoding for this receptor was highly expressed in a number of cytotypes, including small intestinal cells positive for gastric inhibitory peptide, glucagon-like peptide 1, and cholecystokinin in mice. Its actions appear to be related to a paracrine suppression of electrolyte secretion, which can be stimulated by administering α-MSH [144]. The MC4R has been the subject of research inside the hippocampal area, particularly in relation to AD. Via the cAMP-PKA signaling pathway, it has been demonstrated that the MC4R controls hippocampus synaptic plasticity in both healthy mice and an AD animal model [145,146]. Subsequent research revealed that in several AD mice models, MC4R activation prevents the illness from progressing [147,148,149].

4. AD and Rheumatoid Arthritis

One definition of rheumatoid arthritis is a degenerative condition affecting the skeleton. A systemic inflammatory response that damages articular cartilage and bones is a defining feature of the illness [150]. Immune cells typically have a cascading role in the illness process, engaging subsequent cells and mediators. The immune system’s involvement is linked to the activation of the right cells, which causes the release of inflammatory cytokines and matrix metalloproteinases (MMPs) [151]. In the end, these continuing processes lead to the development of excruciating joint swelling and function impairment [152]. The risk factors for RA are not adequately described. There have been suggestions linking the illness to both external and hereditary factors. The etiology of RA is initially associated with CD4+ lymphocytes. These cells activate monocytes, macrophages, and synovial fibroblasts by identifying antigens in the synovial tissue. The cells listed above release metalloproteinases, which contribute to the deterioration of bone and cartilage. These immune cells produce interleukin (IL)-1, IL-6, and TNF-α, which are responsible for the primary inflammatory response in RA, in addition to their degradative activity. Eventually, synovitis—a thicker and swollen tissue—is the result of the complete cascade process [153].
Rheumatoid factor (RF), anti-cyclic citrullinated peptide (anti-CCP), C-reactive protein (CRP), amyloid A protein, and calgranulin are among the suitable biomarkers that are detected in the serum of RA patients [154]. Since amyloid deposition in soft tissues is a result of amyloidosis of the light chains of transthyretin and immunoglobulins, the existence of amyloid structures is particularly interesting. Orthopedic problems are, in fact, directly caused by the accumulating process [155]. According to one study, amyloid deposits have been seen during orthopedic surgeries, particularly in patients older than 70 [156]. Furthermore, research by Donelly et al. shows that tendon sheath-positive markers of amyloid tissue are present in 10% of men over 50 and women over 60 following biopsy [157]. Further research is required on protein amyloid A (SAA, serum amyloid A), which is produced by liver cells. Increased production of SAA is observed in hepatocytes activated by pro-inflammatory cytokines such as TNF-α, IL-1, and IL-6. A study found that some disorders associated with inflammatory pathology can have an excess of SAA. These illnesses include persistent inflammation, which can be caused by long-term infections such as rheumatoid arthritis, osteomyelitis, and tuberculosis, as well as inflammatory bowel disease, genetic disorders, and solid and hematopoietic neoplasms [158]. It is important to remember that pro-inflammatory cytokines are pleiotropic mediators since they stimulate enhanced angiogenesis, the breakdown of connective tissue binder in RA, and increased amyloid activity [159].
Persistent systemic peripheral inflammation affects the AD-specific neurodegenerative processes. When comparing AD patients to healthy controls, it is evident that inflammatory cytokines such as TNF-α, IL-6, IL-1β, TGF-β, IL-12, and IL-18 are active [160]. Because the over-reactivity of the immune system is a common hallmark of both AD and RA, the cytokines described above and their influence are being researched in the etiology of these disorders. It should be taken into consideration when assessing the positive link between AD and RA that AD is far more common in RA patients than in the general population [161]. In a different study, individuals with rheumatic conditions—particularly RA—were shown to have higher cognitive loss as they aged [162]. Anti-inflammatory medication activity has been used to study the consequences of systemic inflammation. Commonly prescribed medications for RA patients—methotrexate and non-steroidal anti-inflammatory medicines (NSAIDs)—lower the incidence of dementia linked to AD, particularly when taken early in the course of the illness [163,164,165]. However, The Alzheimer’s Disease Anti-inflammatory Prevention Trial Follow-up Study (ADAPT-FS) showed that the results do not lend credence to the idea that among persons with a family history of dementia, celecoxib or naproxen prevent AD [166]. It is generally accepted that immune system hyperactivity is the link between these conditions, but more investigation is required to pinpoint the precise relationship between RA and AD. Dysregulation of genes that repress the cell cycle is a common characteristic of both AD and RA, and it also raises the risk of systemic inflammation. Research indicates that age-related alterations in the cell cycle are strongly regulated and that inflammatory changes influence the emergence of pathogenic changes in both illnesses [167].
The existence of coexisting conditions in patients, such as diabetes, peripheral vascular disease, hyperlipidemia, hypertension, and coronary artery disease, was considered in the Chou et al. study [168]. Overall, it was shown that the incidence of AD in individuals with RA was higher than the incidence in individuals without RA. Furthermore, it was demonstrated that among individuals with RA, the relative risk of AD was dramatically elevated by chronic diseases such as diabetes, vascular disease, and coronary artery disease. Interestingly, anti-rheumatic medication therapy has been shown by other research to lower the risk of dementia [169]. Researchers found that cDMARD users had a lower risk of dementia than people who did not take these drugs. Additionally, those on cDMARD had a lower chance of developing dementia following medication use. The usage of MTX (metotrexat) produced the greatest result. Dementia risk was significantly lower after five years of treatment and twice as low at fifteen years. The study’s authors note significant limitations in spite of the findings. Individuals with comorbid conditions such as peripheral vascular disease, interstitial lung disease, anemia, osteoporosis, myocardial infarction, congestive heart failure, and cardiovascular disease were included in the study. Prior to beginning RA treatment, the patients were on statins, analgesics, antihypertensive medications, or proton pump inhibitors [169].
Neurodegenerative abnormalities have an impact on the development of systemic inflammation. The immune system’s activity, which produces inflammatory biomarkers, shows similarities between the pathomechanisms of AD and RA. Numerous bodily functions are mediated by cytokines, which also have an impact on the blood–brain barrier’s integrity and tightness, as well as severely suppress the production of occluding-forming junctions. Additionally, the pathology of amyloid is linked to AD and RA diseases. Aβ plaques in AD make neurons more vulnerable to excitotoxicity, loss of synaptic protein, and cholinergic transmission; in RA, on the other hand, cytokine-stimulated amyloid plays a role in the bone–joint bond’s deterioration. Compared to healthy individuals, RA patients had a much greater incidence of AD. The relationship between the immunological, skeletal, and neurological systems and the organism’s aging processes clarifies the existence of a complex web of interactions. Even though there are some similarities, this mention needs to be handled very carefully. It is possible that the existence of inflammatory markers does not always show a direct cause-and-effect connection between these conditions. It is possible that AD and RA have distinct inflammatory pathways and that the markers listed are the only thing they have in common. To clarify the potential pathways of the immune cascade in both AD and RA and in both diseases concurrently, more research in this area is necessary [170].

5. Conclusions

This paper highlights the intricate connections between Alzheimer’s disease, diabetes, rheumatoid arthritis, and inflammatory bowel diseases, emphasizing the shared mechanisms of metabolic and immune system dysregulation. These seemingly distinct conditions are interwoven through complex interactions between metabolic dysfunction, chronic inflammation, and immune dysregulation, providing insights into their shared etiologies. The metabolic pathways involved in insulin resistance, lipid metabolism, and oxidative stress play significant roles in the pathogenesis of these diseases. For instance, insulin resistance, a hallmark of diabetes, has been implicated in Alzheimer’s disease, where it exacerbates amyloid-beta accumulation and tau phosphorylation. Similarly, lipid metabolism dysregulation is common in rheumatoid arthritis and inflammatory bowel diseases, contributing to systemic inflammation and tissue damage. Chronic inflammation, driven by immune system dysregulation, serves as a critical link between these conditions. The inflammatory cascade, characterized by pro-inflammatory cytokines and immune cell activation, is a shared feature of Alzheimer’s disease, rheumatoid arthritis, and inflammatory bowel diseases. This persistent inflammation not only drives disease progression but also contributes to the development of comorbidities, such as cardiovascular disease, which are prevalent across these conditions. Furthermore, the gut–brain axis emerges as a key player in understanding the connections between these diseases. The bidirectional communication between the gut microbiota and the central nervous system influences both metabolic and immune processes. Dysbiosis, or microbial imbalance, is observed in Alzheimer’s disease and the other diseases described in this paper, suggesting that targeting the gut microbiota could be a promising therapeutic approach.
