Skip to main content

Modeling the neuroimmune system in Alzheimer’s and Parkinson’s diseases

Abstract

Parkinson’s disease (PD) and Alzheimer’s disease (AD) are neurodegenerative disorders caused by the interaction of genetic, environmental, and familial factors. These diseases have distinct pathologies and symptoms that are linked to specific cell populations in the brain. Notably, the immune system has been implicated in both diseases, with a particular focus on the dysfunction of microglia, the brain’s resident immune cells, contributing to neuronal loss and exacerbating symptoms. Researchers use models of the neuroimmune system to gain a deeper understanding of the physiological and biological aspects of these neurodegenerative diseases and how they progress. Several in vitro and in vivo models, including 2D cultures and animal models, have been utilized. Recently, advancements have been made in optimizing these existing models and developing 3D models and organ-on-a-chip systems, holding tremendous promise in accurately mimicking the intricate intracellular environment. As a result, these models represent a crucial breakthrough in the transformation of current treatments for PD and AD by offering potential for conducting long-term disease-based modeling for therapeutic testing, reducing reliance on animal models, and significantly improving cell viability compared to conventional 2D models. The application of 3D and organ-on-a-chip models in neurodegenerative disease research marks a prosperous step forward, providing a more realistic representation of the complex interactions within the neuroimmune system. Ultimately, these refined models of the neuroimmune system aim to aid in the quest to combat and mitigate the impact of debilitating neuroimmune diseases on patients and their families.

Background

Neuroinflammation or central nervous system (CNS) inflammation is present in neurodegenerative disorders. It can contribute to the progression of pathologies like Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis, Huntington’s disease, and multiple sclerosis [1]. In these pathologies, not only is the CNS affected, but alterations of the immune system have also been shown. Under healthy conditions, the immune system is involved in building, maintaining, and repairing the CNS, particularly through extracellular debris’ phagocytosis and production of neurotrophic factors. In neurodegenerative diseases, the immune system sustains neuroinflammation, leading to a pathology progression [2]. This review focuses on AD and PD, complex neurodegenerative disorders that have a profound impact on individuals and society. The interaction between neuroinflammation, immune dysregulation, and the accumulation of abnormal protein aggregates, such as amyloid-beta and Tau in AD and α-synuclein in PD, is complex and has remained largely elusive. Modulating the immune system might offer a possible path toward finding effective treatments for these diseases [3, 4]. Thus, a comprehensive understanding of the pathophysiology of AD and PD, involving complex interactions between the immune system and the CNS, is paramount for treatments, and for the generation of functional models to study these conditions. This review aims to highlight the significance of understanding the pathophysiology and biology of AD and PD and their connections to the immune system. By unraveling the underlying mechanisms and pathways involved in these diseases, we can gain novel insights into potential therapeutic targets and develop strategies for early detection and intervention.

Introduction

Alzheimer’s disease and Parkinson’s disease etiology

AD and PD are among the most prevalent neurodegenerative conditions, inflicting a detrimental outcome on individuals and society worldwide. The impact of these diseases is aggravated by the lack of effective treatments, resulting in increased morbidity and mortality, and representing consequently a major public health challenge [5]. The global socioeconomic bearing of neurodegenerative diseases is increasing significantly as life expectancy rises. Despite extensive research, the limitations of current approaches can be attributed to the complexity of the cellular and molecular mechanisms underlying these diseases. Common pathophysiological mechanisms include abnormal protein dynamics, oxidative stress, mitochondrial dysfunction, DNA damage, neurotrophin dysfunction and exacerbated or altered neuroinflammatory processes (Fig. 1) [6,7,8]. Recently, the discovery of increased circulating levels of pro-inflammatory markers in AD and PD patients suggested a strong interplay between various neuroimmune and inflammatory mechanisms [9, 10]. Therefore, it is essential to understand these pathologies and explore their mechanistic underpinnings, particularly via the development of complete and detailed in vitro models (Fig. 2).

Fig. 1
figure 1

Pathophysiology of Alzheimer’s disease and Parkinson’s disease. The schematic shows the risk factors and features associated with both disease conditions. Image created with BioRender.com

Fig. 2
figure 2

Comparison of healthy versus pathological neuroimmune system: in healthy neuroimmune system (1) microglia are in a homeostatic and surveillant state, (2) with limited infiltration of peripheral immune cells into the central nervous system. In the pathological neuroimmune system, (3) microglia become reactive and display an altered morphology, with increase of (4) phagocytosis, (5) inflammatory markers and (6) peripheral immune cell infiltration. Image created with BioRender.com

Alzheimer’s disease

The most common type of dementia in elderly is AD, which is a non-reversible degeneration of the brain that impairs memory, cognition, personality, and other functions. The most common cause of death in patients with severe AD is aspiration pneumonia, while for the milder cases, it is myocardial infarction [11]. AD risk factors include genetic and environmental factors, such as chronic stress, diet, exercise, smoking, traumatic brain injury, diabetes, and other medical conditions [6, 8, 12]. Although the causes of this disease are not fully understood, AD has been associated with neurodegenerative elements such as amyloid plaques, fragments of amyloid-beta (Aβ) that cluster and disrupt neuronal functions. Aβ is not naturally found in the brain; its presence results from the pathological cleavage of amyloid precursor protein (APP) [13]. Despite the long-held belief in the amyloid cascade hypothesis, the association between amyloid deposits and AD remains complex and not as straightforward as initially thought. Studies suggest that short amyloid fragments can increase the membrane permeability to calcium ions and consequently disrupt the delicate balance of calcium homeostasis within the neuron [14]. Aβ accumulation was also associated with glucose metabolism and cognitive decline. In fact, Aβ tends to accumulate in brain regions with higher glucose metabolism and extensive plaque formation, leading to a gradual decline in glucose metabolism and cognitive function [15]. Aβ plaques’ accumulation in AD is associated with the apolipoprotein E (APOE) gene, particularly the APOE4 allele, which is a major risk factor for late-onset AD. Genes involved in amyloid precursor protein processing, such as APP, presenilin 1 and 2, are linked to early-onset familial AD [16]. Variants in genes related to inflammation and immune responses, like clusterin, complement receptor 1, and triggering receptor expressed on myeloid cells 2, also play a role in Aβ plaque formation [17, 18]. Additionally, neurofibrillary tangles, aggregates of hyperphosphorylated Tau protein, have been associated with AD. It was believed that Tau’s main function in physiological conditions was to stabilize microtubules in the axons of neurons. However, emerging evidence suggests that Tau’s physiological functions involve the regulation of phosphorylation-based signaling pathways [19]. When Tau undergoes hyperphosphorylation and aggregation, these functions are disrupted, resulting in a compromised axonal transport. This, in turn, exacerbates the neurodegenerative environment [19, 20]. Although the exact causes of the misshaping of Tau proteins are not fully understood, they have been linked to various factors, including a genetic variant in APOE gene [21]. Currently, there are no dependable techniques available to predict the onset of AD, resulting in individuals’ unawareness of their risk of developing the condition until symptoms manifest [22]. Discerning the manifestations of normal aging, dementia, and AD can be challenging because symptoms can overlap. During aging, the immune system weakens which is often accompanied by the prevalent occurrence of chronic low-level inflammation, referred to as inflammaging. Mild cognitive impairment (MCI) serves as a crucial stage in this spectrum, where individuals experience cognitive changes beyond normal aging but do not meet the criteria for dementia or AD. Identifying and distinguishing MCI from other cognitive conditions becomes essential for timely intervention and management to potentially slow down cognitive decline [23]. Multiple studies have demonstrated that higher levels of peripheral inflammatory markers in mid- to late-life are associated with an increased risk of dementia and cognitive decline. These events can have complex effects on the neuroinflammatory response in AD, potentially leading to immunosuppressive or anti-inflammatory environments within the brain. As described in depth in Sect. 2.2.1, peripheral inflammation and dysregulation of the immune system in the CNS contribute to the pathogenesis of AD [24, 25].

Parkinson’s disease

PD is a neurological disease, mostly recognized as a movement-related disorder, with deep grey matter degeneration caused by neuronal loss in the substantia nigra, resulting in a lack of dopamine in the basal ganglia [6,7,8, 26]. Along with non-motor symptoms such as insomnia, constipation, dementia, and cognitive and olfactory impairments, the well-described motor symptoms include bradykinesia, stiffness, resting tremors, and postural instability [6,7,8, 12, 27]. PD’s development and association with the death of dopaminergic neurons in the substantia nigra remain unclear [28]. Evidence suggests a link to Lewy bodies, abnormal protein deposits primarily composed of aggregated α-synuclein (α-syn). α-syn, a protein normally present in presynaptic terminals, regulates neurotransmitter release [29, 30]. In neurodegenerative diseases, the imbalance between the production and clearance of α-syn can lead to its misfolding and accumulation into Lewy bodies [31, 32]. Several genetic variants have been associated with α-syn aggregation and increased risk of PD. For instance, mutations in the α-syn gene (SCNA) can lead to abnormal protein folding and aggregation. Additionally, variations in genes involved in α-syn metabolism and clearance, such as leucine-rich repeat kinase 2 gene (LRRK2), glucocerebrosidase gene (GBA), and parkin RBR E3 ubiquitin-protein ligase gene (PRKN), have also been implicated in PD pathogenesis [33]. A recent study examined the impact of Lewy bodies on neurons in the nucleus basalis of Meynert. In patients displaying PD symptoms, the presence of Lewy bodies was associated with mitochondrial damage and neuronal loss. Moreover, Lewy bodies may also be involved in sequestering damaged mitochondria and/or α-syn oligomers, providing protection against their harmful effects [34]. The α-syn aggregates in the brain interfere with the normal functioning of dopaminergic neurons, thus leading to motor impairments, accompanied by depression, anxiety, and apathy [35, 36]. This is attributed to the spread of α-syn pathology to regions beyond the substantia nigra, including cortical areas involved in cognition, mood regulation, and emotional processing [36]. Efforts to predict PD have led to the exploration of potential biomarkers associated with key molecules involved in the disease. Lin et al. investigated the levels of α-syn, Aβ-42, Tau, and nuclear DNA in peripheral plasma, along with regional gray matter volumes, in patients with idiopathic PD. The findings revealed three distinct patterns of gray matter volume atrophy corresponding to different stages of PD. Additionally, the study unveiled correlations between proteinopathy, inflammation markers, gray matter volume patterns, and cognitive performance in PD [37]. While PD can occur at any age, the prevalence of the disease rises significantly in elderly people. The natural decline in the function of the dopaminergic system, accompanied by age-related processes, such as accumulation of oxidative stress, impaired mitochondrial function, altered protein handling mechanisms, as well as altered immune function and neuroinflammation have been implicated in the pathogenesis of PD [38].

Immune system associated with Alzheimer’s and Parkinson’s diseases

In the CNS, components from the innate and adaptive immune system can be found. Microglia are the resident innate immune cells, which originate from the embryonic yolk-sac and colonize the brain in early development. These cells play diverse roles in maintaining the function, plasticity, and integrity of the CNS. Additionally, microglia are actively involved in maintaining homeostasis of the CNS upon various challenges. They contribute significantly to neurogenesis, synapse formation, modification and pruning, vasculature maintenance and repair, axonal myelination, and other essential activities that support CNS activity and health [39, 40]. However, these cells can become reactive, releasing excessive amounts of pro-inflammatory and toxic molecules and perform aberrant phagocytosis that cause synaptic loss and neurodegeneration when exposed to chronic challenges. Over the course of aging, microglia can also become dystrophic and senescent, displaying altered morphology, transcriptome, proteome, lipidome and consequently, impaired functions [41]. With age-associated changes, microglia become less efficient at surveying the brain parenchyma, remodeling synapses, and clearing away damaged cells and toxic debris [42]. Other myeloid cells include monocytes and macrophages, specialized innate immune cells responsible for phagocytosis, release of cytokines and T cells antigen-presentation [43]. In addition, adaptive immunity is the ability to withstand infection and tissue damage caused by specific antigens after previous exposure. This protection comes from adaptive responses from lymphocytes, a bone-marrow lineage derived from hematopoietic stem cells, which are found in the bloodstream. Multiple subtypes of lymphocytes collaborate to coordinate adaptive immune response. For instance, the effector T cells (Teffs), particularly T helper (Th, CD4 +) cells, recognize foreign antigens and coordinate their response with other Teffs, including cytotoxic T cells (CD8 +), regulatory T cells (Tregs) and B cells [25]. CD8+ cells are primarily responsible for recognizing and eliminating target cells, through the release of cytotoxic molecules, such as perforin and granzymes [44]. By contrast, Tregs are specialized subsets of CD4+ T cells that have immunosuppressive functions to avoid self-antigens response [45]. Lastly, B cells, also bone-marrow derived, are a lymphocyte type specialized in antibody production and antigen presentation, to neutralize foreign pathogens and resist infections [46].

‘Neuroinflammation’ is a complex phenomenon referring to the inflammatory response of the CNS to various insults, such as injury, infection, or neurodegeneration [47, 48]. While inflammation is a normal protective response of the immune system, chronic or excessive inflammation can have detrimental effects on the CNS [47]. CNS exacerbated inflammation is implicated in various neurological disorders, including AD and PD.

Neuroinflammation and immune dysfunction in Alzheimer’s disease

In AD, CNS over-inflammation is marked by numerous factors that work together to drive the disease progression. Dysregulated levels of various anti- versus pro-inflammatory cytokines, the phenotypic transformation of immune cells such as microglia in the CNS, and the recruitment of macrophages and lymphocytes from the periphery, contribute to synaptic loss, which is the most accepted pathological correlative of the subsequent cognitive impairment [47].

Among the pathological mechanisms driving AD, the elevated pro-inflammatory cytokine production during healthy middle-age, taking place outside the CNS by endothelial cells and adipocytes upon injury, or inside the CNS via neurons, astrocytes and other immune cells upon inflammation or infection, can be associated with an increased risk of developing dementia or AD later in life. Elevated levels of interleukin (IL)-6 and -1β, as well as tumor necrosis factor alpha (TNF-α) were found in elderly individuals’ blood samples who had a first-degree relative with dementia. High blood cytokine levels were also associated with a greater risk of developing mild cognitive impairment (MCI). This indicates that pro-inflammatory cytokines may contribute at a preclinical stage to the brain chronic inflammation found in AD cases [49, 50]. Studies on the effect of cytokines on AD progression can be found in Table 1.

Table 1 Effects of innate and adaptive immune cells on Alzheimer’s disease progression

The microglial impact on AD pathology is complex and multifaceted [51]. During disease progression, physiological roles of microglia in surveillance, response to damage, and phagocytosis become impaired [52]. Microglia also contribute negatively to the spread of Aβ and Tau pathology. The mechanisms driving these different roles of microglia in AD pathology remain largely unclear, with microglial metabolic alterations being the main accepted hypothesis to explain these events [53]. A decreased microglial responsiveness to cues, and a reduced performance in their essential functions, such as surveillance, trophic support, and phagocytosis, are associated with the microglial changes present in the AD pathology [54]. Interestingly, a microglial state named “dark microglia” was recently found in mice and humans and is predominantly observed during aging and AD pathology. These cells exhibit a condensed and electron-dense cytoplasm and nucleoplasm, along with markers of oxidative stress, which classifies them as “dark” cells [55]. These cells portrait a hyper-ramified morphology, are immunoreactive for CD11b and triggering receptor expressed on myeloid cells 2 (TREM2), and weakly immunoreactive for conventional microglial markers, like P2Y12 and IBA1. These cells further associate with Aβ plaques and surround axon terminals and dendritic spines, and they have been found in many brain regions, including the hippocampus [55, 56]. Details of the latest studies on microglia can be found in Table 1.

As “inflammaging” confers an important risk of developing AD, the progressive release of pro-inflammatory mediators by monocytes and macrophages during aging is also associated with AD onset and progression [57]. When comparing AD patients with healthy patients, circulating monocytes from AD patients were found to produce significantly higher levels of free radicals compared to those of healthy subjects [58]. Inflammatory or chemoattractant cascades are also altered in AD. For instance, the monocyte chemoattractant protein-1 (MCP-1), during chronic inflammation, contributes to an overactivity and excessive recruitment of inflammatory cells, which leads to neuronal damage and cognitive decline [59]. Inflammation and aging are determining factors that affect innate and adaptive immune responses. Several studies demonstrated altered distributions and phenotypic transformation of T helper and cytotoxic T cells in the periphery, resulting in complex implications for AD progression [60]. Details of the latest studies on microglia cells, peripheral monocytes, macrophages, and T cells can be found in Table 1.

Neuroinflammation and immune dysfunction in Parkinson’s disease

Both peripheral and central inflammation have been widely associated with PD pathogenesis and progression. Key natural history studies have revealed associations of inflammation and immune cell activation with disease progression, particularly with risk of differential prognoses, such as higher risk of cognitive impairment and dementia and lower risk of motor dysfunction [68,69,70]. Genetic linkage studies and genome-wide association studies have linked PD risk with mutations in immune-related genes, such as HLA-DR [71,72,73]. Furthermore, PD shows pathological similarities to autoimmune diseases such as inflammatory bowel disease and genetic variants have been found to be shared between PD and other autoimmune conditions [74, 75]. In healthy individuals and early in the PD disease process, microglia are considered beneficial by clearing protein aggregates, releasing neurotropic factors, and performing synaptic pruning essential for learning and memory. However, microglia in the substantia nigra of PD patients were found to be dystrophic, releasing higher levels of pro-inflammatory cytokines IL-1β, IL-6, IL-12, TNF-α, s and inflammatory molecules such as ROS and nitric oxide [76,77,78]. These pro-inflammatory factors can have direct neurotoxic effects on the dopaminergic neurons (DAN); chronic exposure of neurons to these factors can lead to degeneration [79, 80]. Among other mechanisms of glial modulation, both microglia and astrocytes can be directly activated by α-syn, which is recognized by Toll-like receptor (TLR)-2 and TLR-4 on astrocytes and microglia. Upon recognition of α-syn, glia upregulates pro-inflammatory downstream pathways and secrete pro-inflammatory cytokines. In astrocytes, increased inflammasome activity is particularly notable [81]. In the periphery, both innate and adaptive immune cells are affected in PD; innate monocytes, macrophages and neutrophils secrete higher levels of pro-inflammatory cytokines. Like microglia, circulating monocytes and macrophages express higher levels of TLR-2 and TREM2. TREM2 stimulates microglial phagocytosis, maintains metabolic fitness, and suppresses the pro-inflammatory cytokine production. Adaptive immune dysfunction is likely to contribute to earlier symptoms of PD that occur before motor onset [82, 83]. T cells reactive to α-syn can be detected 10 years before motor diagnosis as well as later in the disease process [84]. In addition to their reactivity to α-syn, T cell numbers in PD patients were found to be widely dysregulated; CD3 + cells as a proportion of total leukocytes are lower, but there is a skew towards pro-inflammatory Th1 and Th7 subsets, with lower Th2 and T-reg cells. Concurrently, T cells are more primed for activation and release higher levels of pro-inflammatory cytokines, reflected by increased TNF-α, IL-1β, IL-2 and IL-10 in the serum of PD cases [85,86,87,88,89]. B cells are also implicated in PD pathogenesis, though their involvement is less well elucidated than the T cells. Higher autoreactive α-syn antibodies are detected in PD blood [90] and like T cells, the total B cell number is lower and there is a skew towards pro-inflammatory B cells. There is evidence of increased TNF-α and IL-6 production, higher ratio of IgG to other Ig molecules, indicative of antibody class switching and activated humoral response. Furthermore, B cells in PD blood express higher levels of antigen presentation molecules, suggesting increased support of T cell activation [91, 92]. In healthy individuals, the central and peripheral immune systems are structurally distinct, thanks to the presence of the highly specialized blood–brain barrier (BBB). In PD, the BBB is known to be leaky, allowing for a less-regulated passage of the peripheral immune cells into the brain parenchyma. When peripheral immune cells infiltrate, they can induce neurotoxicity and phenotypically transform microglia and astrocytes, worsening neuroinflammation [93].

Current treatments and clinical trials for Alzheimer’s disease and Parkinson’s disease

One of the common drug targets in AD is acetylcholinesterase, an enzyme that is more active in patients with AD, leading to a lack of acetylcholine in the brain and hence, increased Aβ plaque production (Table 2). Lately, different research groups have focused on the development of drugs able to reduce the Aβ load in AD, primarily using monoclonal antibodies [94]. Among these, lecanemab was approved in January 2023, and aducanumab was granted conditional accelerated approval by the FDA in 2021. The drug will undergo phase 4 clinical trial, which will end in 2030 [95]. Details about these drugs can be found in Table 2.

Table 2 Drugs treating Alzheimer’s and Parkinson’s disease that have passed phase 3 clinical trials

Numerous clinical trials for new treatments against AD are currently active on clinicaltrial.gov. Different research groups are testing drugs, devices, and behaviors (physical activity or different diets) to decelerate the disease or treat individual symptoms (e.g., depression, agitation, psychosis, or epilepsy). Their focus is on drugs to improve Tau protein PET imaging, brain stimulation devices, diet supplements, and physical activity to improve patients’ cognition. Most drugs aim to prevent neurodegeneration and cognitive decline [96,97,98,99,100,101,102,103]. Many groups are investigating new drugs targeting Aβ and Tau. Details on their mechanism of action and phase are listed in Table 3.

Table 3 Drugs against neuroinflammation currently tested in clinical trial found on ClinicalTrials.gov [103] and the ISRCTN registry

For PD, there is a wide variety of drugs available to treat the motor symptoms of the disease, at least in the short term. The majority of treatments against PD target the dopamine system, including dopamine agonists and dopamine breakdown inhibitors (Table 2). Common dopamine breakdown inhibitors target the enzymatic activity of monoamine oxidases, to prevent dopamine breakdown in the system. A non-pharmacological treatment for PD is stereotactic deep-brain stimulation, which uses high frequency stimulation to normalize neuronal firing rates, targeting tremor, stiffness, and bradykinesias [104]. It also has beneficial effects on PD cognitive symptoms [105]. New PD treatments are being developed, including α-syn targeting therapies, neurotropic factors, cell, and gene therapies [106], as well as therapies that target the immune activation in PD. Often, immunomodulation offers a relatively simpler treatment option, utilizing preapproved drugs with known safety and tolerability profiles. Current and prospective immunomodulatory therapies can be categorized into three main groups, broad spectrum immunosuppression, specific immune pathway targeting and microglial targeting (Table 3).

In vitro and preclinical studies for Alzheimer’s and Parkinson’s diseases and the immune system

In vitro and in vivo models can be used to mimic the neuroimmune systems. Even though animal models provide both physiological and behavioral processes, they do not always yield results that can be translated in preclinical drug screening for humans. Cells in 2D cultures are grown on flat surfaces that have been optimized for cell attachment and growth, whereas cells in 3D cell cultures are generally embedded inside a gel-like matrix or grown on a solid scaffold. 2D models do not mimic human brain complexity, creating a need for more physiologically relevant 3D culture formats [6,7,8, 12, 26, 27]. The advantages and disadvantages of both approaches are summarized in Fig. 3.

Fig. 3
figure 3

Advantages and disadvantages of in vivo and in vitro model of the neuroimmune system. The schematic shows the advantages and disadvantages of using animal models compared to cellular models, in 2D or 3D, and organ-on-a-chip. Image created with BioRender.com

In vitro models of the immune system

Numerous studies investigating the immune system in the context of cancer have been published research [127, 128]. The development of effective 3D models has significant implications for disease modeling and drug screening, to develop new chemotherapies or artificial tissues for transplantation [129]. T cell immunotherapy has emerged as a promising approach for treating cancer, infections, and autoimmune diseases. Jin et al. explored the latest advancements in 3D bioprinting and T cell engineering and their medical applications [130]. Among the benefits of using bioengineering approaches, other therapy applications include boosting T cell growth and differentiation into central memory-like and effector memory subsets for therapy applications. Gray et al. [131] and Snow et al. [132] discussed the potential applications of 3D bioprinting and T cell engineering in the field of neurosurgery, including creating immune chips to elucidate links between immune response and disease. Recently, 3D bioprinting has shown great potential in advancing immunotherapy, particularly in the context of natural killer (NK) cells, a type of immune cell that possess the ability to recognize and eliminate target without prior sensitization. The encapsulation of NK cells provides protection and support, allowing for their controlled release and sustained activity at the tumor site. This targeted delivery system enhances the specificity and efficacy of NK cell-based immunotherapy, minimizing off-target effects and maximizing tumor cell killing [133]. CAR-T cell therapy involves genetically modifying a patient’s T cells to express chimeric antigen receptors (CARs) that can recognize and attack cancer cells [134]. CAR-T cells cultured on a 3D scaffold have higher proliferation rate and cytokine production compared to 2D cultures. Moreover, CAR-T cells cultured on a 3D scaffold can improve tumor infiltration and antitumor activity in vivo [130]. Compared to CAR-T cells, CAR-NK cells have several advantages, including their innate ability to recognize and eliminate tumor cells without prior sensitization, lower risk of graft-versus-host disease, and potential for off-the-shelf use due to their allogeneic nature. As for CAR-T cells, 3D scaffolds can improve the survival, proliferation, and infiltration of CAR-NK cells into tumor tissues, leading to enhanced therapeutic outcomes [135]. Researchers also focused on the behavior of macrophages when cultured in 3D [136,137,138]. By precisely manipulating scaffold properties, such as pore size, surface topography, and mechanical cues, researchers can investigate the impact of these factors on macrophage behavior and function. Compared to 2D culture, macrophages in 3D express more cell-specific markers, even if the cells decreased their number, highlighting the influence of the scaffold microenvironment on macrophage behavior [136,137,138]. Bioprinted 3D scaffolds containing macrophages and antibiotics have also been used to reduce Staphylococcus aureus bacterial burdens during infections [139, 140]. This knowledge can be employed to develop innovative strategies for tissue regeneration, immune modulation, and the design of biomaterials that promote favorable immune interactions [141].

Two-dimensional vs three-dimensional models of Alzheimer’s and Parkinson’s disease

Modeling the CNS and neurodegenerative disease in vitro can be challenging due to the complexity of the CNS, the difficulty in accessing the human brain and living primary neurons. Even if post-mortem tissues are often used, these samples represent only the final stage of the pathology and fail modeling the disease progression [6, 142, 143]. An alternative to primary cells are immortalized cell lines. The most commonly used cell line for CNS modeling is the human neuroblastoma cell line SH-SY5Y, which mimics immature catecholaminergic neurons as they express immature neuronal markers and have neurite morphology [144]. Cells can then be exposed to neurotoxins to promote the pathogenesis of AD or PD, or they can be genetically modified to overexpress APP or Tau to mimic AD [145], or α-syn to mimic PD ([146]. SH-SY5Y can also be differentiated into specific neuronal phenotypes. SH-SY5Y have some disadvantages, such as the absence of an established procedure for maintaining them in culture, which results in inconsistent experimental results and variable cell growth [6, 146, 147]. Moreover, SH-SY5Y do not exhibit electrophysiological and electrochemical features normally present in mature neurons [148]. The use of induced pluripotent stem cells (iPSC) to create reproducible models mimicking the human pathophysiology has become a rapidly developing area of research [7, 149, 150]. Somatic cells from AD and PD patients are reprogrammed followed by neuroinduction. In this case, a genetically accurate model of AD and PD is developed [6]. If the somatic cells are isolated from healthy patients, AD and PD models can be generated treating the iPSC-derived neurons in the same way as the immortalized cells [151].

Alzheimer’s disease

There have been several studies using 2D models created from iPSCs derived from AD patients [6, 12, 152]. The formation of abnormally phosphorylated Tau protein, increased expression of glycogen synthase kinase-3, the protein kinase that phosphorylates Tau; and upregulation of genes associated with the oxidative stress response were all achieved in a study using 2D cultures of iPSCs derived from an 82-year-old AD patient [153]. Most significantly, these 2D neuronal cultures lacked glial cell support functions, which are crucial to driving AD pathogenesis [6, 8, 12, 152]. In contrary, self-assembling peptide hydrogels seeded with neuroepithelial stem cells from hiPSCs were used in 3D studies to demonstrate the capability to simulate AD’s in vivo-like responses, such as aberrant translocation of activated P21-activated kinase and redistribution of the actin stabilizing protein drebrin, which were not observed in 2D models. The cytoskeleton dynamics depend on P21-activated kinase and drebrin, and the former is necessary for mechanotransduction pathways and AD pathology [154]. Moreover, in AD 2D models, changing the culture medium regularly can remove the Aβ secreted into the cell culture media, thus interfering with, and affecting the analysis of Aβ aggregation. 3D systems might better mimic the environment of human brain, allowing Aβ deposition and aggregation by limiting the diffusion of secreted Aβ into the cell culture medium and enabling the formation of niches that accumulate high concentrations of Aβ [6, 8, 12, 155, 156]. Additionally, human iPSCs-derived organoids can be engineered to mimic the organizational features of the human brain, e.g., the cerebral cortex to model AD. These brain organoids facilitate the pathological transfer of toxic misfolded proteins, all of which contribute to a higher translational value of brain organoids compared to that of 2D cell models. It was shown that the toxicity of Aβ oligomers was greater in 3D compared to 2D cellular models of AD [6, 8, 12]. This highlights the importance of developing complex 3D in vitro models for specific brain regions, their interactions, and local microenvironments in AD [12, 154, 155, 157]. Organoids can also be used for drug screening [158]. Lee et al., for example, isolated iPSCs from patient blood and differentiated them into neurons and astrocytes to create a 3D human AD neuro-spheroid model. This 3D model physiologically mimicked in vivo response to the drugs tested [157]. Neuronal precursors (NPCs) can also be used to resemble AD. Lomoio et al. recently developed a silk-fibroin/collagen hydrogel for the culture of patient-derived NPCs organoids with familial AD APP London mutation. The cells were cultured for 4.5 months, showing high expression of extracellular Aβ42/40 ratio deposition, and increased neuronal excitability. Moreover, cells showed a decrease in the expression of genes related to transmitter release, neural connectivity and vesicle trafficking, all established neurodegeneration pathways [159]. Kwak et al. embedded NPCs in Matrigel™ and cultured them for 6 weeks. This 3D model confirmed that Aβ42/40 can affect Tau accumulation in neuronal cells [160].

Parkinson’s disease

Many researchers have developed iPSC models specific to PD by reprogramming somatic cells into dopamine neurons, astrocytes, and microglia from PD patients. Complex networks of dopaminergic neurons have been developed through the use of iPSCs and organoid culture. In general, iPSC-derived neurons and organoids from familial and idiopathic PD patients replicate the main pathological disease phenotypes and are useful research tools for understanding the disease’s molecular mechanisms. Additionally, they bridge the gap between human and animal models for target validation. These 3D systems are essential because they generate intricate structures that are impossible to replicate in 2D models [7, 8, 26, 161]. In 2D models, Cooper et al. successfully produced dopaminergic neurons from human iPSCs and, after transplanting the cells into rodent brains, the treated rats showed good survival rates and improved behavior [149, 150]. The 3D model proposed by Moreno et al. used Matrigel™ and phase-guided microfluidics bioreactors differentiated human iPSCs into dopaminergic neurons. Action potential propagation along neurites and other spontaneous electrophysiological activity were reported. Accordingly, their model is reliable, economical, and exhibits biological fidelity for future use in PD modeling and drug discovery [162]. Dopaminergic neurons (DAN) derived from iPSCs were cultured in silk-based 3D scaffolds. Compared with cells in 2D, cells cultured in the scaffolds showed a dense neuronal network, exhibited high levels of α-syn and an altered purine metabolite profile [163]. Abdelrahaman et al. developed a scaffold made of self-assembled tetrapeptide. In this scaffold, DAN derived from human embryonic stem cells (hESC) responded to neurotoxins responsible for DAN loss and showed spontaneous activity over a month in culture [164]. A 3D printed scaffold made with polylactic acid (PLA), chitosan and Levodopa (a drug used to treat PD—Sect. 3) was fabricated as a potential drug delivery system. The scaffold biocompatibility was tested with human adipose-derived mesenchymal stem cells, that were alive and metabolically active over 7 days in culture. Moreover, the scaffold released the drug in PBS for 14 days, making it a suitable candidate for drug delivery [165].

3D models to study the crosstalk between the immune system and nervous system

Cellular communication among neighboring cells stands as an indispensable prerequisite for the preservation of tissue functions and the attainment of homeostatic equilibrium. Despite the advancements in the development of in vitro models (Sect. 4.2), modeling the interfaces between central and peripheral cells remains a challenge [6]. Interactions between neighboring cells serves as a foundational mechanism governing a multitude of critical biological processes, encompassing essential phenomena such as cell signaling, proliferation, and differentiation. Hence, crosstalk between neurons and macrophages plays a pivotal role in modulating macrophage function across diverse physiological perspectives. However, understanding of the exact mechanisms governing the equilibrium between pro-inflammatory and pro-repair responses remains inadequately elucidated. Moreover, microglia, serving as the innate immune cells of the brain, emerge as an exceedingly attractive focus for the investigation of potentially enhanced therapeutic approaches.

Traditionally, in vitro models of AD lack the ability to accurately emulate the intricate patient-specific characteristics exhibited by microglia in a 2D framework. Current in vitro models rely on the direct co-culture of neurons or glia with immune cells, lacking the structural and spatial elements present in vivo. It is therefore challenging to translate drug testing results into clinical applications due to a lack of clinical relevance or physiological intricacy in 2D neuroimmune models.

Recently developed novel 3D in vitro models of monocyte-derived microglia-like cells (MDMi) from AD patients aim to overcome these shortcomings. To simulate the neuro-glial signals present in the brain microenvironment, a co-culture platform was established with MDMi and NPC. Compared to 2D, MDMi in 3D exhibited higher survival and increased microglial marker expression, as well as mature microglial characteristics, such as highly branched morphology. Moreover, changes in cell-to-cell communication, growth factor and cytokine secretions, and amyloid-responses were seen in 3D co-cultures with neuro-glial cells [166]. Studies on the interaction between microglia and neural cells involve organoids derived from human iPSCs. In 2022, Sabate-Soler et al. fabricated midbrain organoids with dopaminergic neuroprogenitors deriving from hiPSCs. The differentiation from iPSC to neuronal progenitors was performed in 15 days, then hiPSCs-derived microglia cells were integrated into the organoids (assembloids) and cultured for 20 or 70 days. Therefore, a specific cell culture medium was optimized to allow the growth of both cell types, containing N2 supplement, 2-mercaptoethanol, IL-34, GM-CSF, BDNF, GDNF and DAPT; while TGFβ3, cAMP, and Activin were excluded as they proved toxic for microglia. After 70 days in culture, each cell type maintained its phenotype and expressed cell-specific markers. The phagocytic activity of microglia was enhanced in the assembloids, and with the cells’ removal, the assembloids size was smaller compared to organoid with neural cells only. Microglia in the assembloids also reduced the oxidative stress and the gene expression of synapsis marker, making them a good model of neuroinflammation in PD [167]. Brüll et al. implemented hiPSCs-derived astrocytes in neural organoids made of human dopaminergic neurons. The addition of astrocytes promoted the formation of a postmitotic organoid that could be used for neurotoxic and pathophysiologic studies [168]. Xu et al. differentiated hiPSCs into macrophage progenitors and NPCs to form organoids containing both cell types. Cells were then matured into macrophages and neurons. Mature organoids showed phagocytosis of apoptotic cells and reduced synapsis formation [169]. Song et al. developed organoids of neural cells from the dorsal or ventral cortex and integrated with hiPSCs-derived microglia-like cells to create a model to study the role of microglia in different neurological disorders [170]. Cai et al. developed an acoustic assembly device that can be used to form neural spheroids. Here, they formed spheroids with mice primary neurons and cultured with Aβ aggregates, showing cell death and neurotoxicity, as it happens in vivo. When microglia cells were added to the neurons and the Aβ, microglia cells migrated and surrounded the Aβ, and their activity was higher compared to organoids without Aβ, increasing the neurotoxicity [171]. Additional structural elements have been included to further enhance 3D cultures; studies have also been performed in cells cultured in 3D hydrogel scaffolds. Raimondi et al. compared semi-interpenetrating polymer networks made of type I collagen (COL) and hyaluronic acid (HA) or poly (ethylene glycol) (PEG). Cortical neurons, astrocytes and microglia from mice were co-cultured in the hydrogels for 21 days. PEG-based hydrogels led to enhanced cell metabolic activity. Furthermore, a complex network between glial cells and neurons formed. PEG-based gels also promoted synapses formation, with better results shown when cells were co-cultured in Col-PEG2000 cells [172]. Chemmarappally et al. developed a polyacrylonitrile/Jeffamine® doped polyacrylonitrile-based fibrous scaffold and co-cultured mature neurons from neuroblastoma cell line SH-SY5Y and astrocytes from glioblastoma cell line U-87MG. After exposing the cells to PD mimicking inhibitors cells showed a higher survival rate compared to cells cultured individually on the scaffolds [173].

These studies successfully incorporated the brain-resident immune cells. It would be interesting to study the paracrine interaction between the immune system and CNS in vitro, particularly given the negative effect that cytokines can have on AD and PD progression (Sect. 2.2). Recently, to assess the role of inflammation in PD progression, Rueda-Gensini et al. co-cultured 3D bioprinted DAN in an electroconductive extracellular matrix-derived scaffold with graphene oxide, with human astrocytes and monocyte-derived macrophages, in a trans-well system. Inflammation was promoted by exposing all the cell types to an aberrant form of α-syn. DAN showed mitochondrial and synaptic dysfunctions, oxidative stress and α-syn accumulation. An increase in TNF-α, IFN-α2, Monocyte Chemoattractant Protein-1 (MCP1), IL-12p70, IL-8, IL-18 and IL-23 levels, all pro-inflammatory cytokines, was observed. The level of pro-inflammatory cytokines was higher when the immune cells were co-cultured with DA in comparison to monoculture [174].

In vivo models for Alzheimer’s disease and Parkinson’s disease

Invertebrate and vertebrate animals have been widely used to study neurodegenerative diseases. Even if in vivo model cannot exactly replicate the complexity of the CNS, different species have been utilized to investigate single biological, molecular and pathological aspects of neurodegenerative diseases that can be translated to humans [175]. As mentioned in Sect. 4.2, studying the human brain is challenging which limits the study of neurodegenerative diseases and their progression. The use of animal models provides more flexibility and reduces these problems [176].

Invertebrate models

The most common invertebrate models used to study neurodegenerative diseases are the fruit fly Drosophila melanogaster (Drosophila) and the nematode worm Caenorhabditis elegans (C. elegans), because they have molecular features similar to humans. As their genome has been fully sequenced, they can easily be genetically modified. This aspect is crucial in AD modeling, as in both Drosophila and C. elegance Aβ is absent, even if APP, PSEN1, and BACE1 are expressed [175]. Thus, transgenic models of both species were developed [177, 178]. Moreover, Drosophila possesses forms of innate [179] and adaptive immunity [180], making it a suitable model for studying neuroimmune dynamics in AD and PD [181]. On the contrary, C. elegance lacks inflammatory cells, microglia, and astrocytes, meaning that only aspects unrelated to neuroinflammation can be investigated using this model [182, 183]. In Drosophila AD models, Aβ are surrounded by glia positive to Draper, the glia engulfment receptor, which activates the signaling pathway of Aβ phagocytose. When Draper is not expressed, locomotion problem and short life span occur [184]. By mutating the transmembrane of a homolog of the mammalian IL-1 receptor (Drosophila Toll gene), a decrease of the Aβ42 neuropathological activity takes place in Drosophila, while the gain of function of the toll receptor increases the Aβ42 neurotoxicity activity. Thus, by suppressing the Tl-NF-κβ pathway, the Aβ42’s neuropathological activity decreases [185]. Furthermore, it was shown in Drosophila models that the increase of ROS can lead to tauopathies progression [186,187,188,189]. Pathogenic Tau and Aβ42 also have a negative effect on behavior, the regulation of neuroinflammatory genes, and the neurodegeneration in Drosophila. It is interesting to note that tauopathy models showed more severe neurological impairment than amyloidopathy models [190]. Drosophila is also used to replicate PD. Mutants of PINK1/Parkin show impaired brain and muscle mitochondrial activity, malfunctioning flight muscle, elevated levels of oxidative stress, male sterility, low life expectancy, decreased motor function, and aberrant DAN morphology [191, 192]. Olsen et al. developed an α-synucleinopathy model to investigate the role of several genes involved in glial modifications in PD, including the ortholog of LRRK2. When these genes were suppressed, neurodegeneration, neural loss and α-syn oligomerization occurred [193].

Vertebrate models

A great variety of vertebrates have been used to model the AD pathology, with rodents being the most common choice for transgenic AD models and non-human primates, such as macaques, for the non-transgenic modeling of the disease [194]. Mouse models are widely used to study AD pathology, due to their ability to be genetically manipulated, mimicking the disease hallmarks and organism’s response to the pathology [195]. Transgenic models involve knock in/out of mutated genes associated with AD pathology, such APP, APOE, α-secretase and presenilins that result in an effective and early onset of Aβ accumulation or Tau hyperphosphorylation. These modifications reproduce many aspects of the pathology, such as neuronal loss [196]; synaptic dysfunction [197] and cognition impairment [198] among others. Even though transgenic mouse models are good at representing the Aβ burden, sporadic forms of the disease are considered more representative of the human condition [199]. This can be achieved using either non-transgenic mouse models—by administrating soluble Aβ oligomers or with novel methods comprising environmental challenges (e.g., chronic inflammation)—or by studying genetic variants that offer greater risk of developing AD. As stated in Sect. 2.2.1, although Aβ plaques are the central cause of the synapse loss and cognitive deficit in AD, chronic inflammation has also been implicated in the onset and progression of these AD-related pathologies [200, 201]. Novel techniques were designed to understand the role of exacerbated inflammation, by using a viral mimetic polyinosinic:polycytidylic acid (poly I:C). The poly I:C model is known to achieve an increased Aβ deposition and cognitive deficits in a non-transgenic mouse [200, 201]. Additionally, several genetic variants of TREM2 (R47H and R62H) have been associated with an increased risk of developing sporadic AD and are being studied in the AD context [202]. Despite the wide usage of the animal models, great discrepancies are observed. Most human cases occur sporadically, while most transgenic mouse models have an early onset [194]. In addition, the pro-inflammatory signaling cascades and phenotypic characteristics of immune cells in AD pathology in transgenic mice, differ from those seen in the human brain. For instance, rodent microglia tend to have a more robust and rapid pro-inflammatory response compared to human microglia. This difference can be attributed to the expression levels and functional variations of specific receptors and signaling pathways [203, 204]. These inter-species variations emphasize the importance of utilizing human models for studying and developing therapeutic interventions for AD [199].

PD does not occur naturally in any mammals except humans, so modeling the disease is limited to focusing on a single or small number of symptoms. Only 7% of PD is caused by genetic mutation [35]. Modeling the disease subtypes is more achievable, though full penetrance of the disease is also likely due to several genetic contributors, as well as environmental factors. Modeling the non-inherited forms of the disease is more complicated, and often one aspect of the disease is studied in isolation. Animal models of PD fall into three main categories, neurotoxin models, genetic/transgenic models, and protein models. Neurotoxin models involve intracerebral lesioning with neurotoxins such as 6-hydroxydopamine (6-OHDA), MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; a prodrug of neurotoxin MPP), rotenone or paraquat. These neurotoxins can induce α-syn aggregation in the brain, as well as causing direct neurotoxicity killing DAN in the lesioned area. Interestingly, neurotoxin models are capable of inducing neuro- and peripheral infiltration [205]. A more direct in vivo model of neuroinflammation is achieved by administrating lipopolysaccharide (LPS) into the brain inducing neuroinflammation. Interestingly, LPS models also show selective DAN loss, replicating PD phenotypes and suggesting a causative role of neuroinflammation in DAN loss [206]. Genetic models of PD can replicate familial forms of the disease with known mutations in PD genes such as PARK7, LRRK2, PRKN, PINK1 or SCNA. Furthermore, transgenic models can be generated to examine genes of interest thought to be involved in PD pathogenesis. Like in AD models, protein aggregation models can be generated, in PD using recombinant α-syn pre-formed fibrils based upon the prion-like transport models [207]. In vivo models allow behavioral testing to be performed. Common behavioral examinations of PD include amphetamine-induced rotation (motor function), rotarod and pole tests (coordination), gait examinations, forced swim and open field tests (anxiety, depression, and mood symptoms). Utilizing these models allows for therapeutic testing in which behavioral rescue can be used as a readout. When considering PD systemic nature, and involvement of the gut and peripheral immune system, in vivo models can offer a more complete picture of the disease than in vitro models.

Organ-on-a-chip of neuroimmune system in Alzheimer’s disease and Parkinson’s disease

The emergence of organ-on-a-chip platforms has allowed for enhanced design capacities and control in building in vitro models capable of mimicking biological, biochemical, physiological, and mechanical phenomenon in living organ systems. Details on materials used for chip fabrication [208,209,210], hydrogels incorporated in the chips [211], and the fabrications techniques used are reviewed elsewhere [194, 212,213,214,215,216,217,218,219]. A schematic of the organ-on-a-chip development can be found in Fig. 4.

Fig. 4
figure 4

Organ-on-a-chip development: the schematic shows the steps required for developing and fabricating a microfluidic chip. Image created with BioRender.com

While some studies primarily focus on successful neuronal cell culture [220], there are a growing number of studies that focus on modeling more complex physiology of permeable barriers relative to the brain–immune axis. For example, a study conducted by Zhou et al. designed a blood–cerebrospinal fluid barrier microfluidic model that allowed two parallel and opposed microchannels separated by a polyester porous membrane to be continuously perfused. This allowed for the monitoring of mass transport and diffusion of molecules from each channel paving the way for further analysis into essential neuroimmune interactions within the blood–cerebrospinal barrier [221]. Similarly, a study published by Brown et al. employed a two-chamber system divided by a polycarbonate membrane made from polydimethylsiloxane (PDMS) and seeded different cell types in opposition to form their neurovascular blood brain barrier unit [222]. Salmon et al. fabricated a chip capable of co-culturing hiPSCs-derived pericytes and endothelial cells that self-assemble into vascularized networks in a temporal/spatial manner [223]. Recently, Kang et al. developed a brain-on-a-chip model, co-culturing human neurons, astrocytes, and microglia. This device was used to develop models of CNS disorders, bacterial infections, cancer, or edema, by introducing Aβ (adding cells expressing APP variants), bacterial conditioned media or bacterial toxin, cancer cells, or ammonium chloride, respectively. The device allowed for both real-time and end-point analysis [224]. Microfluidic platforms have also been used to understand the neuroinflammatory processes in AD. Park et al. developed a 3D triculture model of neurons, astrocytes, and microglia that recapitulated AD features such as Aβ aggregation and phosphorylated Tau accumulation, while also demonstrating the involvement of microglia in neurotoxic activities and neuroinflammation. The inclusion of microglial cells in the model caused significant alterations in neuronal morphology, axonal damage, and a reduction in the surface area of neurons and astrocytes, accompanied by decreased cellular density and substantial losses of both cell types. The negative impact of microglia on neurons was found to be mediated through the activation of interferon (IFN)-γ and TLR-4 signaling pathways. Inhibition of these pathways decreased the production of neurotoxic mediators and mitigated cell death [225]. Another study investigated the interaction between microglia and neutrophils, shedding light on the role of peripheral immune cells in AD neuroinflammation. The results revealed that Aβ-challenged microglia released chemoattractants that recruited neutrophils, resulting in the production of inflammatory mediators. This interaction between microglia and neutrophils contributed to neuroinflammation in AD. The findings suggest that targeting the communication between central and peripheral immune communities could be a potential strategy for alleviating immunological burdens in neuroinflammatory CNS diseases [226]. Overall, both studies provide valuable insights into cell-to-cell interactions and immune responses in AD, offering potential avenues for therapeutic interventions. A brain-on-a-chip model was used to investigate different aspects of neurovascular interactions and neurodegeneration, in two studies. The first study focused on developing a 3D in vitro model of the neurovascular unit (NVU) using six types of cells. This model successfully mimicked the brain microvasculature and demonstrated mature barrier function crucial for maintaining the integrity of the NVU. Additionally, the presence of multiple cell types in the co-culture system was found to modulate the inflammatory response in the NVU through the cytokine-mediated pathway [227]. A following study investigated the effects of particulate matter, specifically diesel exhaust particles (DEP), on neurodegeneration. The platform simulated neuro–glia–vascular interactions in a co-culture of brain endothelial cells (bECs), neurons, and glia. Exposure to DEP in the brain-on-a-chip model resulted in pathological features resembling AD, specifically, phosphorylated Tau and Aβ level changes, excessive production of hydrogen peroxide and reactive oxygen species (H2O2/ROS), and neuronal loss. Results showed that secretion of granulocyte–macrophage colony-stimulating factor by bECs, contributed to microglial activation and led to the overproduction of H2O2/ROS [228]. Jorfi et al. in 2018 cultured neuro-spheroids from engineered human neural stem cells, to overexpress APP and PSEN1, and hiPSCs in microwell array, incorporated in Matrigel™. The platform allowed neuro-spheroids to be held in place during cell differentiation. This brain-on-chip system can be used for studying AD pathogenesis and multiple drug treatments as each neuro-spheroid can be tested individually, and AD cells showed extracellular Aβ aggregates, and intracellular phosphorylated Tau [229]. The same group also assessed the infiltration of peripheral blood mononuclear cells into an AD brain 3D model made of NPC modified to overexpress Aβ, astrocytes and iPSCs-derived microglia, cultured in Matrigel™. The 3D model was cultured in the central chamber of a microfluidic chip, while different cells from the peripheral blood, namely CD8+/CD4+/CD3+ T cells, monocytes, and B cells, were seeded in adjacent chambers, connected by microchannels. Data suggested that the infiltration of CD8+ T cells into the AD 3D model enhanced microglial activation, neuroinflammation and neurodegeneration, by activating INF-γ and neuroinflammatory pathways in the glial cells [230]. A large focus of organ-on-chip platforms for the study of PD attempt to model neuroinflammation to elucidate the physiological mechanisms behind the disease. These models mainly integrate immune cells like microglia, and astrocytes that densely populate the substantia nigra while also taking into consideration infiltrating immune cells like monocytes, as well as certain subtypes of T cells as the disease progresses [76]. One of the first studies that modeled α-syn spreading in a co-cultured system of N9 microglial-like cells and H4 neuroglioma cells on-chip was conducted by Fernandes et al. In this study, a PDMS microfluidic platform was designed to house human H4 neuroglioma cells and N9 microglial cells in two chambers connected by three channels with flow being controlled with integrated pneumatic valves. This device provided the rapid spread of molecules and a regulated microenvironment, thereby imitating the process of paracrine signaling, as observed with the increase in levels of reactive oxygen in the presence of activated N9 cells [231]. Since then, organ-on-chip technologies have paved the way to study of the progression of PD with respect to the BBB. For example, Jacquet et al. examined the effect of inflammatory astrocytes on the BBB by utilizing a PD microfluidic chip model. The chip is composed of 3 lanes allowing for co-culturing endothelial-like vessels (top layer), pericytes (middle), and astrocytes (bottom layer) under perfusion to mimic the in vivo shear forces of the BBB microenvironment. This study demonstrated the same increase in blood vessel diameter as seen in human post-mortem PD brain tissue. This further highlights how inflammatory astrocytes disrupt the BBB via vascular abnormalities which may lead to PD progression [232]. Pediaditakis et al. demonstrated that astrocytes and microglia’s reactivity induced by α-syn on their substantia nigra brain-chip. Their PDMS-based model contained both a vascular channel and brain channel with seeded iPSC-derived dopaminergic neurons in addition to human primary brain astrocytes, microglia and pericytes to create a vascular–neuronal interface. Here, trehalose, a disaccharide known to decrease aggregation of proteins and neurodegeneration, had protective effects against neuroinflammation caused by α-syn, resulting in lower levels of inflammatory cytokines TNF-α and IL-6 [233]. This suggests the significance of neuroinflammation to PD pathology and progression in addition to the versatility of the organ-on-chip platform to accurately model this neuroimmune-specific behavior.

These studies collectively emphasize the importance of studying intercellular crosstalk using human cell-based models to gain insights into neurodegenerative diseases, bridging the gap between physiological and pathological aspects of the human brain. However, despite the growing research on intercellular crosstalk and human cell-based models, studies dedicated to understanding the intricate mechanisms underlying AD and PD remain relatively sparse.

Conclusion and future directions

The intricate interactions and coordination between innate and adaptive immune cells play a pivotal role in the exacerbation of AD and PD pathology. The delicate balance between the overexpression of pro-inflammatory cytokines and the phenotypic transformation of these immune cells is a crucial factor in controlling chronic inflammation, to prevent the onset and progression of neuroimmune diseases. Nevertheless, further research is still required to unravel the complex mechanisms underlying the action and interaction of these immune cells within the context of AD and PD pathology. Modulating the function of these different immune cells holds promise for the development of better targeted and more effective treatments for AD and PD. To study AD and PD, various models have been developed, including animal models, cellular models, and more recently 3D microfluidics models. These models provide a controlled environment to investigate disease mechanisms, evaluate drug candidates, and explore the dynamic interplay between cells and tissues. However, very few in vitro studies explore the dynamic between the immune and the central nervous systems. Understanding how to modulate the immune response could reduce neuroinflammation, delay the disease progression, and eventually provide effective treatments.

Microfluidics models offer several advantages, including the ability to mimic the complex architecture of brain tissue, incorporate multiple cell types, and provide precise control over fluid flow and environmental conditions. Thus, microfluidics models in 3D enable researchers to study the intricate interactions between neuronal cells, immune cells, and the surrounding microenvironment. By using such advanced models, we can gain a deeper understanding of the disease processes and potentially identify novel therapeutic strategies. However, as stated in Sect. 4.5, there is a scarcity of studies on microfluidic chips for AD and PD. Using microfluidic devices could help to reduce the use of animal models, which are not able to provide a complete overview of human pathology. Microfluidic devices will also allow to study the paracrine interaction between immune cells and neurons, which play a key role in the progression of AD and PD.

Availability of data and materials

Not applicable.

Abbreviations

AD:

Alzheimer’s disease

APOE:

Apolipoprotein E

APP:

Amyloid precursor protein

Aβ:

Amyloid beta

BBB:

Brain–blood barrier

BDNF:

Brain-derived neurotrophic factor

CNS:

Central nervous system

DAN:

Dopaminergic neurons

IL:

Interleukin

Ig:

Immunoglobulin

INF:

Interferon

IPSCs:

Induced pluripotent stem cells

MCI:

Mild cognitive impairment

PD:

Parkinson’s disease

PDMS:

Polydimethylsiloxane

TLR:

Toll-like receptor

TNF-α:

Tumor necrosis factor

TREM:

Triggering receptor expressed on myeloid cells

α-syn:

α-Synuclein

References

  1. Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR, Mehrpour M, et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol. 2014;112:24–49.

    Article  CAS  PubMed  Google Scholar 

  2. Hennessy E, Griffin EW, Cunningham C. Astrocytes are primed by chronic neurodegeneration to produce exaggerated chemokine and cell infiltration responses to acute stimulation with the cytokines IL-1β and TNF-α. J Neurosci. 2015;35(22):8411–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Vedam-Mai V. Harnessing the immune system for the treatment of Parkinson’s disease. Brain Res. 2021;1758: 147308.

    Article  CAS  PubMed  Google Scholar 

  4. Zieneldien T, Kim J, Sawmiller D, Cao C. The immune system as a therapeutic target for Alzheimer’s disease. Life. 2022;12(9):1440.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Erkkinen MG, Kim MO, Geschwind MD. Clinical neurology and epidemiology of the major neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2017;10(4): a033118.

    Article  Google Scholar 

  6. Cetin S, Knez D, Gobec S, Kos J, Pišlar A. Cell models for Alzheimer’s and Parkinson’s disease: at the interface of biology and drug discovery. