Skip to main content
Brain Communications logoLink to Brain Communications
. 2025 Feb 25;7(2):fcaf092. doi: 10.1093/braincomms/fcaf092

Could immunotherapy and regulatory T cells be used therapeutically to slow the progression of Alzheimer’s disease?

Victoria Abbott 1, Benjamin E Housden 2,3, Annwyne Houldsworth 4,5,
PMCID: PMC11896979  PMID: 40078868

Abstract

Alzheimer’s disease and other cognitive impairments are a growing problem in the healthcare world with the ageing population. There are currently no effective treatments available; however, it has been suggested that targeting neuroinflammation may be a successful approach in slowing the progression of neurodegeneration. Reducing the destructive hyperinflammatory pathology to maintain homeostasis in neural tissue is a promising option to consider. This review explores the mechanisms behind neuroinflammation and the effectiveness of immunotherapy in slowing the progression of cognitive decline in patients with Alzheimer’s disease. The key components of neuroinflammation in Alzheimer’s disease researched are microglia, astrocytes, cytokines and CD8+ effector T cells. The role of oxidative stress on modulating regulatory T cells and some of the limitations of regulatory T cell–based therapies are also explored. Increasing regulatory T cells can decrease activation of microglia, proinflammatory cytokines and astrocytes; however, it can also increase levels of inflammatory cytokines. There is a complex network of regulatory T cell interactions that reduce Alzheimer’s disease pathology, which is not fully understood. Exploring the current literature, further research into the use of immunotherapy in Alzheimer’s disease is vital to determine the potential of these techniques; however, there is sufficient evidence to suggest that increasing regulatory T cells count does prevent Alzheimer’s disease symptoms and pathology in patients with Alzheimer’s disease. Some exciting innovative therapies are muted to explore in the future. The function of regulatory T cells in the presence of reactive oxygen species and oxidative stress should be investigated further in patients with neurogenerative disorders to ascertain if combination therapies could reduce oxidative stress while also enhancing regulatory T cells function. Could methods of immunotherapy infuse exogenous functional Tregs or enhance the immune environment in favour of endogenous regulatory T cells differentiation, thus reducing neuroinflammation in neurodegenerative pathology, inhibiting the progression of Alzheimer’s disease?

Keywords: regulatory T cells, Alzheimer’s disease, oxidative stress, neuroinflammation, immunotherapy


Abbott et al. describes Alzheimer’s disease pathology associated with neuroinflammation. Tregs are neuroprotective CD4+ T-cells that maintain central nervous system homeostasis by reducing inflammation and inhibiting T cell proliferation and cytokine production. They also reduce microglia-mediated inflammation, inhibit neurotoxicity, neuroinflammation and the progression of neurofibrillary tangles and Aβ production.

Graphical Abstract

Graphical Abstract.

Graphical Abstract

Alzheimer’s disease symptoms and prevalence

Alzheimer's disease is a common neurodegenerative disease characterized by the progressive loss of memory, cognitive function and independence in daily tasks, where 1 in 14 of 65 years old and above are affected as late-onset Alzheimer's disease.1‐5 Patients presenting at a younger age, <65 years are referred to as early-onset Alzheimer's disease.6 Treatment for Alzheimer's disease is extremely costly worldwide estimated at £26.3 billion in the UK.4 Strategies to treat both cognitive decline (CD), including Alzheimer's disease and mild cognitive impairment (MCI) are important areas to address.3,7‐10 Criteria for pathological diagnosis of Alzheimer's disease include the presence of extracellular amyloid-beta (Aβ) plaques and intraneuronal neurofibrillary tangles (NFT) made of phosphorylated tau protein. Both abnormal accumulations of protein lead to a disruption in cell function, neuroinflammation and oxidative stress, all which have been linked to neuronal cell death.11,12

Current treatments, prevention and risk factors

Research has identified multiple key risk factors for developing Alzheimer's disease. These include age, genetics and family history. According to most of the literature produced on the subject, age is the most significant risk factor for CD and Alzheimer's disease. Reports suggest that the estimated 19% prevalence of Alzheimer's disease in people ages 74–85 increases to between 30 and 50% in populations over the age of 85.13‐16 Therefore, preventions that aim to increase the age at onset (AAO) have a clear benefit as, especially with the ageing population, the burden of care on families and the NHS will become unsustainable.

In order to understand the pathogenesis of Alzheimer's disease, neuroinflammatory immunology and oxidative stress, the use of genome-wide association studies (GWAS) has allowed researchers to identify genetics as risk factors for Alzheimer's disease may give further clues as to the pathogenesis of the disease, especially where some polymorphisms cause reduced or dysfunctional function. There is sufficient evidence to show that multiple genetic variants, including apolipoprotein E-ε4 (APOE), amyloid precursor protein (APP), presenilin 1 (PSEN1) and presenilin 2 (PSEN2), increase an individual’s risk for AD.17,18 However, given that neither genetics or age are modifiable factors, research must focus on understanding and targeting modifiable factors to be effective in the search for effective preventions and therapeutics. APOE is overexpressed following oxidative stress leading to a greater degree of oxidative insults in the brain.19

Currently, there is no cure for CD or Alzheimer's disease, although there are multiple treatments offered by the NHS aimed to reduce symptoms but not reverse or stop them.20,21 These treatments include acetylcholinesterase (AChE) inhibitors (which increase acetylcholine levels in the brain and therefore increase nerve transmission22) and memantine (NMDA receptor antagonist23), as well as cognitive therapies.20 Several studies have shown the limited efficacy of these current treatments, although all three techniques are considered to delay the progression of CD and AD.22‐26 Current research focuses on the treatment of Alzheimer's disease through immunotherapy with multiple drugs showing promising results in delaying on the onset of CD and progression of Alzheimer's disease. In 2023, the use of monoclonal antibodies that target Aβ, Lecanemab and aducanumab (Aduhelm) were approved by the by the Food and Drug dministration (FDA) to treat AD.27 The drug is an intravenous infusion therapy using monoclonal antibodies to target Aβ plagues,27,28 similarly to a drug, Aducanumab, which was approved in 2021. Few studies have evaluated the effectiveness of Lecanemab on later stage Alzheimer's disease, thus further studies are required to determine the impact of the drug on these patients.29 Aducanumab was discontinued in 2024, reportedly due to the manufacturing costs,30,31 highlighting the major issue with monoclonal antibody therapies not being assessable. Many studies also debated the drug’s clinical efficacy and safety,32‐34 which lead to the drug not being approved for use in Europe and the UK.30 Nevertheless, Aducanumab and Lecanemab opened the door into this new era of Alzheimer's disease drug research focusing on immunotherapy to combat Alzheimer's disease symptoms.29 Disease-modifying drugs, such as these, have the potential to decrease the number of MCI cases developing into dementia and increase the life expectancy and quality of life for patients with Alzheimer’s disease.10 Overall, there is a growing need for effective Alzheimer’s disease treatments without side effects, such as serious symptoms experienced in 1–2% of patients treated with these drugs who report headache, seizures, delirium, impaired speech, problems with vision and microbleeds in the brain. Recently, Donanemab was approved by the Food and Drug Administration (FDA) to treat AD.35 Although Donanemab has been approved by the FDA in the United States and is available for patients, it is not able to be prescribed in the UK on the NHS, while being available to private health patients. Donanemab was trialled in patients who were experiencing early symptoms of Alzheimer’s disease with tau and amyloid pathology and was shown to slow down clinical progression at 76 weeks.35

Search strategy

Regulatory T cells and the immune role in Alzheimer’s disease

In recent years, there has been a significant shift of focus in the literature onto the importance of the immune system and its role in Alzheimer’s disease (Fig. 1).21,36,37 A key example being the use of monoclonal antibodies to treat Aβ plagues.27‐30,32‐34 Accumulating research suggests that there is potential for therapeutics for Alzheimer’s disease using immunotherapy to target neuroinflammation, a key aspect of Alzheimer’s disease.21,36‐38 One of these methods of modifying the immune response is the use of ex vivo expanded regulatory T cell (Treg) infusions.39 Tregs are a subset of neuroprotective CD4+ T cells, which are identified by the expression of forkhead box protein P3 (Foxp3).40 The main role of Tregs is to maintain homeostasis within the central nervous system (CNS), by inhibiting T cell proliferation and cytokine production.41‐43 Tregs also have been shown to modulate adaptive immune reactions and reduce microglia and macrophage-mediated inflammation.41 Microglia have been suggested to accelerate neurotoxicity, neuroinflammation and the progression of NFT and Aβ pathogenesis in Alzheimer’s disease.37,39,43 Furthermore, studies have found that this cell population is dysfunctional in multiple neurodegenerative diseases.39,44 Thus, increasing Treg count may result in slower pathogenesis,39,41 demonstrating that Tregs have clear potential as a therapeutic target in Alzheimer’s disease. Also, Tregs play a crucial role in autoimmunity, but there is limited research on the effects of disrupting this balance and the risk of neuronal damage.42,45 Recently, research has focused on the different cell types affected by Tregs,39,41‐43,46 the effect of oxidative stress47 and the debated therapeutic potential of the use of Treg infusions.41,46,48,49 Whilst theoretically promising, the complex web of mechanisms involving Tregs in Alzheimer’s disease is not entirely understood.

Figure 1.

Figure 1

Search strategy for discussion. The databases listed above were searched using the terms described above. Each sections search results were then defined further, ending with a final screening for relevance.

Healthy brains of mice and humans have populations of brain resident CD69 Tregs, and these have been shown to expand rapidly in several neuroinflammatory conditions. Brain-resident Tregs are proposed to control astrogliosis and restrain inflammatory responses and polarize microglia into neuroprotective states.50

In this review, the term ‘natural Tregs’ refers to the Tregs in the body, which have not been manipulated, expanded artificially or transplanted by researchers. These cells are the Tregs present in the body that naturally occur, with varying levels based on a multitude of factors. The term ‘Treg infusions’ will be used in this review to refer to techniques that increase the frequency of Tregs within the body, including adoptive cell transfer, which uses Tregs from a donor,51 as well as the use of ex vivo expanded Tregs, which involves taking the cells out of the body and proliferating them, before returning them back to the body.39,41

Research questions and aims

Several research questions are posed in this review, including, what are the mechanisms of action of Tregs on cell types in the brain; does an increase in the Treg count produce neuroprotection against Alzheimer’s disease; what side effects are there in Treg infusion and how can these findings be translated into effective therapies?

The review aims to assess the therapeutic potential of ex vivo expanded Treg infusions by exploring the key cellular mechanisms underlying Tregs. This research will help develop understanding of the role of Tregs in Alzheimer’s disease, investigate any underlying limitations to the proposed therapy and suggest future research to aid in the development of novel treatments for Alzheimer’s disease.

Evidence for increasing Treg levels in Alzheimer’s disease

Treg levels are reduced in Alzheimer’s disease

A variety of studies have established that Tregs play a critical role in homeostasis of the immune system.42,48,52 Given that the immune role in Alzheimer’s disease studies is an increasingly significant area of interest,21,36,38 Alzheimer’s disease Treg studies have become a major focus in the field.43,46,47,53 There is some inconsistency within the literature surrounding the levels and functionality of Tregs in Alzheimer’s disease, but most studies report a depletion of Treg count and function.54 For example, a 2010 study found that percentages of Treg in blood are decreased in patients with Alzheimer’s disease compared with healthy controls, with the lowest percentages of all subtypes of Tregs in patients with more severe AD.55 Interestingly, a more recent study found that there was no significant difference between Treg count in patients with Alzheimer’s diseasecompared to MCI controls53; however, the lack of healthy controls in this study may account for the inconclusive data. The reported reduction in Tregs in patients with Alzheimer’s disease implies that there may be a benefit in increasing levels to match healthy control pairs.

Treg function is reduced in Alzheimer’s disease

In addition to the reduced frequencies of Tregs reported in patients with Alzheimer’s disease, there is evidence to suggest that the Tregs present in Alzheimer’s disease have reduced suppressive function and different immunophenotypes to healthy patient Tregs,39 contrasting with healthy brain Tregs, as described earlier.50 This study also found that there are significant differences in immunophenotypes present in patients with Alzheimer’s disease, with reduced CD25 surface expression on Alzheimer’s disease Tregs.39 The CD25 marker is reportedly crucial for the survival and expansion of Tregs,54,56 supporting the theory that reduced frequencies of Tregs are present in Alzheimer’s disease. Conversely, to the previous study, Rosenkranz et al.57 found an increase in suppressive activity in patients with Alzheimer’s disease in comparison to young healthy controls, but no difference when compared to age-matched controls. In addition, another study found no significant difference in the percentages of CD25− Tregs between healthy controls and patients with Alzheimer’s disease, but they did find lower percentages of this subtype of Treg in individuals with MCI. This study was unable to explore the functions of Tregs due to insufficient numbers of cells from donors, which may have affected other data in their investigation.58 Despite the inconsistencies in findings in these studies, most researchers appear to have accepted the theory that Tregs in patients with Alzheimer’s disease have reduced function39,41,59 due to numbers, although the mechanism behind this and whether there are differences in immunophenotypes remains unclear.

Increasing Tregs reduces the symptoms of Alzheimer’s disease

It is important to discuss the effect of Tregs on the symptoms of Alzheimer’s disease. Whilst there is evidence that Treg infusions reduce and prevent Alzheimer’s disease pathology,39,46 this reduction needs to translate into the symptoms of Alzheimer’s disease to be an effective treatment. The literature suggests that Tregs play a vital role in modulating disease progression in Alzheimer’s disease mice models. Baek et al.59 showed that lower percentages of Tregs increase the rate of CD and increase Aβ plagues. Thus, increasing Treg levels in blood could attenuate the progression of Aβ plagues. Supporting this, other studies demonstrated that amplification of Tregs restored cognitive function and delayed the onset of CD in mice models expressing chimeric mouse/human amyloid precursor and a mutant human presenilin 1 protein (APP/PS1).60‐63 Together, these studies suggest that Tregs are essential in learning and memory, which are key aspects of Alzheimer’s disease. The data also suggest that the immunosuppressive role of Tregs may be the key in the prevention of Alzheimer’s disease pathology. Although this is a new field of research in Alzheimer’s disease and currently, there is sparse data about how these findings translate into human patients, some recent findings demonstrate a clear potential in further research to determine the extent to which Treg therapies can reduce/prevent symptoms of Alzheimer’s disease.

The main focus of this review has explored ex vivo Treg expansion, it is also important to consider in vivo Treg expansion as a promising area of research and therapeutic approach, involving the adoptive transfer of these immune cells. Tregs express an abundance of interleukin-2 receptors and congenital deficiency or dysfunction of this receptor reduces the differentiation of Treg cell lineage, thus has been shown to be essential for the development of Tregs.64 In a recent phase 1 trial of amyotrophic lateral sclerosis (ALS), patients receiving interleukin-2 and CTLA-4Ig, biomarkers of OS were observed as well as indicators of neurodegeneration and neuroinflammation.65,66 Recombinant human IL-2 (Aldesleukin) has been administered in a phase 2a trial for patients with ALS, in a randomized, double-blind, placebo-controlled trial and found to expand the Treg population, reducing plasma myeloid activating cytokines, while suppressing peripheral proinflammatory monocytes.66,67 This trial has served as a proof-of-concept trial as to the impact of in vivo Treg expansion on patients with Alzheimer’s disease. It has yet to be determined if this type of therapy for in vivo Treg expansion will modify neuroinflammation in patients with AD.67

Tregs can be modulated by various factors

Oxidative stress promotes instability of Tregs

Oxidative stress describes an imbalance between free radicals and antioxidants. Free radicals are highly reactive molecules, due to an uneven number of electrons, which can bind to other molecules to result in damage to DNA, proteins and tissue when in excess.68‐71 Reactive oxygen species (ROS) are a subset of free radicals containing oxygen that can induce inflammation,72 which is shown to be a key player in Alzheimer’s disease pathology.3,37,73 Oxidative stress has often been associated with CD and AD,74,75 and there is evidence to suggest that Tregs may play a role in this mechanism. As previously mentioned, data suggest that patients with Alzheimer’s disease may have an increased percentage of CD25− Tregs,39 and CD25 knockout Tregs have been shown to increase oxidative stress and apoptosis.56 Furthermore, there is evidence that oxidative stress promotes instability of Tregs.47 Together with the previous study, this demonstrates there is a clear link between ROS and Tregs in the progression of Alzheimer’s disease. Understanding this link may be useful to determine the therapeutic potential of Tregs. It is possible that the higher levels of oxidative stress found in patients with Alzheimer’s disease may decrease the efficacy of Treg infusions in comparison to the use of similar methods to treat other diseases, but there is yet to be any research investigating this. There is also evidence that microglia may be activated by excessive ROS, leading to further neuroinflammation.68 The studies imply that there is a benefit to increasing Treg levels in Alzheimer’s disease, as the association between oxidative stress and Alzheimer’s disease may be partially due to the instability of Tregs caused by ROS.

Furthermore, oxidative damage leads to an increase in IL-6 production, which has been shown to inhibit Foxp3 expression in Treg differentiation,76 suggesting that oxidative stress is the cause of a decrease of Treg levels in patients with Alzheimer’s disease. Since this inhibition of expression takes place in the differentiation of the cells, oxidative stress may not affect infused Tregs; however, more research is needed to investigate this.

Superoxide dismutases can modulate oxidative stress

Superoxide dismutase (SOD) enzymes catalyze the dismutation of superoxide anions (a type of ROS) and play a central role in defence against oxidative stress. They are typically associated with metals, such as copper, zinc, iron or manganese.68,77 In Alzheimer’s disease, mutations in the gene encoding for SOD2 have been identified, resulting in a loss of antioxidant activity.68 Interestingly, SOD2 is dependent on manganese, which has also been found to be significantly reduced in AD.78 There is evidence to suggest that SOD2 is upregulated in patients with Alzheimer’s disease, although this may be a compensatory mechanism due to the increased oxidative stress.78 Studies have shown that reducing SOD2 activity in Alzheimer’s disease models increases disease-like pathology,70 demonstrating clearly that oxidative stress and SOD enzymes play an important role in Alzheimer’s disease pathology. Using this, Treg levels could potentially be increased naturally by increasing the activity of SOD. Given the dysfunctional role of SOD enzymes in Alzheimer’s disease, it is possible that an infusion of Tregs would have a limited effect as they become unstable from ROS, and it may be necessary to measure the antioxidant status of patients with Alzheimer’s disease before administering these novel treatments. Further research is necessary to better understand the mechanisms of oxidative stress and Tregs.

Antioxidants can modulate oxidative stress

Antioxidant-rich food is well known to have many health benefits. Antioxidants are natural polyhydroxylated phenolic compounds with low molecular weights and can eliminate ROS. Antioxidants reduce oxidative stress and therefore can be used to prevent AD.71 Many of these foods have been demonstrated to alter Aβ aggregation, improve neuronal physiology and enhance cognitive abilities.79‐81 This demonstrates again that ROS plays an important role in Alzheimer’s disease pathology. Whilst there are issues with antioxidants crossing the blood–brain barrier,81 advancements in therapies for Alzheimer’s disease could produce a drug that uses antioxidants alongside Treg-based therapies to target both aspects of the disease. Potentially, creating a drug with a lower dosage of Tregs, requiring fewer intravenous infusions and reduced side effects, may provide a more assessable treatment. Without elucidating the mechanisms of oxidative stress in Alzheimer’s disease, it is possible that new Tregs would not differentiate, and Treg infusions will not have long-term effects.

Tregs mechanism of action in Alzheimer’s disease

Tregs modulate neuroinflammation

Extensive research has shown that Tregs play an important role in modulating neuroinflammation in Alzheimer’s disease.21,37,38,41 Neuroinflammation has been suggested as an essential therapeutic target due to the role of neuroinflammation in aggravating Aβ pathology and Alzheimer’s disease development.82,83 There is also evidence to suggest that ongoing inflammation aids the phosphorylation of tau.73,84 There are various known mechanisms underpinning this neuroinflammation that will be discussed in the next section, with focus on the interactions Tregs have with different cell types (Fig. 2) and how these affected cells interact with Aβ and tau pathologies.

Figure 2.

Figure 2

The mechanisms underpinning the effect of increased regulatory T cells on neuroinflammation. Regulatory T cells (Tregs) modulate many pathways in Alzheimer's disease to reduce inflammation. Tregs modulate astrogliosis to prevent reactive astrocytes.85 They also inhibit microgliosis.58 Both functions lead to reduced amyloid-beta plagues and neurofibrillary tangles of tau protein.61,62 Furthermore, Tregs produce many anti-inflammatory cytokines such as interleukin-10 (IL-10) and transforming growth factor-beta TGF-β.86 Image adapted from Jaber87 and information from Le Page et al.,58 Chay,et al.,85 Park et al.86 and Chakrabarty et al.88 Created in BioRender. Houldsworth, A. (2025) https://BioRender.com/i62v491.89

Tregs modulate microglial activation

Microglia act as the first defence in the brain by phagocytosing dead cells and cell debris and in Alzheimer’s disease, microglia respond to Aβ plague. Microglia also phagocytose misfolded protein, such as both soluble and fibrillary forms of Aβ.90,91 These cells can also trigger both pro-inflammatory and anti-inflammatory cytokines into action. Whilst the preliminary activation of microglia is vital for containing and preventing Aβ plaque, sustained activation leads microglia to skew towards defective subtype, which can cause long-term damage to the CNS.91‐93 These microglia increase Aβ plagues and tau proliferation, which then activate more microglia, creating a self-proliferating pathology.94,95 The suppressive function of Tregs can regulate the inflammatory microglial phenotype.51 This has been demonstrated using ex vivo expansion and adoptive transfer studies, showing the increased numbers and function of Tregs can reduce the levels of reactive microglia, as well as altering the inflammatory phenotype.39,51,59

Further suggesting the significance of microglia in Alzheimer’s disease pathology, GWAS have identified the APOE E4 gene variant (which is found in a gene that are expressed by microglia) to be associated with AD.91,96 A 2022 study also found that the function of microglial Aβ clearance is compromised in Alzheimer’s disease, although they found this can be improved by activation of PIEZO1 channels.97 The complex mechanisms in which microglial function is reduced in Alzheimer’s disease is not yet fully understood, further studies exploring this may provide insight into finding new therapeutic targets for understanding and treating Alzheimer’s disease.

Although studies show Treg-induced reduction in microglia to have a positive effect in reducing and preventing Aβ plague,39,41,51,59 there remains a question about the discriminatory abilities and localization of Tregs.98 The balance of microglia is important to maintain normal functioning as they play a crucial role in modulating synaptic plasticity through the expression and release of neurotrophic factors, such as brain-derived neurotrophic factor (BDNF). BDNF is released via the microglial phosphatidylinositol 3-kinase/BDNF signalling pathway and is essential for neuronal growth, learning and memory.99‐102 It has been shown that decreased levels of BDNF are associated with Alzheimer’s disease, suggesting that BDNF may be useful in the prevention of AD.99 Whilst this review is limited to the role of BDNF as a microglial-derived neurotrophic factor as an example, it is important to recognize that there are various other microglial-derived neurotrophic factors that are affected by microglial count and may play a role in Alzheimer’s disease. Therefore, the control of Tregs function, migration and survival is essential in reducing microglial-meditated Alzheimer’s disease pathology and further research into controlling and understanding the balance of Tregs and microglia would prove beneficial in the steps towards a safe and effective Treg drug for Alzheimer’s disease. Studies trialling ex vivo expanded Tregs on animal models fail to mention the importance of exploring this balance to develop an effective drug. In the future, it would be interesting to see studies focused on the potential negative impact of excessive Tregs on microglia in Alzheimer’s disease brains to better understand the consequences of over-dosing this new drug type.

Some abnormal signalling has been reported across a network of microglial genes in their expression of mRNA transcript alterations as a shared genetic risk in neurodegenerative disorders. These aberrant gene expressions affect physical interactions between CCRX4 (CD152), a receptor on Tregs, and microglial receptors. This changed signalling may contribute to neurodegeneration.103 However, ex vivo expanded adoptive transfer Tregs modulate the activation and polarization of microglia in TMT-induced neurodegeneration animal models, and this may be a mechanism that could shift the activation of microglia from inflammatory clustered forms to quiescent conformations, thus reducing the formation of dystrophic neurites.86

Tregs activate cytokines

Another way that Tregs can act to modulate neuroinflammation is by producing immunosuppressive cytokines, such as interleukin 10 (IL-10), interleukin 35 (IL-35) and transforming growth factor beta.104 Cytokines play an important role in regulating immune responses and therefore can modulate neuroinflammation in Alzheimer’s disease. For example, IL-10 suppresses inflammation by inhibiting the secretion of IL-1β, IL-6 and tumour necrosis factor alpha by microglia.105 A study by Le Page et al.58 found IL-10 levels to be higher in both Alzheimer’s disease and MCI patients in comparison to healthy controls. However, the effects of IL-10 on Alzheimer’s disease pathology are still debated, with some studies showing excessive IL-10 in Alzheimer’s disease models to worsen CD and Aβ plague,105 and others showing increased levels of IL-10 to improve spatial learning in APP/PS1 mice.106 Despite this, transforming growth factor beta has been shown to promote microglial and macrophage-mediated Aβ clearance107 demonstrating that cytokines play a significant role in Alzheimer’s disease. Further investigation of cytokine interactions in Alzheimer’s disease is recommended to better understand the significance of these within the disease.

There is sufficient evidence to suggest that interleukin-2 (IL-2) improves amyloid pathology and memory in Alzheimer’s disease mice.108 Interestingly, IL-2 also boosts the suppressive function of Tregs not only suggesting that Tregs also may a role in improving Alzheimer’s disease hallmarks but also highlighting IL-2 as a potential target for Treg enhancement and Alzheimer’s disease treatment.108,109 IL-2 is necessary for Foxp3 expression and signalling to maintain survival of Tregs,110,111 thus vital for development and stability. This is demonstrated further by studies using IL-2 treatments to amplify Tregs, which have been shown in restore cognitive functions in APP/PS1 mice.60 However, there is a low presence of IL-2 in Alzheimer’s disease brains and Tregs are poor at producing IL-20.112,113 Taken together, this suggests that for Treg infusions to have long-term effects in Alzheimer’s disease, levels of IL-2 may also need to be increased within the CNS.

Both pro-inflammatory and anti-inflammatory cytokines are elevated in the plasma of patients with Alzheimer’s disease, reflecting the changes in the immune system.58,114 It is unclear whether this is a consequence or causing factor of Alzheimer’s disease, highlighting the need for further research into the dynamics of the disease. Whilst the mechanisms and impact of some of the Treg-induced cytokines on Alzheimer’s disease may be partially unclear, the fact that Tregs have been shown to produce immunosuppressive cytokines remains and is therefore relevant to study further when exploring the therapeutic potential of Tregs.

Tregs modulate astrocytes

Astrocytes make up a significant part of the CNS; however, for decades, they were believed to play just a supporting role in the brain and were largely overlooked.115 Astrocytes have many structural, metabolic and neuroprotective functions, including the modulation of synapses.116,117 As part of the immune response, astrocytes undergo reactive astrogliosis, a process which includes increased gene expression of structural proteins and proliferation. In Alzheimer’s disease, reactive astrocytes build up around Aβ plagues, have chronic activation and dysfunctional signalling.118,119 Studies have shown that excessive Aβ upregulates astrogliosis,85 thus reactive astrocytes play a key role in neuroinflammation in Alzheimer’s disease. Similarly to microglia, Tregs can modulate astrocytes to prevent neuroinflammation.120 Studies have also found that a depletion of Tregs increases the activation of astrocytes,121 supporting the benefit of increasing Tregs as a method to prevent neuroinflammation in Alzheimer’s disease.

There is evidence that astrocytes can protect the CNS from oxidative injury. However, other reports suggest astrocytes may be a source of ROS and in turn, excessive ROS may lead to may reactive astrocytes,69,122 suggesting they play a dual role. Astrocytes can also phagocytose Aβ and secrete Aβ-degrading proteases, such as APOE.123 Whilst astrocytes play an important role in maintaining a healthy CNS, the balance of reactive astrocytes is critical in preventing neuroinflammation. Exploring the mechanisms that Tregs modulate astrocytes is critical for safe application of Treg-based therapies to prevent over-modulation of vital astrocytes.

Tregs modulate CD8+ T cells

Another main function of Tregs is modulating CD8+ effector T cells. These cells play a critical role by producing antiviral cytokines and removing virus-infected cells.124 CD8+ T cells can also produce inflammatory cytokines such as TNF, which then activate microglia and other immune cells.125 Recent studies have found an increased number of CD8+ T cells present in patients with Alzheimer’s disease, which result in an increase in neuroinflammation.126,127 Although, other studies found no significant difference in CD8+ T cell frequencies compared with healthy controls.59 These inconsistencies may exist due to differences in the stage of disease studied. Despite this, it has been shown that Tregs can modulate CD8+ T cell expansion and differentiation128,129 to prevent further neuroinflammation. Although, the mechanisms behind this are not well understood. One study showed that IL-2 produced by CD8+ T cells expand Tregs, which then limit CD8+ cell responses.129 Thus, increasing Tregs can modulate CD8+ T cells, and in turn, can modulate neuroinflammation through this pathway. Exploring CD8+ T cells further could reveal a deeper insight into Alzheimer’s disease pathology, as their mechanisms in Alzheimer’s disease remain largely unknown.

Terminally differentiated effector T cells (TEMRA) are associated with CD and Alzheimer’s disease, having a more cytotoxic phenotype, also connected with neurodegeneration, neuroinflammation and plasma markers of AD.130 More specifically, CD8+ TEMRA cells were shown to be associated with neural injury and Alzheimer’s disease, whereas CD4+ TEMRA have a role in the interplay of inflammatory mechanisms and CD8+ activation.130

Tregs reduce amyloid burden

There is a great focus within the research placed on Aβ plagues in Alzheimer’s disease as they are established as one of the most significant hallmarks of Alzheimer’s disease. Investigating ways of reducing Aβ plagues is a continuing approach in Alzheimer’s disease drug development, and many studies have looked at how Tregs effect Aβ plagues. Some studies have shown that a depletion of Tregs results in increased Aβ plagues.59,131 Supporting this, other studies have demonstrated that increased levels of Tregs lead to increased Aβ plague clearance.39,61‐63,132‐163 Contrasting this, Dansokho et al.60 found that there was no significant difference in Aβ burden in Treg-depleted APP/PS1 and non-depleted mice, although it was found that decreases in plaque-associated microglial activity and it was concluded that Tregs have a neuroprotective effect on Alzheimer’s disease models. The reasons behind the inconsistent finds are unclear, although it may be due to the modulation of Tregs at different stages of disease. Dansokho et al. altered Treg levels when mice were 5–6 weeks of age, compared to Treg modulation in 4- to 5-month-old mice in a conflicting study by Baruch et al.133 Despite findings by Dansokho et al., most researchers have come to the consensus that increasing levels of Tregs does reduce or prevent the development of Aβ plagues. Taken together, the findings still indicate that increasing levels of Tregs in Alzheimer’s disease is beneficial in reducing/preventing symptoms and therefore must be explored further to potentially develop a new drug type of Alzheimer’s disease.

Tregs reduce phosphorylated tau

The second key hallmark in Alzheimer’s disease pathology is the NFT of phosphorylated tau protein. Whilst the impact increasing Tregs have on tau pathology is less prominent in the literature compared to the effect on Aβ plagues, it is necessary to discuss tau due to its prominence in the disease pathology. Data from several studies suggest that ongoing inflammation from microglia in Alzheimer’s disease facilitates tau phosphorylation,73,134 therefore implying that increasing Treg levels should prevent/reduce phosphorylated tau NFT, because Tregs reduce microglia and inflammation. A study by Yang et al.62 demonstrated mitigation of tau deposits by ex vivo-expanded Tregs, suggesting that Tregs can play a role in reducing tau Alzheimer’s disease pathologies. Another study found that Alzheimer’s disease patients’ levels of natural Tregs are correlated with the levels of NFT and Tau.53 Levels of tau have also been shown to predict the rate of CD.135,136 As increasing Tregs can restore cognitive function,59,60 this together implies that increasing Tregs could also reduce levels of tau. Adding to this, it has been shown that Tregs suppress microglia, which have been shown to accelerate development of NFT.63 It would be interesting to see more studies focusing on the impact of increased Tregs on tau pathogenesis in Alzheimer’s disease as there is limited research available on this concept, although it is clear from current research that there are various mechanisms triggered by an increase in Tregs that lead to reduced tau pathogenesis.

Chimeric antigen receptor (CAR) Tregs immunotherapy for Alzheimer’s disease

There have been some significant advances in cancer therapy using CAR T cell therapy and this concept, using engineering CAR-Treg cells for treating patients with Alzheimer’s disease as an immunotherapy, despite there being numerous challenges to this approach.137,138 Designing an antigen target that is appropriate for the required immune outcome being one of the issues to address. Another being maintaining Treg phenotype and stability of function over time. Pathogenic conversion was observed for Tregs to TH17 cells in autoimmune arthritis.139

Limitations of Treg infusions

Tregs act as an immunosuppressant

Although Tregs have an important immunoregulatory role, some immunosuppressant role of Tregs in Alzheimer’s disease is now recognized within the literature42,54,133; however, there is a relatively small portion of the literature that is concerned with the control of the immunosuppressive function of Treg infusions. To date, several studies have explored the relationship between immunosuppressants and cancer, as well as the importance of immune system equilibrium.45,140,141 The body needs an effective immune response to fight infections and keep cancer at bay142; however, if this immune response is overactive, then immune cells can cause tissue damage. This is known as autoimmune disease.140,141,143 Emerging theories of Alzheimer’s disease propose that Alzheimer’s disease is an autoimmune disease,144 which is supported by many of the mechanisms of Tregs used to reduce Alzheimer’s disease pathologies.41,42,59

One issue of using immunosuppressants is that they can weaken the whole body’s immune system and leave patients vulnerable to infections.145 This concern is highlighted by Ochs et al., who showed that decreasing Treg levels can assist with immunity against pathogenic microorganisms,146 suggesting increasing Treg levels may decrease immunity against infections. Supporting this, one study using a viral model for multiple sclerosis reported higher Treg frequencies to suppress immune response and increased viral replication; however, it was also found that Tregs did reduce immunopathology by reducing neuroinflammation.147 Furthermore, a 2021 study has suggested a link between mortality and the routine use of immunosuppressants in SARS-CoV-2 (COVID-19) patients.148 However, Harden et al.149 studied the use of Treg therapy in kidney transplants and concluded that Treg therapy was safe and resulted in fewer incidences of opportunistic infection in comparison to patients who were treated with other immunosuppressants. Despite this suggesting Treg infusions are safe, it is important to note that Harden et al. only show that Treg infusion are safer than other immunosuppressants that are typically associated with high risks.145 Whilst there are currently no data available on these risks in Alzheimer’s disease, most studies on other diseases appear to be consistent in findings that increased levels of Tregs result in increased risk of infection,146,147,149 suggesting that more research and consideration of risks is vital to produce a safe treatment.

As people age, their immune system naturally weakens.150 This is an important consideration relating to Alzheimer’s disease treatment as the prevalence of Alzheimer’s disease also increases with age14‐16; thus, many of the older patients may not be suitable candidates for immunosuppressant Treg infusions and concerns with safety of potential drugs arise. Studies have demonstrated that immunosuppressants can have dangerous consequences in older patients who may have compromised immune systems.151 However, more recent studies on the use of immunosuppressants to treat Crohn’s disease found no difference in the efficacy and safety of immunosuppressants between older and younger patients.152 Discrepancies in the data imply that the use of Tregs in older patients with Alzheimer’s disease may need to be assessed on an individual basis to assess the condition of their immune system to determine whether patients are at high risk of infection and side effects.

Cancer and autoimmunity

One of the biggest concerns with ex vivo expanded Treg infusions is the potential increased risk for cancer. Due to the immunosuppressive role of Tregs, theoretical speculations about the relationship between Tregs and cancer arise (Fig. 3). Whilst the literature on increasing Treg levels as a therapeutic for Alzheimer’s disease fails to mention this potential problem, other studies have found an increased frequency of Tregs in peripheral blood of cancer patients compared with healthy controls. These studies also show that Tregs accumulate within tumours, implying there is a clear link between Treg levels and cancer.155,156 One study by Curiel et al.153 describes increased tumour Tregs to predict reduced survival and treatment response, suggesting that Tregs may a significant role in cancer development. Conversely, another study showed that Treg levels present in renal cancer tumours did not predict survival,157 with a third study finding that an increased number of Tregs predicted improved survival in Hodgkin lymphoma patients.158,159 The inconsistencies in the data from these studies may be due to the differences in cancer types or the methodology used in these studies used to identify Tregs using Foxp3 expression.153‐158 Not all Foxp3-expressing cells are functional Tregs.160 Thus, without a functional test, inaccuracies in identifying Tregs may occur. However, it remains necessary for research to investigate the relationship between Tregs and cancer further before moving forward to clinical trials using Tregs to treat Alzheimer’s disease (Figure 3).

Figure 3.

Figure 3

Balancing autoimmunity and cancer. Represent the fine balance of regulating the immune response and maintaining homeostasis in tissues. In immunotherapies, excessive suppression of the immune system can result in an increased risk of cancer,153 whereas an excessive enhancement of the immune system can result in autoimmune diseases.145 Image adapted from Desai et al.145 Singh et al.152, Curiel et al.153 and 154.

Furthermore, similarly to AD,14‐16 cancer is often considered to be an age-related disease.161 Meaning that patients with Alzheimer’s disease are already at a higher risk of developing cancer than the average population,162 and Tregs may also increase this risk further. It is, therefore, recommended that any clinical trials involving increasing Tregs in patients with Alzheimer’s disease should carefully consider this potential risk when selecting participants. This major limitation in the use of Treg therapies is largely ignored by animal model studies, although the reason for this remains unclear. This may be due to the use of antigen-specific technologies, although there is still a risk of non-specific immunosuppression with the use of polyclonal Tregs.39,163

Lack of understanding of mechanism of suppression, survival and plasticity

Due to the potential risks associated with overexpression of Tregs, there is a clear need to understand the mechanism with Tregs and their interactions with other cell types. The high need for a balance within multiple aspects of the treatments means that an understanding of how these cells can survive or be suppressed is essential to produce a safe and effective drug. For example, the immunosuppressant mechanisms can lead to an increased risk of cancer or reduced ability to fight infections.155,156 Tregs have been shown to exhibit high levels of plasticity due to interactions with cytokines. For example, a subset of Tregs has been shown to lose expression of Foxp2 in a depletion of IL-2 and pro-inflammatory cytokines. This results in a change in phenotype to an effector T cell.164,165 Instability of the Foxp3 protein is a concern for researchers exploring Treg-based therapies, as the differentiation of Tregs into effector T cells (eTregs) could lead to increased inflammation.125 IL-2 could potentially be used to prevent plasticity of Tregs, due to the role played in Tregs survival and stability.110,111 IL-2 has been used in combination with Treg therapies to increase the longevity of Treg survival,60,166 demonstrating the potential to target Treg control using IL-2. Despite this, the mechanisms of suppression, survival and plasticity of Tregs remain partially unknown. Research focused on better understanding the mechanisms behind Treg cell stability are needed to control and retain stable Tregs to produce a predictable, safe and effective treatment for Alzheimer’s disease.

Limited long-term effects

Long-term effects are an important factor to consider when assessing the safety of any new treatment. Due to the novelty of Treg therapies, there are limited data available on the long-term impact of these therapies. A Treg adoptive cell therapy trial in renal transplant patients, where patients received ex vivo expanded polyclonal Tregs, reported no record of adverse effects, infections, or rejections 2 years post-transplant.163 The data from this study provide enough information to support the safety of Treg therapy in Alzheimer’s disease clinical trials, although longitudinal studies over longer periods of time would confirm this. In 2018, Thonhoff et al.167 published a study on Treg infusions in 3 ALS patients and found that infusions slowed progression rates of disease, again suggesting that Treg infusions are safe and effective in treating neuroinflammation. Although Alzheimer’s disease and ALS are vastly different in many ways, both conditions are characterized by neuroinflammation and activation of microglia. Thus, Thonhoff et al.167 provide promising results for the therapeutic potential of Treg infusions in AD. Clinical trials for Treg therapies are yet to commence; however, the data from studies in other diseases show that treatment should be safe and effective in Alzheimer’s disease.

Given the role of oxidative stress, it is apparent that Tregs that differentiate after the infusion treatments would continue to have the same issues of reduces frequencies and function. Few trials have addressed the long-term survival of Tregs from infusions, due to the increased apoptosis due to low IL-2 levels,168 and they may not be able to survive as intended. As previously mentioned, clinical trials have established that Treg populations can be restored and support the suppressor function of Tregs.64 Treatment using ex vivo expanded Tregs may require continuous treatment for the desired effects, although it is unclear how frequently infusions would be necessary to maintain Treg levels. However, mice model trials60 and a clinical trial exploring a combined therapy of Tregs, with a dose of IL-2 has been recently published, demonstrating the beneficial effects of IL-2 on Treg functions.166 It is unclear whether further doses of IL-2 were required, although this demonstrates a potential resolution to this issue that needs to be investigated further.

A 2017 study described being unable to explore the functions of Tregs due to an insufficient number of cells from participants.58 This highlights another issue with this type of treatment. Many cells are required for polyclonal expansion and if doctors cannot extract enough cells to expand, patients may not be suitable for treatment using ex vivo expansion techniques,163 although adoptive transfer may still be feasible.

Cost of treatment

When investigating a potential new treatment for a debilitating disease such as Alzheimer’s disease, therapies using ex vivo expanded cell technologies are notoriously expensive. Finding ways to reduce these costs and make treatments assessable is crucial to facilitate the widespread usage and success of new technologies. In the UK, the National Institute for Health and Clinical Excellence (NICE) uses an Incremental Cost Effectiveness Ratio (ICER) of up to £30 000 per quality-adjusted life year (QALY) as the system to approve new drug therapies.169 Given that a similar Treg therapy for cancer (Carvykti) reportedly costs $465 000 for a single infusion87 and that there is evidence discussed above to suggest that Treg therapies in Alzheimer’s disease would require multiple infusions, the likelihood of such therapies being approved in the UK appears next to impossible. This highlights the importance of exploring new techniques and methods to discover assessable drug therapies for all. Whilst Tregs can prevent Alzheimer’s disease symptoms and pathologies, the search for a cost-effective alternative to Treg infusions is vital in the treatment of Alzheimer’s disease globally. It should be mentioned that in vivo Treg expansion therapy using IL-2 immunotherapy is a cheaper and viable option compared to ex vivo Treg expansion therapy.67

Conclusion and future research directions

Within the past decade, there has been a shift in focus in Alzheimer’s disease research towards immunotherapy and the therapeutic potential of Treg-based therapies and researchers remain cautiously optimistic about the potential of these immunotherapy approaches. Given the current literature, it can be concluded that increasing levels of Tregs can prevent neuroinflammation and the hallmark symptoms and pathologies of Alzheimer’s disease; however, the plethora of interactions involving Tregs are not fully understood. Promising results have demonstrated the ability of Tregs to reduce neuroinflammation in Alzheimer’s disease models. This study is unable to encompass the entire web of cellular pathways in which Tregs play a role in, however, a few significant pathways mentioned in the literature have been explored. Treg therapies act to modulate key immune components of the CNS, such as microglia, astrocytes and cytokines, to reduce Aβ and tau pathologies in animal models. Furthermore, Treg infusions have been demonstrated to slow CD and the progression of memory loss in Alzheimer’s disease mice models, highlighting the undeniable potential of the use of Treg therapies for Alzheimer’s disease. However, the inconsistencies of findings between studies demonstrate a lack of understanding of these mechanisms.

Tregs can be modulated by various factors. This review focuses on oxidative stress as an example. Increased oxidative stress in Alzheimer’s disease is reported to result in a depletion of Tregs. However, it is difficult to determine whether this is a cause or symptom of the disease. A deeper understanding how Tregs are modulated within the CNS may provide insight into the onset of Alzheimer’s disease and new prevention techniques. It would be interesting to see further research investigating this relationship further, including the effects using Treg therapies alongside an antioxidant-rich diet and IL-2. Tregs are also modulated by many cytokines, such as IL-6 and IL-2, understanding the complex interactions of these may allow for therapies that naturally increase Treg levels without the need for infusions.

The successful use of Treg therapies in organ transplants, ALS and Crohn’s disease demonstrates the safety and efficacy of these novel treatments; however, there are limited data on the long-term consequences. Whilst the potential is promising, further research into understanding the complex mechanisms surrounding Tregs is crucial to fully understand the risks of increasing frequencies of Tregs in patients with Alzheimer’s disease. For example, without the knowledge of how these infused cells survive, migrate and proliferate, questions around the dangers of non-specific immunosuppression, autoimmunity and cancer remain. A greater understanding of Treg homeostasis and ‘normal’ function needs to be understood by comparing control groups with patients with Alzheimer’s disease to ascertain safe levels for new clinical trials.

The effects of ROS, OS and Treg function must be investigated in patients with neurogenerative disorders to ascertain if combination therapies could reduce OS while enhancing Treg function. Indeed, can the immunotherapy infuse functional Tregs or could immunotherapy enhance the environment to activate existing Tregs and the T-cell differentiation? Trials to explore further the impact of in vivo Treg expansion using IL-2 are eagerly anticipated when translated to human patients with neurodegenerative conditions.64

Further, new generations of therapy with novel approaches of combination therapies, using different cocktails of monoclonal antibodies, cytokines and CAR Tregs may prove to be successful immunotherapy approaches for Alzheimer’s disease. CAR constructs are able to bind to neurotoxic Aβ monomers, deposited extracellularly.138

A recent proof of concept study showing that adoptive transfer of specific TCR Aβ regulatory Tregs restores brain homeostasis in mice may be an important new immunotherapy strategy in human AD patients.63

It is important to note that much of the research covered in this review explores animal models and human studies need to be trialled to confirm the findings from bench to bedside. Careful consideration must be taken when selecting participants for clinical trials of Treg-based therapies to reduce potential risks. Although until the extensive modulation and activation triggered by Tregs is understood, researchers may not truly comprehend the long-term side effects. A potential limitation of the current study is that the scope may be too broad to provide a comprehensive review of the impact of Tregs. However, Alzheimer’s disease progression is caused by a multitude of factors, and it appears that Treg therapies can target many of these, suggesting a bright future as a potential treatment for AD.

Acknowledgements

The graphical abstract was created in BioRender. Houldsworth, A. (2025) https://biorender.com/i61g810

Contributor Information

Victoria Abbott, Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK.

Benjamin E Housden, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK.

Annwyne Houldsworth, Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK; Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK.

Funding

None declared.

Competing interests

The authors report no competing interests.

Data availability

Data sharing is not applicable to this article as no new data were created or analysed in this study.

References

  • 1. NHS . Alzheimer’s disease—NHS. 2021. Accessed 22 March 2024. https://www.nhs.uk/conditions/alzheimers-disease/
  • 2. Breijyeh  Z, Karaman  R. Comprehensive review on Alzheimer’s disease: Causes and treatment. Molecules. 2020;25:5789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Rostagno  AA. Pathogenesis of Alzheimer’s disease. Int J Mol Sci.  2022;24:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Prince  M, Knapp  M, Guerchet  M, McCrane  P. Dementia UK: Update (2nd ed). Alzheimer's Society; 2014. [Google Scholar]
  • 5. Alzheimer’s disease facts and figures. Alzheimer’s & Dementia. 2021;17:327–406. [DOI] [PubMed] [Google Scholar]
  • 6. Panegyres  PK, Chen  HY. Differences between early and late onset Alzheimer’s disease. Am J Neurodegener Dis. 2013;2:300. [PMC free article] [PubMed] [Google Scholar]
  • 7. Martin  E, Velayudhan  L. Neuropsychiatric symptoms in mild cognitive impairment: A literature review. Dement Geriatr Cogn Disord. 2020;49:146–155. [DOI] [PubMed] [Google Scholar]
  • 8. Kasper  S, Bancher  C, Eckert  A, et al.  Management of mild cognitive impairment (MCI): The need for national and international guidelines. World J Biol Psychiatry.  2020;21:579–594. [DOI] [PubMed] [Google Scholar]
  • 9. Petersen  RC, Smith  GE, Waring  SC, Ivnik  RJ, Kokmen  E, Tangelos  EG. Aging, memory, and mild cognitive impairment. Int Psychogeriatr. 1997;9:65–69. [DOI] [PubMed] [Google Scholar]
  • 10. Davis  M, O`Connell  T, Johnson  S, et al.  Estimating Alzheimer’s disease progression rates from normal cognition through mild cognitive impairment and stages of dementia. Curr Alzheimer Res. 2018;15:777–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Deture  MA, Dickson  DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener.  2019;14:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Fan  L, Mao  C, Hu  X, et al.  New insights into the pathogenesis of Alzheimer’s disease. Front Neurol. 2020;10:1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Heyman  A, Wilkinson  WE, Hurwitz  BJ, et al.  Alzheimer’s disease: Genetic aspects and associated clinical disorders. Ann Neurol. 1983;14:507–515. [DOI] [PubMed] [Google Scholar]
  • 14. Henderson  AS. The risk factors for Alzheimer’s disease: A review and a hypothesis. Acta Psychiatr Scand. 1988;78:257–275. [DOI] [PubMed] [Google Scholar]
  • 15. Ferri  CP, Prince  M, Brayne  C, et al.  Global prevalence of dementia: A Delphi consensus study. Lancet. 2005;366:2112–2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Armstrong  RA, Richard  P, Armstrong  A. Risk factors for Alzheimer’s disease. Folia Neuropathol. 2019;57:87–105. [DOI] [PubMed] [Google Scholar]
  • 17. Bekris  LM, Yu  CE, Bird  TD, Tsuang  DW. Genetics of Alzheimer disease. J Geriatr Psychiatry Neurol. 2010;23:213–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Giri  M, Zhang  M, Lü  Y. Genes associated with Alzheimer’s disease: An overview and current status. Clin Interv Aging. 2016;11:665–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Shea  TB, Rogers  E, Ashline  D, Ortiz  D, Sheu  M-S. Apolipoprotein E deficiency promotes increased oxidative stress and compensatory increases in antioxidants in brain tissue.  Free Radic Biol Med. 2002;33:1115–1120. [DOI] [PubMed] [Google Scholar]
  • 20. NHS . Alzheimer’s disease—Treatment. 2021. Accessed 26 March 2024. https://www.nhs.uk/conditions/alzheimers-disease/treatment/
  • 21. Valiukas  Z, Ephraim  R, Tangalakis  K, Davidson  M, Apostolopoulos  V, Feehan  J. Immunotherapies for Alzheimer’s disease—A review. Vaccines (Basel).  2022;10:1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. McGleenon  BM, Dynan  KB, Passmore  AP. Acetylcholinesterase inhibitors in Alzheimer’s disease. Br J Clin Pharmacol. 1999;48:471–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. McShane  R, Westby  MJ, Roberts  E, et al.  Memantine for dementia. Cochrane Database Syst Rev. 2019;3:CD003154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Hampel  HJ, Us  E, Bokde  ALW, Buschert  V, Bokde  ALW, Hampel  H. Cognitive intervention in Alzheimer disease. Nat Rev Neurol. 2010;6:508–517. [DOI] [PubMed] [Google Scholar]
  • 25. Wilkinson  D, Fox  NC, Barkhof  F, Phul  R, Lemming  O, Scheltens  P. Memantine and brain atrophy in Alzheimer’s disease: A 1-year randomized controlled trial. J Alzheimers Dis. 2012;29:459–469. [DOI] [PubMed] [Google Scholar]
  • 26. Fox  C, Crugel  M, Maidment  I, et al.  Efficacy of memantine for agitation in Alzheimer’s dementia: A randomised double-blind placebo controlled trial. PLoS One. 2012;7:e35185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. van Dyck  CH, Swanson  CJ, Aisen  P, et al.  Lecanemab in early Alzheimer’s disease. N Engl J Med.  2023;388:9–21. [DOI] [PubMed] [Google Scholar]
  • 28. U.S Food and Drink Administration. FDA Grants Accelerated Approval for Alzheimer’s Disease Treatment | FDA. n.d. Accessed 26 March 2024. https://www.fda.gov/news-events/press-announcements/fda-grants-accelerated-approval-alzheimers-disease-treatment
  • 29. Qiao  Y, Chi  Y, Zhang  Q, Ma  Y. Safety and efficacy of lecanemab for Alzheimer’s disease: A systematic review and meta-analysis of randomized clinical trials. Front Aging Neurosci. 2023;15:1169499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Alzheimer's Research UK. Development and sale of Alzheimer’s drug, aducanumab, ceased—Alzheimer’s Research UK. 2024. Accessed 27 March 2024. https://www.alzheimersresearchuk.org/news/development-and-sale-of-alzheimers-drug-aducanumab-ceased/
  • 31. Ross  EL, Weinberg  MS, Arnold  SE. Cost-effectiveness of Aducanumab and Donanemab for early Alzheimer disease in the US. JAMA Neurol. 2022;79:478–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Cummings  J, Aisen  P, Lemere  C, Atri  A, Sabbagh  M, Salloway  S. Aducanumab produced a clinically meaningful benefit in association with amyloid lowering. Alzheimers Res Ther. 2021;13:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Knopman  DS, Jones  DT, Greicius  MD. Failure to demonstrate efficacy of aducanumab: An analysis of the EMERGE and ENGAGE trials as reported by Biogen, December 2019. Alzheimers Dement. 2021;17:696–701. [DOI] [PubMed] [Google Scholar]
  • 34. Haeberlein  SB, von Hehn  C, Tian  Y, et al.  Emerge and engage topline results: Phase 3 studies of aducanumab in early Alzheimer’s disease. Alzheimer’s & Dementia. 2020;16:e047259. [Google Scholar]
  • 35. Sims  JR, Zimmer  JA, Evans  CD, et al.  TRAILBLAZER-ALZ 2 investigators, Donanemab in early symptomatic Alzheimer disease: The TRAILBLAZER-ALZ 2 randomized clinical trial. JAMA. 2023;330:512–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Jevtic  S, Sengar  AS, Salter  MW, McLaurin  JA. The role of the immune system in Alzheimer disease: Etiology and treatment. Ageing Res Rev. 2017;40:84–94. [DOI] [PubMed] [Google Scholar]
  • 37. Sinyor  B, Mineo  J, Ochner  C. Alzheimer’s disease, inflammation, and the role of antioxidants. J Alzheimers Dis Rep. 2020;4:175–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Heneka  MT, Carson  MJ, Khoury  JE, et al.  Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Faridar  A, Thome  AD, Zhao  W, et al.  Restoring regulatory T-cell dysfunction in Alzheimer’s disease through ex vivo expansion.  Brain Commun. 2020;2:fcaa112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Vaeth  M, Wang  YH, Eckstein  M, et al.  Tissue resident and follicular Treg cell differentiation is regulated by CRAC channels. Nat Commun.  2019;10:1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Faridar  A, Vasquez  M, Thome  AD, et al.  Ex vivo expanded human regulatory T cells modify neuroinflammation in a preclinical model of Alzheimer’s disease. Acta Neuropathol Commun. 2022;10:144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Kondělková  K, Vokurková  D, Krejsek  J, Borská  L, Fiala  Z, Ctirad  A. Regulatory T cells (Treg) and their roles in immune system with respect to immunopathological disorders. Acta Medica (Hradec Kralove, Czech Republic). 2016;53:73–77. [DOI] [PubMed] [Google Scholar]
  • 43. Askin  B, Wegmann  S. Interaction of microglia and T cells: A deadly duo behind Tau-mediated neurodegeneration. Signal Transduct Target Ther.  2023;8:317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Kustrimovic  N, Comi  C, Magistrelli  L, et al.  Parkinson’s disease patients have a complex phenotypic and functional Th1 bias: Cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naïve and drug-treated patients. J Neuroinflammation. 2018;15:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Togashi  Y, Shitara  K, Nishikawa  H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat Rev Clin Oncol.  2019;16(6):356–371. [DOI] [PubMed] [Google Scholar]
  • 46. Stym-Popper  G, Matta  K, Chaigneau  T, et al.  Dorothée, regulatory T cells decrease C3-positive reactive astrocytes in Alzheimer-like pathology. J Neuroinflammation. 2023;20:164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Shimojima  Y, Kishida  D, Ichikawa  T, Takamatsu  R, Nomura  S, Sekijima  Y. Oxidative stress promotes instability of regulatory T cells in antineutrophil cytoplasmic antibody-associated vasculitis. Front Immunol. 2021;12:789740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Hong  J, Kim  BS. Regulatory T cell-mediated tissue repair. Adv Exp Med Biol. 2018;1064:221–233. [DOI] [PubMed] [Google Scholar]
  • 49. Olson  KE, Mosley  RL, Gendelman  HE. The potential for treg-enhancing therapies in nervous system pathologies. Clin Exp Immunol. 2023;211:108–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Liston  A, Dooley  J, Yshii  L. Brain-resident regulatory T cells and their role in health and disease.  Immunol Lett. 2022;248:26–30. [DOI] [PubMed] [Google Scholar]
  • 51. Yang  HJ, Park  SY, Baek  H, et al.  Adoptive therapy with amyloid-β specific regulatory T cells alleviates Alzheimer’s disease. Theranostics. 2022;12:7668–7680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Smigiel  KS, Srivastava  S, Stolley  JM, Campbell  DJ. Regulatory T cell homeostasis: Steady-state maintenance and modulation during inflammation. Immunol Rev. 2014;259:40–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Oberstein  TJ, Taha  L, Spitzer  P, et al.  Imbalance of circulating Th17 and regulatory T cells in Alzheimer’s disease: A case control study. Front Immunol. 2018;9:1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Ciccocioppo  F, Lanuti  P, Pierdomenico  L, et al.  The characterization of regulatory T-cell profiles in Alzheimer’s disease and multiple sclerosis. Sci Rep.  2019;9(1):8788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Saresella  M, Calabrese  E, Marventano  I, et al.  PD1 negative and PD1 positive CD4+ T regulatory cells in mild cognitive impairment and Alzheimer’s disease. J Alzheimers Dis. 2010;21:927–938. [DOI] [PubMed] [Google Scholar]
  • 56. Toomer  KH, Lui  JB, Altman  NH, Ban  Y, Chen  X, Malek  TR. Essential and non-overlapping IL-2Rα-dependent processes for thymic development and peripheral homeostasis of regulatory T cells. Nat Commun. 2019;10:1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Rosenkranz  D, Weyer  S, Tolosa  E, et al.  Higher frequency of regulatory T cells in the elderly and increased suppressive activity in neurodegeneration. J Neuroimmunol. 2007;188:117–127. [DOI] [PubMed] [Google Scholar]
  • 58. Le Page  A, Garneau  H, Dupuis  G, et al.  Differential phenotypes of myeloid-derived suppressor and T regulatory cells and cytokine levels in amnestic mild cognitive impairment subjects compared to mild Alzheimer diseased patients. Front Immunol. 2017;8:275965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Baek  H, Ye  M, Kang  GH, et al.  Neuroprotective effects of CD4 + CD25 + foxp3 + regulatory T cells in a 3xTg-AD Alzheimer’s disease model. Oncotarget. 2016;7:69347–69357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Dansokho  C, Ait Ahmed  D, Aid  S, et al.  Regulatory T cells delay disease progression in Alzheimer-like pathology. Brain. 2016;139:1237–1251. [DOI] [PubMed] [Google Scholar]
  • 61. Schwartz  M, Deczkowska  A. Neurological disease as a failure of brain–immune crosstalk: The multiple faces of neuroinflammation. Trends Immunol. 2016;37:668–679. [DOI] [PubMed] [Google Scholar]
  • 62. Yang  Y, Yang  J, Cho  H, et al.  Therapeutic effects of Aβ-specific regulatory T cells in Alzheimer’s disease: A study in 5xFAD mice. Int J Mol Sci.  2024;25:783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Yeapuri  P, Machhi  J, Lu  Y, et al.  Amyloid-β specific regulatory T cells attenuate Alzheimer’s disease pathobiology in APP/PS1 mice. Mol Neurodegener. 2023;18:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Chinen  T, Kannan  AK, Levine  AG, et al.  An essential role for the IL-2 receptor in Treg cell function. Nat Immunol. 2016;17:1322–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Thonhoff  JR, Beers  DR, Zhao  W, et al.  A phase 1 proof-of-concept study evaluating safety, tolerability, and biological marker responses with combination therapy of CTLA4-Ig and interleukin-2 in amyotrophic lateral sclerosis.  Front Neurol. 2024;15:1415106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Camu  W, Mickunas  M, Veyrune  J-L, et al.  Repeated 5-day cycles of low dose aldesleukin in amyotrophic lateral sclerosis (IMODALS): A phase 2a randomised, double-blind, placebo-controlled trial.  EBioMedicine. 2020;59:102844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Faridar  A, Eid  AM, Thome  AD, et al.  A phase 1 open-label pilot study of low-dose interleukine-2 immunotherapy in patients with Alzheimer’s disease.  Transl Neurodegener. 2023;12:54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Houldsworth  A. Role of oxidative stress in neurodegenerative disorders: A review of reactive oxygen species and prevention by antioxidants. Brain Commun. 2023;6:fcad356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Chen  Y, Qin  C, Huang  J, et al.  The role of astrocytes in oxidative stress of central nervous system: A mixed blessing. Cell Prolif. 2020;53:e12781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Esposito  L, Raber  J, Kekonius  L, et al.  Reduction in mitochondrial superoxide dismutase modulates Alzheimer’s disease-like pathology and accelerates the onset of behavioral changes in human amyloid precursor protein transgenic mice. J Neurosci.  2006;26:5167–5179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Pritam  P, Deka  R, Bhardwaj  A, et al.  Antioxidants in Alzheimer’s disease: Current therapeutic significance and future prospects. Biology (Basel). 2022;11:212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Pizzino  G, Irrera  N, Cucinotta  M, et al.  Oxidative stress: Harms and benefits for human health. Oxid Med Cell Longev. 2017;2017:8416763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Sy  M, Kitazawa  M, Medeiros  R, et al.  Inflammation induced by infection potentiates tau pathological features in transgenic mice. Am J Pathol. 2011;178:2811–2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Liguori  I, Russo  G, Curcio  F, et al.  Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Franzoni  F, Scarfò  G, Guidotti  S, Fusi  J, Asomov  M, Pruneti  C. Oxidative stress and cognitive decline: The neuroprotective role of natural antioxidants. Front Neurosci. 2021;15:729757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Sakaguchi  S, Sakaguchi  N. Regulatory T cells in immunologic self-tolerance and autoimmune disease. Int Rev Immunol. 2005;24:211–226. [DOI] [PubMed] [Google Scholar]
  • 77. Saxena  A, Lakshmi  J, Bhattacharjya  R, Singh  PK, Mishra  B, Tiwari  A. Marine Antioxidants: Preparations, Syntheses, Appl. Elsevier; 2022:89–118. [Google Scholar]
  • 78. De Leo  ME, Borrello  S, Passantino  M, et al.  Oxidative stress and overexpression of manganese superoxide dismutase in patients with Alzheimer’s disease. Neurosci Lett. 1998;250:173–176. [DOI] [PubMed] [Google Scholar]
  • 79. Frassetto  L, Morris  RC, Sellmeyer  DE, Todd  K, Sebastian  A. Diet, evolution and aging–the pathophysiologic effects of the post-agricultural inversion of the potassium-to-sodium and base-to-chloride ratios in the human diet. Eur J Nutr. 2001;40:200–213. [DOI] [PubMed] [Google Scholar]
  • 80. Mathew  B, Biju  R. Neuroprotective effects of garlic a review. Libyan J Med. 2008;3:23–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Arslan  J, Jamshed  H, Qureshi  H. Early detection and prevention of Alzheimer’s disease: Role of oxidative markers and natural antioxidants. Front Aging Neurosci. 2020;12:231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Fakhoury  M. Microglia and astrocytes in Alzheimer’s disease: Implications for therapy. Curr Neuropharmacol. 2018;16:508–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Park  JS, Kam  TI, Lee  S, et al.  Blocking microglial activation of reactive astrocytes is neuroprotective in models of Alzheimer’s disease. Acta Neuropathol Commun. 2021;9:78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Lee  DC, Rizer  J, Selenica  MLB, et al.  LPS- induced inflammation exacerbates phospho-tau pathology in rTg4510 mice. J Neuroinflammation. 2010;7:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Chay  K-O, Nam Koong  KY, Hwang  S, Kim  J-K, Bae  CS. NADPH oxidase mediates β-amyloid peptide-induced neuronal death in mouse cortical cultures. Chonnam Med J. 2017;53:196–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Park  S-Y, Yang  H, Ye  M, et al.  Neuroprotective effects of ex vivo-expanded regulatory T cells on trimethyltin-induced neurodegeneration in mice.  J Neuroinflammation. 2022;19:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Jaber  N. FDA Approves Carvykti CAR T-Cell Therapy for Multiple Myeloma—NCI, National Cancer Institute 2022. Accessed 8 April 2024. https://www.cancer.gov/news-events/cancer-currents-blog/2022/fda-carvykti-multiple-myeloma
  • 88. Chakrabarty  P, Li  A, Ceballos-Diaz  C, et al.  IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior.  Neuron. 2015;85:519–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Ito  M, Komai  K, Mise-Omata  S, et al.  Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature. 2019; 565:246–250. [DOI] [PubMed] [Google Scholar]
  • 90. Lee  CYD, Landreth  GE. The role of microglia in amyloid clearance from the AD brain. J Neural Transm (Vienna). 2010;117:949–960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Muzio  L, Viotti  A, Martino  G. Microglia in neuroinflammation and neurodegeneration: From understanding to therapy. Front Neurosci. 2021;15:742065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Daria  A, Colombo  A, Llovera  G, et al.  Young microglia restore amyloid plaque clearance of aged microglia. EMBO J. 2017;36:583–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Hickman  SE, Allison  EK, Khoury  JE. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. J Neurosci. 2008;28:8354–8360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Rajendran  L, Paolicelli  RC. Microglia-mediated synapse loss in Alzheimer’s disease. J Neurosci. 2018;38:2911–2919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Khandelwal  PJ, Dumanis  SB, Herman  AM, Rebeck  GW, Moussa  CEH. Wild type and P301L mutant Tau promote neuro-inflammation and α-synuclein accumulation in lentiviral gene delivery models. Mol Cell Neurosci. 2012;49:44–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Kang  SS, Ebbert  MTW, Baker  KE, et al.  Microglial translational profiling reveals a convergent APOE pathway from aging, amyloid, and tau. J Exp Med. 2018;215:2235–2245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Jäntti  H, Sitnikova  V, Ishchenko  Y, et al.  Microglial amyloid beta clearance is driven by PIEZO1 channels. J Neuroinflammation. 2022;19:147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Campbell  DJ. Control of regulatory T cell migration, function and homeostasis. J Immunol. 2015;195:2507–2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Bathina  S, Das  UN. Brain-derived neurotrophic factor and its clinical implications. Arch Med Sci. 2015;11:1164–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Saw  G, Krishna  K, Gupta  N, et al.  Epigenetic regulation of microglial phosphatidylinositol 3-kinase pathway involved in long-term potentiation and synaptic plasticity in rats. Glia. 2020;68:656–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Rauti  R, Cellot  G, D’Andrea  P, et al.  BDNF impact on synaptic dynamics: Extra or intracellular long-term release differently regulates cultured hippocampal synapses. Mol Brain. 2020;13:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Nagahara  AH, Merrill  DA, Coppola  G, et al.  Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. 2009;15:331–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Bonham  LW, Karch  CM, Fan  CC, et al.  International FTD-Genomics Consortium (IFGC), International Parkinson’s Disease Genetics Consortium (IPDGC), International Genomics of Alzheimer’s Project (IGAP), CXCR4 involvement in neurodegenerative diseases. Transl Psychiatry. 2018;8:73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Schmidt  A, Oberle  N, Krammer  PH. Molecular mechanisms of Treg-mediatedt cell suppression. Front Immunol. 2012;3:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Chakrabarty  P, Li  A, Ceballos-Diaz  C, et al.  IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron. 2015;85:519–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Kiyota  T, Ingraham  KL, Swan  RJ, Jacobsen  MT, Andrews  SJ, Ikezu  T. AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP + PS1 mice. Gene Ther.  2012;19:724–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Wyss-Coray  T, Lin  C, Yan  F, et al.  TGF-β1 promotes microglial amyloid-β clearance and reduces plaque burden in transgenic mice. Nat Med.  2001;7:612–618. [DOI] [PubMed] [Google Scholar]
  • 108. Alves  S, Churlaud  G, Audrain  M, et al.  Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain. 2017;140:aww330. [DOI] [PubMed] [Google Scholar]
  • 109. Harris  F, Berdugo  YA, Tree  T. IL-2-based approaches to Treg enhancement. Clin Exp Immunol. 2023;211:149–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Yu  A, Zhu  L, Altman  NH, Malek  TR. A low interleukin-2 receptor signaling threshold supports the development and homeostasis of T regulatory cells. Immunity. 2009;30:204–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Fan  MY, Low  JS, Tanimine  N, et al.  Differential roles of IL-2 signaling in developing versus mature Tregs. Cell Rep. 2018;25:1204–1213.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Whyte  CE, Singh  K, Burton  OT, et al.  Context-dependent effects of IL-2 rewire immunity into distinct cellular circuits. J Exp Med.  2022;219:e20212391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Yshii  L, Pasciuto  E, Bielefeld  P, et al.  Astrocyte-targeted gene delivery of interleukin 2 specifically increases brain-resident regulatory T cell numbers and protects against pathological neuroinflammation. Nat Immunol.  2022;23:878–891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Brosseron  F, Krauthausen  M, Kummer  M, Heneka  MT. Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: A comparative overview. Mol Neurobiol. 2014;50:534–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Sofroniew  MV, Vinters  HV. Astrocytes: Biology and pathology. Acta Neuropathol. 2010;119:7–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Chung  WS, Allen  NJ, Eroglu  C. Astrocytes control synapse formation, function, and elimination. Cold Spring Harb Perspect Biol. 2015;7:a020370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Chung  WS, Clarke  LE, Wang  GX, et al.  Astrocytes mediate synapse elimination through MEGF10 and MERTK pathways. Nature. 2013;504:394–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Sofroniew  MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009;32:638–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Wisniewski  HM, Wegiel  J. Spatial relationships between astrocytes and classical plaque components. Neurobiol Aging. 1991;12:593–600. [DOI] [PubMed] [Google Scholar]
  • 120. Shi  L, Sun  Z, Su  W, et al.  Treg cell-derived osteopontin promotes microglia-mediated white matter repair after ischemic stroke. Immunity. 2021;54:1527–1542.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Wang  S, Shi  Y, Zhang  Y, Yuan  F, Mao  M, Ma  J. Tregs depletion aggravates activation of astrocytes by modulating IL-10/GXP4 following cerebral infarction. Front Immunol. 2023;14:1255316. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 122. Johnstone  M, Gearing  AJH, Miller  KM. A central role for astrocytes in the inflammatory response to beta-amyloid; chemokines, cytokines and reactive oxygen species are produced. J Neuroimmunol. 1999;93:182–193. [DOI] [PubMed] [Google Scholar]
  • 123. Ries  M, Sastre  M. Mechanisms of Aβ clearance and degradation by glial cells. Front Aging Neurosci. 2016;8:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Sigal  LJ. Activation of CD8 T lymphocytes during viral infections. Encyclopedia of Immunobiology. 2016;4:286–290. [Google Scholar]
  • 125. Garber  C, Soung  A, Vollmer  LL, et al.  T cells promote microglia-mediated synaptic elimination and cognitive dysfunction during recovery from neuropathogenic flaviviruses. Nat Neurosci.  2019;22:1276–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Jorfi  M, Park  J, Hall  CK, et al.  Infiltrating CD8+ T cells exacerbate Alzheimer’s disease pathology in a 3D human neuroimmune axis model. Nat Neurosci.  2023;26(9):1489–1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Speciale  L, Calabrese  E, Saresella  M, et al.  Lymphocyte subset patterns and cytokine production in Alzheimer’s disease patients. Neurobiol Aging. 2007;28:1163–1169. [DOI] [PubMed] [Google Scholar]
  • 128. Chappert  P, Leboeuf  M, Rameau  P, et al.  Antigen-specific Treg impair CD8+ T-cell priming by blocking early T-cell expansion. Eur J Immunol. 2010;40:339–350. [DOI] [PubMed] [Google Scholar]
  • 129. McNally  A, Hill  GR, Sparwasser  T, Thomas  R, Steptoe  RJ. CD4+CD25+ regulatory T cells control CD8+ T-cell effector differentiation by modulating IL-2 homeostasis. Proc Natl Acad Sci U S A. 2011;108:7529–7534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Winford  E, Lutshumba  J, Martin  BJ, et al.  Terminally differentiated effector memory T cells associate with cognitive and AD-related biomarkers in an aging-based community cohort. Immun Ageing.  2024;21:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Zhang  H, Cao  F, Zhou  Y, Wu  B, Li  C. Peripheral immune cells contribute to the pathogenesis of Alzheimer’s disease. Mol Neurobiol. 2025;62:264–270. [DOI] [PubMed] [Google Scholar]
  • 132. Sakaguchi  S, Yamaguchi  T, Nomura  T, Ono  M. Regulatory T cells and immune tolerance. Cell. 2008;133:775–787. [DOI] [PubMed] [Google Scholar]
  • 133. Baruch  K, Rosenzweig  N, Kertser  A, et al.  Breaking immune tolerance by targeting foxp3 + regulatory T cells mitigates Alzheimer’s disease pathology. Nat Commun.  2015;6:7967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Maphis  N, Xu  G, Kokiko-Cochran  ON, et al.  Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138:1738–1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Blom  ES, Giedraitis  V, Zetterberg  H, et al.  Rapid progression from mild cognitive impairment to Alzheimer’s disease in subjects with elevated levels of Tau in cerebrospinal fluid and the APOE ε4/ε4 genotype. Dement Geriatr Cogn Disord. 2009;27:458–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Buerger  K, Ewers  M, Andreasen  N, et al.  Phosphorylated tau predicts rate of cognitive decline in MCI subjects: A comparative CSF study. Neurology. 2005;65:1502–1503. [DOI] [PubMed] [Google Scholar]
  • 137. Requejo Cier  CJ, Valentini  N, Lamarche  C. Unlocking the potential of Tregs: Innovations in CAR technology.  Front Mol Biosci. 2023;10:1267762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Saetzler  V, Riet  T, Schienke  A, et al.  Development of Beta-amyloid-specific CAR-Tregs for the treatment of Alzheimer’s disease.  Cells. 2023;12:2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Komatsu  N, Okamoto  K, Sawa  S, et al.  Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis.  Nat Med. 2014;20:62–68. [DOI] [PubMed] [Google Scholar]
  • 140. Sakowska  J, Arcimowicz  Å?, Jankowiak  M, et al.  Autoimmunity and cancer—Two sides of the same coin. Front Immunol. 2022;13:793234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Elkoshi  Z. Cancer and autoimmune diseases: A tale of two immunological opposites? . Front Immunol. 2022;13:821598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Candeias  SM, Gaipl  US. The immune system in cancer prevention, development and therapy. Anticancer Agents Med Chem. 2016;16:101–107. [DOI] [PubMed] [Google Scholar]
  • 143. Richard-Eaglin  A, Smallheer  BA. Immunosuppressive/autoimmune disorders. Nurs Clin North Am. 2018;53:319–334. [DOI] [PubMed] [Google Scholar]
  • 144. Weaver  DF. Alzheimer’s disease as an innate autoimmune disease (AD2): A new molecular paradigm. Alzheimers Dement. 2022;19:1086–1098. [DOI] [PubMed] [Google Scholar]
  • 145. Desai  RJ, Bateman  BT, Huybrechts  KF, et al.  Risk of serious infections associated with use of immunosuppressive agents in pregnant women with autoimmune inflammatory conditions: Cohort study. BMJ. 2017;356:895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Ochs  HD, Oukka  M, Torgerson  TR. TH17 cells and regulatory T cells in primary immunodeficiency diseases. J Allergy Clin Immunol. 2009;123:977–985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Martinez  NE, Karlsson  F, Sato  F, et al.  Protective and detrimental roles for regulatory T cells in a viral model for multiple sclerosis. Brain Pathol. 2014;24:436–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Myint  PK, Carter  B, Barlow-Pay  F, et al.  Routine use of immunosuppressants is associated with mortality in hospitalised patients with COVID-19. Ther Adv Drug Saf. 2021;12:2042098620985690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Harden  PN, Game  DS, Sawitzki  B, et al.  Feasibility, long-term safety, and immune monitoring of regulatory T cell therapy in living donor kidney transplant recipients. Am J Transplant.  2021;21:1603–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Lawton  G. You’re only as young as your immune system. New Sci. 2020;245:44–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Kuypers  DRJ. Immunotherapy in elderly transplant recipients. Drugs Aging.  2009;26(9):715–737. [DOI] [PubMed] [Google Scholar]
  • 152. Singh  S, Stitt  LW, Zou  G, et al.  Early combined immunosuppression may be effective and safe in older patients with Crohn’s disease: Post hoc analysis of REACT. Aliment Pharmacol Ther. 2019;49:1188–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Curiel  TJ, Coukos  G, Zou  L, et al.  Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med.  2004;10(9):942–949. [DOI] [PubMed] [Google Scholar]
  • 154. Town  Terrence. Tregs Suppress Astrogliosis and Improve Recovery After Stroke | ALZFORUM, n.d. Accessed 8 April 2024. https://www.alzforum.org/news/research-news/tregs-suppress-astrogliosis-and-improve-recovery-after-stroke
  • 155. Curiel  TJ. Regulatory T cells and treatment of cancer. Curr Opin Immunol. 2008;20:241–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Zou  W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6:295–307. [DOI] [PubMed] [Google Scholar]
  • 157. Siddiqui  SA, Frigola  X, Bonne-Annee  S, et al.  Tumor-infiltrating Foxp3-CD4+CD25+ T cells predict poor survival in renal cell carcinoma. Clin Cancer Res. 2007;13:2075–2081. [DOI] [PubMed] [Google Scholar]
  • 158. Tzankov  A, Meier  C, Hirschmann  P, Went  P, Pileri  SA, Dirnhofer  S. Correlation of high numbers of intratumoral FOXP3 + regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin’s lymphoma. Haematologica. 2008;93:193–200. [DOI] [PubMed] [Google Scholar]
  • 159. Carreras  J, Lopez-Guillermo  A, Fox  BC, et al.  High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood. 2006;108:2957–2964. [DOI] [PubMed] [Google Scholar]
  • 160. Curiel  TJ. Regulatory T-cell development: Is Foxp3 the decider?. Nat Med. 2007;13:250–253. [DOI] [PubMed] [Google Scholar]
  • 161. White  MC, Holman  DM, Boehm  JE, Peipins  LA, Grossman  M, Jane Henley  S. Age and cancer risk: A potentially modifiable relationship. Am J Prev Med. 2014;46:S7–S15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Risk Factors: Age—NCI. n.d. Accessed 4 April 2024. https://www.cancer.gov/about-cancer/causes-prevention/risk/age
  • 163. Zhang  Q, Lu  W, Liang  C-L, et al.  Chimeric antigen receptor (CAR) treg: A promising approach to inducing immunological tolerance.  Front Immunol. 2018;9:2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. NovalRivas  M, Burton  OT, Wise  P, et al.  Regulatory T cell reprogramming toward a th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015;42:512–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Zhou  X, Bailey-Bucktrout  SL, Jeker  LT, et al.  Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009;10:1000–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Theil  A, Tuve  S, Oelschlägel  U, et al.  Adoptive transfer of allogeneic regulatory T cells into patients with chronic graft-versus-host disease. Cytotherapy. 2015;17:473–486. [DOI] [PubMed] [Google Scholar]
  • 167. Thonhoff  JR, Beers  DR, Zhao  W, et al.  Expanded autologous regulatory T-lymphocyte infusions in ALS: A phase I, first-in-human study, neurology(R). Neuroimmunol Neuroinflam. 2018;5:e465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Alzhrani  A, Bottomley  M, Wood  K, Hester  J, Issa  F. Identification, selection, and expansion of non-gene modified alloantigen-reactive Tregs for clinical therapeutic use. Cell Immunol. 2020;357:104214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. McCabe  C, Claxton  K, Culyer  AJ. The NICE cost-effectiveness threshold: What it is and what that means. Pharmacoeconomics. 2008;26:733–744. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analysed in this study.


Articles from Brain Communications are provided here courtesy of Oxford University Press

RESOURCES