Ultimately, a deeper understanding of the pathophysiological mechanisms shared by these diseases can revolutionize how their treatment and management are approached. By focusing on the commonalities, we can develop holistic, multi-targeted therapies that address the underlying causes rather than just the symptoms of these conditions. This integrative approach is promising for improving patient outcomes and quality of life across a spectrum of chronic diseases, ushering in a new era of personalized and precise medical care. The diseases discussed in the paper involve complex networks of interactions. These include genetic predispositions, environmental triggers, immune responses, and metabolic processes. The challenge lies in disentangling these overlapping factors to identify specific pathways that contribute to each disease. The interactions between inflammation, immune dysregulation, and metabolic changes are not fully understood, making it difficult to establish clear causal links. Additionally, more longitudinal studies are crucial for understanding how metabolic and immune dysregulation contributes to the onset and progression of these diseases. Without long-term data, it is difficult to determine how early metabolic or immune changes influence the development of these conditions. Another aspect is a huge need for biomarker research. However, the complexity of multifactorial interactions, the scarcity of longitudinal studies, difficulties in biomarker identification and validation, and the limitations of animal models pose significant hurdles. Overcoming these challenges requires a multifaceted approach, including the development of more sophisticated research methodologies, greater investment in long-term studies, and enhanced collaboration between basic scientists and clinicians. Only by addressing these limitations can we hope to unravel the intricate connections between these diseases and develop more effective diagnostic and therapeutic strategies.

Author Contributions

Conceptualization: J.D. and B.M.; Data curation: J.D. and J.M.; Writing—original draft: J.D. and J.M.; Visualization: J.D.; Funding acquisition: J.D. and B.M.; Supervision: I.W. and B.M.; Investigation: I.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical University of Bialystok, grant number B.SUB.24.166, B.SUB.24.503 and B.SUB.24.297.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

B.M. has received consultation and/or lecture honoraria from Abbott, Wiener, Roche, Cormay, Biameditek, and TK Biotech. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Heneka, M.T.; Carson, M.J.; Khoury, J.E.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s Disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [PubMed]
  2. Kanashiro, A.; Hiroki, C.H.; da Fonseca, D.M.; Birbrair, A.; Ferreira, R.G.; Bassi, G.S.; Fonseca, M.D.; Kusuda, R.; Cebinelli, G.C.M.; da Silva, K.P.; et al. The Role of Neutrophils in Neuro-Immune Modulation. Pharmacol. Res. 2020, 151, 104580. [Google Scholar] [CrossRef] [PubMed]
  3. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of Neuroinflammation in Neurodegeneration Development. Signal Transduct. Target. Ther. 2023, 8, 267. [Google Scholar] [CrossRef]
  4. Garwood, C.J.; Pooler, A.M.; Atherton, J.; Hanger, D.P.; Noble, W. Astrocytes Are Important Mediators of Aβ-Induced Neurotoxicity and Tau Phosphorylation in Primary Culture. Cell Death Dis. 2011, 2, e167. [Google Scholar] [CrossRef]
  5. Carrero, I.; Gonzalo, M.R.; Martin, B.; Sanz-Anquela, J.M.; Arévalo-Serrano, J.; Gonzalo-Ruiz, A. Oligomers of β-Amyloid Protein (Aβ1-42) Induce the Activation of Cyclooxygenase-2 in Astrocytes via an Interaction with Interleukin-1β, Tumour Necrosis Factor-α, and a Nuclear Factor κ-B Mechanism in the Rat Brain. Exp. Neurol. 2012, 236, 215–227. [Google Scholar] [CrossRef]
  6. Venegas, C.; Kumar, S.; Franklin, B.S.; Dierkes, T.; Brinkschulte, R.; Tejera, D.; Vieira-Saecker, A.; Schwartz, S.; Santarelli, F.; Kummer, M.P.; et al. Microglia-Derived ASC Specks Cross-Seed Amyloid-β in Alzheimer’s Disease. Nature 2017, 552, 355–361. [Google Scholar] [CrossRef] [PubMed]
  7. Frank-Cannon, T.C.; Alto, L.T.; McAlpine, F.E.; Tansey, M.G. Does Neuroinflammation Fan the Flame in Neurodegenerative Diseases? Mol. Neurodegener. 2009, 4, 47. [Google Scholar] [CrossRef]
  8. Iliff, J.J.; Wang, M.; Liao, Y.; Plogg, B.A.; Peng, W.; Gundersen, G.A.; Benveniste, H.; Vates, G.E.; Deane, R.; Goldman, S.A.; et al. A Paravascular Pathway Facilitates CSF Flow Through the Brain Parenchyma and the Clearance of Interstitial Solutes, Including Amyloid β. Sci. Transl. Med. 2012, 4, 147ra111. [Google Scholar] [CrossRef]
  9. Da Mesquita, S.; Louveau, A.; Vaccari, A.; Smirnov, I.; Cornelison, R.C.; Kingsmore, K.M.; Contarino, C.; Onengut-Gumuscu, S.; Farber, E.; Raper, D.; et al. Functional Aspects of Meningeal Lymphatics in Aging and Alzheimer’s Disease. Nature 2018, 560, 185. [Google Scholar] [CrossRef]
  10. He, X.F.; Liu, D.X.; Zhang, Q.; Liang, F.Y.; Dai, G.Y.; Zeng, J.S.; Pei, Z.; Xu, G.Q.; Lan, Y. Voluntary Exercise Promotes Glymphatic Clearance of Amyloid Beta and Reduces the Activation of Astrocytes and Microglia in Aged Mice. Front. Mol. Neurosci. 2017, 10, 144. [Google Scholar] [CrossRef]
  11. Harrison, I.F.; Ismail, O.; Machhada, A.; Colgan, N.; Ohene, Y.; Nahavandi, P.; Ahmed, Z.; Fisher, A.; Meftah, S.; Murray, T.K.; et al. Impaired Glymphatic Function and Clearance of Tau in an Alzheimer’s Disease Model. Brain 2020, 143, 2576. [Google Scholar] [CrossRef] [PubMed]
  12. Iliff, J.J.; Chen, M.J.; Plog, B.A.; Zeppenfeld, D.M.; Soltero, M.; Yang, L.; Singh, I.; Deane, R.; Nedergaard, M. Impairment of Glymphatic Pathway Function Promotes Tau Pathology after Traumatic Brain Injury. J. Neurosci. 2014, 34, 16180. [Google Scholar] [CrossRef]
  13. He, X.F.; Li, L.L.; Xian, W.B.; Li, M.Y.; Zhang, L.Y.; Xu, J.H.; Pei, Z.; Zheng, H.Q.; Hu, X. Quan Chronic Colitis Exacerbates NLRP3-Dependent Neuroinflammation and Cognitive Impairment in Middle-Aged Brain. J. Neuroinflammation 2021, 18, 153. [Google Scholar] [CrossRef] [PubMed]
  14. Clarke, L.E.; Liddelow, S.A.; Chakraborty, C.; Münch, A.E.; Heiman, M.; Barres, B.A. Normal Aging Induces A1-like Astrocyte Reactivity. Proc. Natl. Acad. Sci. USA 2018, 115, E1896–E1905. [Google Scholar] [CrossRef]
  15. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic Reactive Astrocytes Are Induced by Activated Microglia. Nature 2017, 541, 481. [Google Scholar] [CrossRef] [PubMed]
  16. Diabetes Facts and Figures|International Diabetes Federation. Available online: https://idf.org/about-diabetes/diabetes-facts-figures/ (accessed on 15 July 2024).
  17. Chen, Y.; Yu, Q.; Gong, C.X. Molecular Connection Between Diabetes and Dementia. Adv. Exp. Med. Biol. 2019, 1128, 103–131. [Google Scholar] [CrossRef] [PubMed]
  18. Kubis-Kubiak, A.M.; Rorbach-Dolata, A.; Piwowar, A. Crucial Players in Alzheimer’s Disease and Diabetes Mellitus: Friends or Foes? Mech. Ageing Dev. 2019, 181, 7–21. [Google Scholar] [CrossRef]
  19. Gudala, K.; Bansal, D.; Schifano, F.; Bhansali, A. Diabetes Mellitus and Risk of Dementia: A Meta-Analysis of Prospective Observational Studies. J. Diabetes Investig. 2013, 4, 640–650. [Google Scholar] [CrossRef]
  20. Rorbach-Dolata, A.; Piwowar, A. Neurometabolic Evidence Supporting the Hypothesis of Increased Incidence of Type 3 Diabetes Mellitus in the 21st Century. Biomed. Res. Int. 2019, 2019, 1435276. [Google Scholar] [CrossRef]
  21. Huang, C.C.; Chung, C.M.; Leu, H.B.; Lin, L.Y.; Chiu, C.C.; Hsu, C.Y.; Chiang, C.H.; Huang, P.H.; Chen, T.J.; Lin, S.J.; et al. Diabetes Mellitus and the Risk of Alzheimer’s Disease: A Nationwide Population-Based Study. PLoS ONE 2014, 9, e87095. [Google Scholar] [CrossRef]
  22. Ramos-Rodriguez, J.J.; Molina-Gil, S.; Ortiz-Barajas, O.; Jimenez-Palomares, M.; Perdomo, G.; Cozar-Castellano, I.; Lechuga-Sancho, A.M.; Garcia-Alloza, M. Central Proliferation and Neurogenesis Is Impaired in Type 2 Diabetes and Prediabetes Animal Models. PLoS ONE 2014, 9, e89229. [Google Scholar] [CrossRef]
  23. Gage, F.H. Mammalian Neural Stem Cells. Science 2000, 287, 1433–1438. [Google Scholar] [CrossRef] [PubMed]
  24. Lang, B.T.; Yan, Y.; Dempsey, R.J.; Vemuganti, R. Impaired Neurogenesis in Adult Type-2 Diabetic Rats. Brain Res. 2009, 1258, 25–33. [Google Scholar] [CrossRef] [PubMed]
  25. Beauquis, J.; Saravia, F.; Coulaud, J.; Roig, P.; Dardenne, M.; Homo-Delarche, F.; De Nicola, A. Prominently Decreased Hippocampal Neurogenesis in a Spontaneous Model of Type 1 Diabetes, the Nonobese Diabetic Mouse. Exp. Neurol. 2008, 210, 359–367. [Google Scholar] [CrossRef]
  26. Guo, J.; Yu, C.; Li, H.; Liu, F.; Feng, R.; Wang, H.; Meng, Y.; Li, Z.; Ju, G.; Wang, J. Impaired Neural Stem/Progenitor Cell Proliferation in Streptozotocin-Induced and Spontaneous Diabetic Mice. Neurosci. Res. 2010, 68, 329–336. [Google Scholar] [CrossRef] [PubMed]
  27. Li, J.; Tang, Y.; Cai, D. IKKβ/NF-ΚB Disrupts Adult Hypothalamic Neural Stem Cells to Mediate Neurodegenerative Mechanism of Dietary Obesity and Pre-Diabetes. Nat. Cell Biol. 2012, 14, 999. [Google Scholar] [CrossRef] [PubMed]
  28. Qin, L.M.; Bouchard, R.; Pugazhenthi, S. Regulation of Cyclic AMP Response Element-Binding Protein during Neuroglial Interactions. J. Neurochem. 2016, 136, 918–930. [Google Scholar] [CrossRef]
  29. Cukierman-Yaffe, T.; Gerstein, H.C.; Williamson, J.D.; Lazar, R.M.; Lovato, L.; Miller, M.E.; Coker, L.H.; Murray, A.; Sullivan, M.D.; Marcovina, S.M.; et al. Relationship Between Baseline Glycemic Control and Cognitive Function in Individuals With Type 2 Diabetes and Other Cardiovascular Risk Factors: The Action to Control Cardiovascular Risk in Diabetes-Memory in Diabetes (ACCORD-MIND) Trial. Diabetes Care 2009, 32, 221. [Google Scholar] [CrossRef]
  30. Yaffe, K.; Blackwell, T.; Whitmer, R.A.; Krueger, K.; Barrett-Connor, E. Glycosylated Hemoglobin Level and Development of Mild Cognitive Impairment or Dementia in Older Women. J. Nutr. Health Aging 2006, 10, 293–295. [Google Scholar]
  31. Ohara, T.; Doi, Y.; Ninomiya, T.; Hirakawa, Y.; Hata, J.; Iwaki, T.; Kanba, S.; Kiyohara, Y. Glucose Tolerance Status and Risk of Dementia in the Community: The Hisayama Study. Neurology 2011, 77, 1126–1134. [Google Scholar] [CrossRef]
  32. Ding, J.; Strachan, M.W.J.; Reynolds, R.M.; Frier, B.M.; Deary, I.J.; Fowkes, F.G.R.; Lee, A.J.; McKnight, J.; Halpin, P.; Swa, K.; et al. Diabetic Retinopathy and Cognitive Decline in Older People with Type 2 Diabetes: The Edinburgh Type 2 Diabetes Study. Diabetes 2010, 59, 2883–2889. [Google Scholar] [CrossRef]
  33. Crane, P.K.; Walker, R.; Hubbard, R.A.; Li, G.; Nathan, D.M.; Zheng, H.; Haneuse, S.; Craft, S.; Montine, T.J.; Kahn, S.E.; et al. Glucose Levels and Risk of Dementia. N. Engl. J. Med. 2013, 369, 540. [Google Scholar] [CrossRef] [PubMed]
  34. Sommerfield, A.J.; Deary, I.J.; Frier, B.M. Acute Hyperglycemia Alters Mood State and Impairs Cognitive Performance in People with Type 2 Diabetes. Diabetes Care 2004, 27, 2335–2340. [Google Scholar] [CrossRef] [PubMed]
  35. Spauwen, P.J.J.; Köhler, S.; Verhey, F.R.J.; Stehouwer, C.D.A.; Van Boxtel, M.P.J. Effects of Type 2 Diabetes on 12-Year Cognitive Change: Results from the Maastricht Aging Study. Diabetes Care 2013, 36, 1554–1561. [Google Scholar] [CrossRef] [PubMed]
  36. Cukierman, T.; Gerstein, H.C.; Williamson, J.D. Cognitive Decline and Dementia in Diabetes--Systematic Overview of Prospective Observational Studies. Diabetologia 2005, 48, 2460–2469. [Google Scholar] [CrossRef]
  37. Ravona-Springer, R.; Moshier, E.; Schmeidler, J.; Godbold, J.; Akrivos, J.; Rapp, M.; Grossman, H.T.; Wysocki, M.; Silverman, J.M.; Haroutunian, V.; et al. Changes in Glycemic Control Are Associated with Changes in Cognition in Non-Diabetic Elderly. J. Alzheimers Dis. 2012, 30, 299. [Google Scholar] [CrossRef]
  38. Ravona-Springer, R.; Heymann, A.; Schmeidler, J.; Moshier, E.; Godbold, J.; Sano, M.; Leroith, D.; Johnson, S.; Preiss, R.; Koifman, K.; et al. Trajectories in Glycemic Control over Time Are Associated with Cognitive Performance in Elderly Subjects with Type 2 Diabetes. PLoS ONE 2014, 9, e97384. [Google Scholar] [CrossRef]
  39. Sue Kirkman, M.; Briscoe, V.J.; Clark, N.; Florez, H.; Haas, L.B.; Halter, J.B.; Huang, E.S.; Korytkowski, M.T.; Munshi, M.N.; Odegard, P.S.; et al. Diabetes in Older Adults: Consensus Report. J. Am. Geriatr. Soc. 2012, 60, 2342. [Google Scholar] [CrossRef] [PubMed]
  40. Pugazhenthi, S.; Qin, L.; Reddy, P.H. Common Neurodegenerative Pathways in Obesity, Diabetes, and Alzheimer’s Disease. Biochim. Biophys. Acta 2017, 1863, 1037. [Google Scholar] [CrossRef]
  41. Karmi, A.; Iozzo, P.; Viljanen, A.; Hirvonen, J.; Fielding, B.A.; Virtanen, K.; Oikonen, V.; Kemppainen, J.; Viljanen, T.; Guiducci, L.; et al. Increased Brain Fatty Acid Uptake in Metabolic Syndrome. Diabetes 2010, 59, 2171. [Google Scholar] [CrossRef]
  42. Baker, L.D.; Cross, D.J.; Minoshima, S.; Belongia, D.; Stennis Watson, G.; Craft, S. Insulin resistance is associated with alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with pre-diabetes or early type 2 diabetes. Arch. Neurol. 2011, 68, 51. [Google Scholar] [CrossRef]
  43. Sȩdzikowska, A.; Szablewski, L. Insulin and Insulin Resistance in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 9987. [Google Scholar] [CrossRef] [PubMed]
  44. Neth, B.J.; Craft, S. Insulin Resistance and Alzheimer’s Disease: Bioenergetic Linkages. Front. Aging Neurosci. 2017, 9, 345. [Google Scholar] [CrossRef] [PubMed]
  45. Moult, P.R.; Harvey, J. Hormonal Regulation of Hippocampal Dendritic Morphology and Synaptic Plasticity. Cell Adhes. Migr. 2008, 2, 269. [Google Scholar] [CrossRef] [PubMed]
  46. Van Der Heide, L.P.; Kamal, A.; Artola, A.; Gispen, W.H.; Ramakers, G.M.J. Insulin Modulates Hippocampal Activity-Dependent Synaptic Plasticity in a N-Methyl-d-Aspartate Receptor and Phosphatidyl-Inositol-3-Kinase-Dependent Manner. J. Neurochem. 2005, 94, 1158–1166. [Google Scholar] [CrossRef]
  47. Gratuze, M.; Joly-Amado, A.; Vieau, D.; Buée, L.; Blum, D. Mutual Relationship between Tau and Central Insulin Signalling: Consequences for AD and Tauopathies? Neuroendocrinology 2018, 107, 181–195. [Google Scholar] [CrossRef]
  48. Skeberdis, V.A.; Lan, J.Y.; Zheng, X.; Zukin, R.S.; Bennett, M.V.L. Insulin Promotes Rapid Delivery of N-Methyl-d- Aspartate Receptors to the Cell Surface by Exocytosis. Proc. Natl. Acad. Sci. USA 2001, 98, 3561. [Google Scholar] [CrossRef]
  49. Wu, S.P.; Lu, K.T.; Chang, W.C.; Gean, P.W. Involvement of Mitogen-Activated Protein Kinase in Hippocampal Long-Term Potentiation. J. Biomed. Sci. 1999, 6, 409–417. [Google Scholar] [CrossRef]
  50. Sanna, P.P.; Cammalleri, M.; Berton, F.; Simpson, C.; Lutjens, R.; Bloom, F.E.; Francesconi, W. Phosphatidylinositol 3-Kinase Is Required for the Expression But Not for the Induction or the Maintenance of Long-Term Potentiation in the Hippocampal CA1 Region. J. Neurosci. 2002, 22, 3359. [Google Scholar] [CrossRef]
  51. Abbott, M.A.; Wells, D.G.; Fallon, J.R. The Insulin Receptor Tyrosine Kinase Substrate P58/53 and the Insulin Receptor Are Components of CNS Synapses. J. Neurosci. 1999, 19, 7300. [Google Scholar] [CrossRef]
  52. Spinelli, M.; Fusco, S.; Mainardi, M.; Scala, F.; Natale, F.; Lapenta, R.; Mattera, A.; Rinaudo, M.; Li Puma, D.D.; Ripoli, C.; et al. Brain Insulin Resistance Impairs Hippocampal Synaptic Plasticity and Memory by Increasing GluA1 Palmitoylation through FoxO3a. Nat. Commun. 2017, 8, 2009. [Google Scholar] [CrossRef] [PubMed]
  53. Nelson, T.J.; Sun, M.K.; Hongpaisan, J.; Alkon, D.L. Insulin, PKC Signaling Pathways and Synaptic Remodeling during Memory Storage and Neuronal Repair. Eur. J. Pharmacol. 2008, 585, 76–87. [Google Scholar] [CrossRef]
  54. van der Heide, L.P.; Ramakers, G.M.J.; Smidt, M.P. Insulin Signaling in the Central Nervous System: Learning to Survive. Prog. Neurobiol. 2006, 79, 205–221. [Google Scholar] [CrossRef] [PubMed]
  55. Kaur, S.; Singh, S.; Arora, A.; Ram, P.; Kumar, S.; Kumar, P.; Abed, S.N. Pharmacology of GABA and Its Receptors. In Frontiers in Pharmacology of Neurotransmitters; Springer: Singapore, 2020; pp. 241–292. [Google Scholar] [CrossRef]
  56. Sandoval-Salazar, C.; Ramírez-Emiliano, J.; Trejo-Bahena, A.; Oviedo-Solís, C.I.; Solís-Ortiz, M.S. A High-Fat Diet Decreases GABA Concentration in the Frontal Cortex and Hippocampus of Rats. Biol. Res. 2016, 49, 15. [Google Scholar] [CrossRef] [PubMed]
  57. Arnold, S.E.; Arvanitakis, Z.; Macauley-Rambach, S.L.; Koenig, A.M.; Wang, H.Y.; Ahima, R.S.; Craft, S.; Gandy, S.; Buettner, C.; Stoeckel, L.E.; et al. Brain Insulin Resistance in Type 2 Diabetes and Alzheimer Disease: Concepts and Conundrums. Nat. Rev. Neurol. 2018, 14, 168. [Google Scholar] [CrossRef]
  58. Strachan, M.W.J.; Reynolds, R.M.; Marioni, R.E.; Price, J.F. Cognitive Function, Dementia and Type 2 Diabetes Mellitus in the Elderly. Nat. Rev. Endocrinol. 2011, 7, 108–114. [Google Scholar] [CrossRef]
  59. Wang, J.; Gong, B.; Zhao, W.; Tang, C.; Varghese, M.; Nguyen, T.; Bi, W.; Bilski, A.; Begum, S.; Vempati, P.; et al. Epigenetic Mechanisms Linking Diabetes and Synaptic Impairments. Diabetes 2014, 63, 645–654. [Google Scholar] [CrossRef]
  60. Qiu, W.Q.; Folstein, M.F. Insulin, Insulin-Degrading Enzyme and Amyloid-Beta Peptide in Alzheimer’s Disease: Review and Hypothesis. Neurobiol. Aging 2006, 27, 190–198. [Google Scholar] [CrossRef]
  61. Haque, R.; Nazir, A. Insulin-Degrading Enzyme: A Link between Alzheimer’s and Type 2 Diabetes Mellitus. CNS Neurol. Disord. Drug Targets 2014, 13, 259–264. [Google Scholar] [CrossRef]
  62. Akter, K.; Lanza, E.A.; Martin, S.A.; Myronyuk, N.; Rua, M.; Raffa, R.B. Diabetes Mellitus and Alzheimer’s Disease: Shared Pathology and Treatment? Br. J. Clin. Pharmacol. 2011, 71, 365. [Google Scholar] [CrossRef]
  63. Janson, J.; Laedtke, T.; Parisi, J.E.; O’Brien, P.; Petersen, R.C.; Butler, P.C. Increased Risk of Type 2 Diabetes in Alzheimer Disease. Diabetes 2004, 53, 474–481. [Google Scholar] [CrossRef] [PubMed]
  64. Maj, M.; Hoermann, G.; Rasul, S.; Base, W.; Wagner, L.; Attems, J. The Microtubule-Associated Protein Tau and Its Relevance for Pancreatic Beta Cells. J. Diabetes Res. 2016, 2016, 1964634. [Google Scholar] [CrossRef] [PubMed]
  65. Martinez-Valbuena, I.; Valenti-Azcarate, R.; Amat-Villegas, I.; Riverol, M.; Marcilla, I.; de Andrea, C.E.; Sánchez-Arias, J.A.; del Mar Carmona-Abellan, M.; Marti, G.; Erro, M.E.; et al. Amylin as a Potential Link between Type 2 Diabetes and Alzheimer Disease. Ann. Neurol. 2019, 86, 539–551. [Google Scholar] [CrossRef] [PubMed]
  66. Toledo, J.B.; Arnold, M.; Kastenmüller, G.; Chang, R.; Baillie, R.A.; Han, X.; Thambisetty, M.; Tenenbaum, J.D.; Suhre, K.; Thompson, J.W.; et al. Metabolic Network Failures in Alzheimer’s Disease: A Biochemical Road Map. Alzheimers Dement. 2017, 13, 965–984. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, Z.; Zhong, C. Decoding Alzheimer’s Disease from Perturbed Cerebral Glucose Metabolism: Implications for Diagnostic and Therapeutic Strategies. Prog. Neurobiol. 2013, 108, 21–43. [Google Scholar] [CrossRef] [PubMed]
  68. De La Monte, S.M.; Tong, M. Brain Metabolic Dysfunction at the Core of Alzheimer’s Disease. Biochem. Pharmacol. 2014, 88, 548–559. [Google Scholar] [CrossRef]
  69. Diniz Pereira, J.; Gomes Fraga, V.; Morais Santos, A.L.; Carvalho, M.D.G.; Caramelli, P.; Braga Gomes, K. Alzheimer’s Disease and Type 2 Diabetes Mellitus: A Systematic Review of Proteomic Studies. J. Neurochem. 2021, 156, 753–776. [Google Scholar] [CrossRef]
  70. Yoshino, S.; Fujimoto, K.; Takada, T.; Kawamura, S.; Ogawa, J.; Kamata, Y.; Kodera, Y.; Shichiri, M. Molecular Form and Concentration of Serum A2-Macroglobulin in Diabetes. Sci. Rep. 2019, 9, 12927. [Google Scholar] [CrossRef]
  71. Chung, T.J.; Hsu, K.Y.; Chen, J.H.; Liu, J.S.; Chang, H.W.; Li, P.F.; Huang, C.L.; Shieh, Y.S.; Lee, C.H. Association of Salivary Alpha 2-Macroglobulin Levels and Clinical Characteristics in Type 2 Diabetes. J. Diabetes Investig. 2016, 7, 190–196. [Google Scholar] [CrossRef]
  72. Gravina, A.; Panarese, I.; Trotta, M.; Pellegrino, R.; Ferraraccio, F.; Galdiero, M.; Alfano, R.; Loguercio, C.; Romano, M.; Grieco, P.; et al. Relationship between Salivary Alpha-2 Macroglobulin and HbA1c among Patients with Type-2 Diabetes Mellitus: A Cross-Sectional Study. Indian. J. Endocrinol. Metab. 2019, 23, 184–187. [Google Scholar] [CrossRef]
  73. James, K.; Merriman, J.; Gray, R.S.; Duncan, L.J.; Herd, R. Serum Alpha 2-Macroglobulin Levels in Diabetes. J. Clin. Pathol. 1980, 33, 163–166. [Google Scholar] [CrossRef]
  74. Seddighi, S.; Varma, V.; Thambisetty, M. A2-Macroglobulin in Alzheimer’s Disease: New Roles for an Old Chaperone. Biomark. Med. 2018, 12, 311–314. [Google Scholar] [CrossRef]
  75. Varma, V.R.; Varma, S.; An, Y.; Hohman, T.J.; Seddighi, S.; Casanova, R.; Beri, A.; Dammer, E.B.; Seyfried, N.T.; Pletnikova, O.; et al. Alpha-2 Macroglobulin in Alzheimer’s Disease: A Marker of Neuronal Injury through the RCAN1 Pathway. Mol. Psychiatry 2017, 22, 13–23. [Google Scholar] [CrossRef] [PubMed]
  76. Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms Underlying Inflammation in Neurodegeneration. Cell 2010, 140, 918. [Google Scholar] [CrossRef] [PubMed]
  77. Hanzel, C.E.; Pichet-Binette, A.; Pimentel, L.S.B.; Iulita, M.F.; Allard, S.; Ducatenzeiler, A.; Do Carmo, S.; Cuello, A.C. Neuronal Driven Pre-Plaque Inflammation in a Transgenic Rat Model of Alzheimer’s Disease. Neurobiol. Aging 2014, 35, 2249–2262. [Google Scholar] [CrossRef]
  78. Di Filippo, M.; Chiasserini, D.; Gardoni, F.; Viviani, B.; Tozzi, A.; Giampà, C.; Costa, C.; Tantucci, M.; Zianni, E.; Boraso, M.; et al. Effects of Central and Peripheral Inflammation on Hippocampal Synaptic Plasticity. Neurobiol. Dis. 2013, 52, 229–236. [Google Scholar] [CrossRef]
  79. Franceschi, C. Inflammaging as a Major Characteristic of Old People: Can It Be Prevented or Cured? Nutr. Rev. 2007, 65, S173–S176. [Google Scholar] [CrossRef] [PubMed]
  80. Shoelson, S.E.; Lee, J.; Goldfine, A.B. Inflammation and Insulin Resistance. J. Clin. Investig. 2006, 116, 1793. [Google Scholar] [CrossRef]
  81. Skelly, D.T.; Hennessy, E.; Dansereau, M.A.; Cunningham, C. A Systematic Analysis of the Peripheral and CNS Effects of Systemic LPS, IL-1Β, TNF-α and IL-6 Challenges in C57BL/6 Mice. PLoS ONE 2013, 8, e69123. [Google Scholar] [CrossRef]
  82. Krstic, D.; Madhusudan, A.; Doehner, J.; Vogel, P.; Notter, T.; Imhof, C.; Manalastas, A.; Hilfiker, M.; Pfister, S.; Schwerdel, C.; et al. Systemic Immune Challenges Trigger and Drive Alzheimer-like Neuropathology in Mice. J. Neuroinflammation 2012, 9, 151. [Google Scholar] [CrossRef]
  83. Swardfager, W.; Lanctt, K.; Rothenburg, L.; Wong, A.; Cappell, J.; Herrmann, N. A Meta-Analysis of Cytokines in Alzheimer’s Disease. Biol. Psychiatry 2010, 68, 930–941. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, R.; Miller, R.G.; Madison, C.; Jin, X.; Honrada, R.; Harris, W.; Katz, J.; Forshew, D.A.; McGrath, M.S. Systemic Immune System Alterations in Early Stages of Alzheimer’s Disease. J. Neuroimmunol. 2013, 256, 38. [Google Scholar] [CrossRef] [PubMed]
  85. Bermejo, P.; Martín-Aragón, S.; Benedí, J.; Susín, C.; Felici, E.; Gil, P.; Ribera, J.M.; Villar, Á.M. Differences of Peripheral Inflammatory Markers between Mild Cognitive Impairment and Alzheimer’s Disease. Immunol. Lett. 2008, 117, 198–202. [Google Scholar] [CrossRef]
  86. Koyama, A.; O’Brien, J.; Weuve, J.; Blacker, D.; Metti, A.L.; Yaffe, K. The Role of Peripheral Inflammatory Markers in Dementia and Alzheimer’s Disease: A Meta-Analysis. J. Gerontol. A Biol. Sci. Med. Sci. 2013, 68, 433. [Google Scholar] [CrossRef] [PubMed]
  87. Tan, Z.S.; Beiser, A.S.; Vasan, R.S.; Roubenoff, R.; Dinarello, C.A.; Harris, T.B.; Benjamin, E.J.; Au, R.; Kiel, D.P.; Wolf, P.A.; et al. Inflammatory Markers and the Risk of Alzheimer Disease: The Framingham Study. Neurology 2007, 68, 1902–1908. [Google Scholar] [CrossRef]
  88. Magaki, S.; Mueller, C.; Dickson, C.; Kirsch, W. Increased Production of Inflammatory Cytokines in Mild Cognitive Impairment. Exp. Gerontol. 2007, 42, 233–240. [Google Scholar] [CrossRef]
  89. Peng, L.; Xu, L.; Ouyang, W. Role of Peripheral Inflammatory Markers in Postoperative Cognitive Dysfunction (POCD): A Meta-Analysis. PLoS ONE 2013, 8, e79624. [Google Scholar] [CrossRef]
  90. Banks, W.A.; Kastin, A.J. Blood to Brain Transport of Interleukin Links the Immune and Central Nervous Systems. Life Sci. 1991, 48, PL117–PL121. [Google Scholar] [CrossRef]
  91. Gutierrez, E.G.; Banks, W.A.; Kastin, A.J. Murine Tumor Necrosis Factor Alpha Is Transported from Blood to Brain in the Mouse. J. Neuroimmunol. 1993, 47, 169–176. [Google Scholar] [CrossRef]
  92. Dilger, R.N.; Johnson, R.W. Aging, Microglial Cell Priming, and the Discordant Central Inflammatory Response to Signals from the Peripheral Immune System. J. Leukoc. Biol. 2008, 84, 932–939. [Google Scholar] [CrossRef]
  93. Thomson, C.A.; McColl, A.; Cavanagh, J.; Graham, G.J. Peripheral Inflammation Is Associated with Remote Global Gene Expression Changes in the Brain. J. Neuroinflammation 2014, 11, 73. [Google Scholar] [CrossRef] [PubMed]
  94. Fuschillo, G.; Celentano, V.; Rottoli, M.; Sciaudone, G.; Gravina, A.G.; Pellegrino, R.; Marfella, R.; Romano, M.; Selvaggi, F.; Pellino, G. Influence of Diabetes Mellitus on Inflammatory Bowel Disease Course and Treatment Outcomes. A Systematic Review with Meta-Analysis. Dig. Liver Dis. 2023, 55, 580–586. [Google Scholar] [CrossRef]
  95. Ordás, I.; Eckmann, L.; Talamini, M.; Baumgart, D.C.; Sandborn, W.J. Ulcerative Colitis. Lancet 2012, 380, 1606–1619. [Google Scholar] [CrossRef] [PubMed]
  96. Abraham, C.; Cho, J.H. Inflammatory Bowel Disease. N. Engl. J. Med. 2009, 361, 2066–2078. [Google Scholar] [CrossRef] [PubMed]
  97. Silverberg, M.S.; Satsangi, J.; Ahmad, T.; Arnott, I.D.R.; Bernstein, C.N.; Brant, S.R.; Caprilli, R.; Colombel, J.F.; Gasche, C.; Geboes, K.; et al. Toward an Integrated Clinical, Molecular and Serological Classification of Inflammatory Bowel Disease: Report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can. J. Gastroenterol. Hepatol. 2005, 19, 5A–36A. [Google Scholar] [CrossRef] [PubMed]
  98. Torres, J.; Mehandru, S.; Colombel, J.F.; Peyrin-Biroulet, L. Crohn’s Disease. Lancet 2017, 389, 1741–1755. [Google Scholar] [CrossRef]
  99. Zhang, B.; Wang, H.E.; Bai, Y.M.; Tsai, S.J.; Su, T.P.; Chen, T.J.; Wang, Y.P.; Chen, M.H. Inflammatory Bowel Disease Is Associated with Higher Dementia Risk: A Nationwide Longitudinal Study. Gut 2021, 70, 85–91. [Google Scholar] [CrossRef]
  100. Zingel, R.; Bohlken, J.; Kostev, K. Association Between Inflammatory Bowel Disease and Dementia: A Retrospective Cohort Study. J. Alzheimers Dis. 2021, 80, 1471–1478. [Google Scholar] [CrossRef]
  101. Rønnow Sand, J.; Troelsen, F.S.; Horváth-Puhó, E.; Henderson, V.W.; Sørensen, H.T.; Erichsen, R. Risk of Dementia in Patients with Inflammatory Bowel Disease: A Danish Population-Based Study. Aliment. Pharmacol. Ther. 2022, 56, 831–843. [Google Scholar] [CrossRef]
  102. Huang, J.; Su, B.; Karhunen, V.; Gill, D.; Zuber, V.; Ahola-Olli, A.; Palaniswamy, S.; Auvinen, J.; Herzig, K.H.; Keinänen-Kiukaanniemi, S.; et al. Inflammatory Diseases, Inflammatory Biomarkers, and Alzheimer Disease: An Observational Analysis and Mendelian Randomization. Neurology 2023, 100, 568–581. [Google Scholar] [CrossRef]
  103. Amoroso, C.; Perillo, F.; Strati, F.; Fantini, M.C.; Caprioli, F.; Facciotti, F. The Role of Gut Microbiota Biomodulators on Mucosal Immunity and Intestinal Inflammation. Cells 2020, 9, 1234. [Google Scholar] [CrossRef] [PubMed]
  104. Szandruk-Bender, M.; Wiatrak, B.; Szeląg, A. The Risk of Developing Alzheimer’s Disease and Parkinson’s Disease in Patients with Inflammatory Bowel Disease: A Meta-Analysis. J. Clin. Med. 2022, 11, 3704. [Google Scholar] [CrossRef]
  105. Sartor, R.B.; Wu, G.D. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology 2017, 152, 327. [Google Scholar] [CrossRef]
  106. Szandruk-Bender, M.; Rutkowska, M.; Merwid-Ląd, A.; Wiatrak, B.; Szeląg, A.; Dzimira, S.; Sobieszczańska, B.; Krzystek-Korpacka, M.; Kucharska, A.Z.; Matuszewska, A.; et al. Cornelian Cherry Iridoid-Polyphenolic Extract Improves Mucosal Epithelial Barrier Integrity in Rat Experimental Colitis and Exerts Antimicrobial and Antiadhesive Activities In Vitro. Oxid. Med. Cell Longev. 2020, 2020, 7697851. [Google Scholar] [CrossRef] [PubMed]
  107. Schirmer, M.; Garner, A.; Vlamakis, H.; Xavier, R.J. Microbial Genes and Pathways in Inflammatory Bowel Disease. Nat. Rev. Microbiol. 2019, 17, 497. [Google Scholar] [CrossRef] [PubMed]
  108. Dinan, T.G.; Cryan, J.F. Gut Instincts: Microbiota as a Key Regulator of Brain Development, Ageing and Neurodegeneration. J. Physiol. 2017, 595, 489. [Google Scholar] [CrossRef]
  109. Burokas, A.; Moloney, R.D.; Dinan, T.G.; Cryan, J.F. Microbiota Regulation of the Mammalian Gut-Brain Axis. Adv. Appl. Microbiol. 2015, 91, 1–62. [Google Scholar] [CrossRef]
  110. Wiatrak, B.; Balon, K.; Jawień, P.; Bednarz, D.; Jęśkowiak, I.; Szeląg, A. The Role of the Microbiota-Gut-Brain Axis in the Development of Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 4862. [Google Scholar] [CrossRef]
  111. Villain, N.; Dubois, B. Alzheimer’s Disease Including Focal Presentations. Semin. Neurol. 2019, 39, 213–226. [Google Scholar] [CrossRef]
  112. Doroszkiewicz, J.; Groblewska, M.; Mroczko, B. The Role of Gut Microbiota and Gut–Brain Interplay in Selected Diseases of the Central Nervous System. Int. J. Mol. Sci. 2021, 22, 10028. [Google Scholar] [CrossRef]
  113. Brandalise, F.; Roda, E.; Ratto, D.; Goppa, L.; Gargano, M.L.; Cirlincione, F.; Priori, E.C.; Venuti, M.T.; Pastorelli, E.; Savino, E.; et al. Hericium Erinaceus in Neurodegenerative Diseases: From Bench to Bedside and Beyond, How Far from the Shoreline? J. Fungi 2023, 9, 551. [Google Scholar] [CrossRef]
  114. Hong, S.M.; Yoon, D.H.; Lee, M.K.; Lee, J.K.; Kim, S.Y. A Mixture of Ginkgo Biloba L. Leaf and Hericium Erinaceus (Bull.) Pers. Fruit Extract Attenuates Scopolamine-Induced Memory Impairments in Mice. Oxid. Med. Cell Longev. 2022, 2022, 9973678. [Google Scholar] [CrossRef]
  115. Roda, E.; Priori, E.C.; Ratto, D.; De Luca, F.; Di Iorio, C.; Angelone, P.; Locatelli, C.A.; Desiderio, A.; Goppa, L.; Savino, E.; et al. Neuroprotective Metabolites of Hericium Erinaceus Promote Neuro-Healthy Aging. Int. J. Mol. Sci. 2021, 22, 6379. [Google Scholar] [CrossRef]
  116. Shirvani, M.; Nouri, F.; Sarihi, A.; Habibi, P.; Mohammadi, M. Neuroprotective Effects of Dehydroepiandrosterone and Hericium Erinaceus in Scopolamine-Induced Alzheimer’s Diseases-like Symptoms in Male Rats. Cell Biochem. Biophys. 2024, 1–12. [Google Scholar] [CrossRef] [PubMed]
  117. Sutthibutpong, T.; Posansee, K.; Liangruksa, M.; Termsaithong, T.; Piyayotai, S.; Phitsuwan, P.; Saparpakorn, P.; Hannongbua, S.; Laomettachit, T. Combining Deep Learning and Structural Modeling to Identify Potential Acetylcholinesterase Inhibitors from Hericium Erinaceus. ACS Omega 2024, 9, 16311–16321. [Google Scholar] [CrossRef]
  118. Diling, C.; Xin, Y.; Chaoqun, Z.; Jian, Y.; Xiaocui, T.; Jun, C.; Ou, S.; Yizhen, X. Extracts from Hericium Erinaceus Relieve Inflammatory Bowel Disease by Regulating Immunity and Gut Microbiota. Oncotarget 2017, 8, 85838. [Google Scholar] [CrossRef] [PubMed]
  119. Westfall, S.; Lomis, N.; Kahouli, I.; Dia, S.Y.; Singh, S.P.; Prakash, S. Microbiome, Probiotics and Neurodegenerative Diseases: Deciphering the Gut Brain Axis. Cell Mol. Life Sci. 2017, 74, 3769. [Google Scholar] [CrossRef]
  120. Pistollato, F.; Cano, S.S.; Elio, I.; Vergara, M.M.; Giampieri, F.; Battino, M. Role of Gut Microbiota and Nutrients in Amyloid Formation and Pathogenesis of Alzheimer Disease. Nutr. Rev. 2016, 74, 624–634. [Google Scholar] [CrossRef] [PubMed]
  121. Erny, D.; De Angelis, A.L.H.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host Microbiota Constantly Control Maturation and Function of Microglia in the CNS. Nat. Neurosci. 2015, 18, 965. [Google Scholar] [CrossRef]
  122. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Guan, N.L.; Kundu, P.; et al. The Gut Microbiota Influences Blood-Brain Barrier Permeability in Mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef]
  123. Park, S.; Kim, J.; Chun, J.; Han, K.; Soh, H.; Kang, E.A.; Lee, H.J.; Im, J.P.; Kim, J.S. Patients with Inflammatory Bowel Disease Are at an Increased Risk of Parkinson’s Disease: A South Korean Nationwide Population-Based Study. J. Clin. Med. 2019, 8, 1191. [Google Scholar] [CrossRef] [PubMed]
  124. Jiang, C.; Li, G.; Huang, P.; Liu, Z.; Zhao, B. The Gut Microbiota and Alzheimer’s Disease. J. Alzheimers Dis. 2017, 58, 1–15. [Google Scholar] [CrossRef]
  125. Zhou, Y.; Smith, D.; Leong, B.J.; Brännström, K.; Almqvist, F.; Chapman, M.R. Promiscuous Cross-Seeding between Bacterial Amyloids Promotes Interspecies Biofilms. J. Biol. Chem. 2012, 287, 35092. [Google Scholar] [CrossRef] [PubMed]
  126. Friedland, R.P. Mechanisms of Molecular Mimicry Involving the Microbiota in Neurodegeneration. J. Alzheimers Dis. 2015, 45, 349–362. [Google Scholar] [CrossRef]
  127. Neish, A.S. Microbes in Gastrointestinal Health and Disease. Gastroenterology 2009, 136, 65. [Google Scholar] [CrossRef]
  128. Leylabadlo, H.E.; Ghotaslou, R.; Feizabadi, M.M.; Farajnia, S.; Moaddab, S.Y.; Ganbarov, K.; Khodadadi, E.; Tanomand, A.; Sheykhsaran, E.; Yousefi, B.; et al. The Critical Role of Faecalibacterium Prausnitzii in Human Health: An Overview. Microb. Pathog. 2020, 149, 104344. [Google Scholar] [CrossRef]
  129. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of Brain Amyloidosis with Pro-Inflammatory Gut Bacterial Taxa and Peripheral Inflammation Markers in Cognitively Impaired Elderly. Neurobiol. Aging 2017, 49, 60–68. [Google Scholar] [CrossRef]
  130. Brierley, S.M.; Linden, D.R. Neuroplasticity and Dysfunction after Gastrointestinal Inflammation. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 611–627. [Google Scholar] [CrossRef] [PubMed]
  131. Prigent, A.; Chapelet, G.; De Guilhem de Lataillade, A.; Oullier, T.; Durieu, E.; Bourreille, A.; Duchalais, E.; Hardonnière, K.; Neunlist, M.; Noble, W.; et al. Tau Accumulates in Crohn’s Disease Gut. FASEB J. 2020, 34, 9285–9296. [Google Scholar] [CrossRef]
  132. Lionnet, A.; Wade, M.A.; Corbillé, A.G.; Prigent, A.; Paillusson, S.; Tasselli, M.; Gonzales, J.; Durieu, E.; Rolli-Derkinderen, M.; Coron, E.; et al. Characterisation of Tau in the Human and Rodent Enteric Nervous System under Physiological Conditions and in Tauopathy. Acta Neuropathol. Commun. 2018, 6, 65. [Google Scholar] [CrossRef]
  133. Dugger, B.N.; Hoffman, B.R.; Scroggins, A.; Serrano, G.E.; Adler, C.H.; Shill, H.A.; Belden, C.M.; Sabbagh, M.N.; Caviness, J.N.; Driver Dunckley, E.; et al. Tau Immunoreactivity in Peripheral Tissues of Human Aging and Select Tauopathies. Neurosci. Lett. 2019, 696, 132. [Google Scholar] [CrossRef]
  134. Prigent, A.; Lionnet, A.; Durieu, E.; Chapelet, G.; Bourreille, A.; Neunlist, M.; Rolli-Derkinderen, M.; Derkinderen, P. Enteric Alpha-Synuclein Expression Is Increased in Crohn’s Disease. Acta Neuropathol. 2019, 137, 359–361. [Google Scholar] [CrossRef] [PubMed]
  135. Fenyi, A.; Leclair-Visonneau, L.; Clairembault, T.; Coron, E.; Neunlist, M.; Melki, R.; Derkinderen, P.; Bousset, L. Detection of Alpha-Synuclein Aggregates in Gastrointestinal Biopsies by Protein Misfolding Cyclic Amplification. Neurobiol. Dis. 2019, 129, 38–43. [Google Scholar] [CrossRef] [PubMed]
  136. Dong, L.; Shen, Y.; Li, H.; Zhang, R.; Yu, S.; Wu, Q. Shared Genes of PPARG and NOS2 in Alzheimer’s Disease and Ulcerative Colitis Drive Macrophages and Microglia Polarization: Evidence from Bioinformatics Analysis and Following Validation. Int. J. Mol. Sci. 2023, 24, 5651. [Google Scholar] [CrossRef]
  137. Gravina, A.G.; Pellegrino, R.; Durante, T.; Palladino, G.; Imperio, G.; D’Amico, G.; Trotta, M.C.; Dallio, M.; Romeo, M.; D’Amico, M.; et al. The Melanocortin System in Inflammatory Bowel Diseases: Insights into Its Mechanisms and Therapeutic Potentials. Cells 2023, 12, 1889. [Google Scholar] [CrossRef]
  138. Catania, A. The Melanocortin System in Leukocyte Biology. J. Leukoc. Biol. 2007, 81, 383–392. [Google Scholar] [CrossRef] [PubMed]
  139. Catania, A. Neuroprotective Actions of Melanocortins: A Therapeutic Opportunity. Trends Neurosci. 2008, 31, 353–360. [Google Scholar] [CrossRef]
  140. Mountjoy, K.G.; Mortrud, M.T.; Low, M.J.; Simerly, R.B.; Cone, R.D. Localization of the Melanocortin-4 Receptor (MC4-R) in Neuroendocrine and Autonomic Control Circuits in the Brain. Mol. Endocrinol. 1994, 8, 1298–1308. [Google Scholar] [CrossRef]
  141. Tracey, K.J. Physiology and Immunology of the Cholinergic Antiinflammatory Pathway. J. Clin. Investig. 2007, 117, 289. [Google Scholar] [CrossRef]
  142. Giuliani, D.; Ottani, A.; Altavilla, D.; Bazzani, C.; Squadrito, F.; Guarini, S. Melanocortins and the Cholinergic Anti-Inflammatory Pathway. Adv. Exp. Med. Biol. 2010, 681, 71–87. [Google Scholar] [CrossRef]
  143. Gravina, A.G.; Pellegrino, R.; Durante, T.; Palladino, G.; Imperio, G.; D’Amico, G.; Trotta, M.C.; Dallio, M.; Romeo, M.; D’Amico, M.; et al. P072 Melanocortin MC3 and MC5 Receptors Expression Is Different According to Colonic Disease Activity in Crohn’s Disease and Ulcerative Colitis Biopsies: Results from an Immunohistochemical Observational Study. J. Crohns Colitis 2022, 16, i176–i177. [Google Scholar] [CrossRef]
  144. Panaro, B.L.; Tough, I.R.; Engelstoft, M.S.; Matthews, R.T.; Digby, G.J.; Møller, C.L.; Svendsen, B.; Gribble, F.; Reimann, F.; Holst, J.J.; et al. The Melanocortin-4 Receptor Is Expressed in Enteroendocrine L Cells and Regulates the Release of Peptide YY and Glucagon-Like Peptide 1 In Vivo. Cell Metab. 2014, 20, 1018. [Google Scholar] [CrossRef]
  145. Shen, Y.; Fu, W.Y.; Cheng, E.Y.L.; Fu, A.K.Y.; Ip, N.Y. Melanocortin-4 Receptor Regulates Hippocampal Synaptic Plasticity through a Protein Kinase A-Dependent Mechanism. J. Neurosci. 2013, 33, 464. [Google Scholar] [CrossRef]
  146. Shen, Y.; Tian, M.; Zheng, Y.; Gong, F.; Fu, A.K.Y.; Ip, N.Y. Stimulation of the Hippocampal POMC/MC4R Circuit Alleviates Synaptic Plasticity Impairment in an Alzheimer’s Disease Model. Cell Rep. 2016, 17, 1819–1831. [Google Scholar] [CrossRef]
  147. Giuliani, D.; Bitto, A.; Galantucci, M.; Zaffe, D.; Ottani, A.; Irrera, N.; Neri, L.; Cavallini, G.M.; Altavilla, D.; Botticelli, A.R.; et al. Melanocortins Protect against Progression of Alzheimer’s Disease in Triple-Transgenic Mice by Targeting Multiple Pathophysiological Pathways. Neurobiol. Aging 2014, 35, 537–547. [Google Scholar] [CrossRef] [PubMed]
  148. Giuliani, D.; Galantucci, M.; Neri, L.; Canalini, F.; Calevro, A.; Bitto, A.; Ottani, A.; Vandini, E.; Sena, P.; Sandrini, M.; et al. Melanocortins Protect against Brain Damage and Counteract Cognitive Decline in a Transgenic Mouse Model of Moderate Alzheimer’s Disease. Eur. J. Pharmacol. 2014, 740, 144–150. [Google Scholar] [CrossRef] [PubMed]
  149. Zhou, Y.; Chawla, M.K.; Rios-Monterrosa, J.L.; Wang, L.; Zempare, M.A.; Hruby, V.J.; Barnes, C.A.; Cai, M. Aged Brains Express Less Melanocortin Receptors, Which Correlates with Age-Related Decline of Cognitive Functions. Molecules 2021, 26, 6266. [Google Scholar] [CrossRef] [PubMed]
  150. Doan, T.; Massarotti, E. Rheumatoid Arthritis: An Overview of New and Emerging Therapies. J. Clin. Pharmacol. 2005, 45, 751–762. [Google Scholar] [CrossRef]
  151. Choy, E.H.S.; Panayi, G.S. Cytokine Pathways and Joint Inflammation in Rheumatoid Arthritis. N. Engl. J. Med. 2001, 344, 907–916. [Google Scholar] [CrossRef]
  152. 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]
  153. Feldmann, M.; Maini, R.N. The Role of Cytokines in the Pathogenesis of Rheumatoid Arthritis. Rheumatology (Oxford) 1999, 38 Suppl 2, 3–7. [Google Scholar] [CrossRef]
  154. Zheng, X.; Wu, S.L.; Hincapie, M.; Hancock, W.S. Study of the Human Plasma Proteome of Rheumatoid Arthritis. J. Chromatogr. A 2009, 1216, 3538–3545. [Google Scholar] [CrossRef]
  155. Bodi, K.; Prokaeva, T.; Spencer, B.; Eberhard, M.; Connors, L.H.; Seldin, D.C. AL-Base: A Visual Platform Analysis Tool for the Study of Amyloidogenic Immunoglobulin Light Chain Sequences. Amyloid 2009, 16, 1. [Google Scholar] [CrossRef] [PubMed]
  156. 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. Alzheimers Dis. 2019, 69, 59–70. [Google Scholar] [CrossRef] [PubMed]
  157. 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] [PubMed]
  158. 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] [PubMed]
  159. 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] [CrossRef]
  160. Walker, K.A.; Gottesman, R.F.; Wu, A.; Knopman, D.S.; Gross, A.L.; Mosley, T.H.; Selvin, E.; Windham, B.G. Systemic Inflammation during Midlife and Cognitive Change over 20 Years: The ARIC Study. Neurology 2019, 92, e1256. [Google Scholar] [CrossRef]
  161. 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, e0186475. [Google Scholar] [CrossRef]
  162. 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. Alzheimers Dis. 2012, 31, 669–676. [Google Scholar] [CrossRef]
  163. Sanchez-Santos, M.T.; Garriga, C.; Judge, A.; Batra, R.N.; Price, A.J.; Liddle, A.D.; Javaid, M.K.; Cooper, C.; Murray, D.W.; Arden, N.K. Development and Validation of a Clinical Prediction Model for Patient-Reported Pain and Function after Primary Total Knee Replacement Surgery. Sci. Rep. 2018, 8, 3381. [Google Scholar] [CrossRef] [PubMed]
  164. 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] [PubMed]
  165. 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]
  166. The Alzheimer’s Disease Anti-inflammatory Prevention Trial Research Group. Results of a Follow-up Study to the Randomized Alzheimer’s Disease Anti-Inflammatory Prevention Trial (ADAPT). Alzheimers Dement. 2013, 9, 714–723. [Google Scholar] [CrossRef]
  167. 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. [Google Scholar] [CrossRef]
  168. 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. [Google Scholar] [CrossRef]
  169. 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 Dementia Transl. Res. Clin. Interv. 2017, 3, 612. [Google Scholar] [CrossRef]
  170. Trzeciak, P.; Herbet, M.; Dudka, J. Common Factors of Alzheimer’s Disease and Rheumatoid Arthritis—Pathomechanism and Treatment. Molecules 2021, 26, 6038. [Google Scholar] [CrossRef]
Figure 1. Common aspects of the diseases with additional aspects specific to each disease.
Figure 1. Common aspects of the diseases with additional aspects specific to each disease.
Jcm 13 05057 g001
Table 1. Table presenting the comparison of characteristics of inflammatory bowel diseases and Alzheimer’s disease.
Table 1. Table presenting the comparison of characteristics of inflammatory bowel diseases and Alzheimer’s disease.
AspectUlcerative ColitisCrohn’s DiseaseConnection to ADRef.
Primary FeaturesInflammation limited to mucosal surface; continuous from rectum proximallyTransmural inflammation; skip lesions; can affect any part of GI tract from mouth to anusBoth UC and CD linked to increased risk of dementia; connection to neuroinflammation and amyloid deposition[95,96,99,100,101]
Clinical PresentationProctitis, left-sided colitis, pancolitis; limited to colonPersistent diarrhea, weight loss, abdominal pain; can involve entire GI tractHigher incidence of dementia in IBD patients; possible earlier onset and increased detection due to frequent medical visits[95,98,99,100,101]
Endoscopic and Histological FindingsContinuous mucosal inflammation, can include caecal patch in some casesTransmural inflammation with skip lesions; can involve any part of GI tractSimilar histological patterns in neurodegeneration and inflammation seen in both IBD and AD[95,96,103,104]
Gut MicrobiotaDysbiosis with decreased SCFA-producing bacteria, increased mucosa-associated bacteriaReduced diversity, decreased SCFA-producing bacteria, increased pro-inflammatory bacteriaAltered gut microbiota linked to AD through immune and inflammatory pathways[103,104,105,106]
Impact on Gut-Brain AxisIncreased gut permeability; potential impact on CNS due to dysbiosisGut–brain axis dysregulation; potential for microbial metabolites to affect CNSGut microbiota influence on neuroinflammation and amyloid deposition; increased gut permeability impacts CNS exposure[107,108,109,110]
Role of SCFA and Inflammatory CytokinesLower SCFA-producing bacteria; increased pro-inflammatory cytokinesAltered SCFA production and increased pro-inflammatory cytokinesSCFA and cytokines impact neuroinflammation and potentially AD progression[103,104,111,112]
Potential for Cross-Seeding of AmyloidsNot well-defined but potential for amyloids to affect neuroinflammationEvidence of microbial amyloids contributing to proteinopathy and inflammationGut-derived amyloids may impact AD pathology through cross-seeding and neuroinflammation[119,120,121]
Tau Protein ExpressionGenerally not elevated in UCHigher tau expression in enteric nervous system (ENS)Elevated tau in ENS linked to AD; potential shared mechanisms in tau pathology[123,124,125,126]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Doroszkiewicz, J.; Mroczko, J.; Winkel, I.; Mroczko, B. Metabolic and Immune System Dysregulation: Unraveling the Connections between Alzheimer’s Disease, Diabetes, Inflammatory Bowel Diseases, and Rheumatoid Arthritis. J. Clin. Med. 2024, 13, 5057. https://doi.org/10.3390/jcm13175057

AMA Style

Doroszkiewicz J, Mroczko J, Winkel I, Mroczko B. Metabolic and Immune System Dysregulation: Unraveling the Connections between Alzheimer’s Disease, Diabetes, Inflammatory Bowel Diseases, and Rheumatoid Arthritis. Journal of Clinical Medicine. 2024; 13(17):5057. https://doi.org/10.3390/jcm13175057

Chicago/Turabian Style

Doroszkiewicz, Julia, Jan Mroczko, Izabela Winkel, and Barbara Mroczko. 2024. "Metabolic and Immune System Dysregulation: Unraveling the Connections between Alzheimer’s Disease, Diabetes, Inflammatory Bowel Diseases, and Rheumatoid Arthritis" Journal of Clinical Medicine 13, no. 17: 5057. https://doi.org/10.3390/jcm13175057

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

Article Metrics

Back to TopTop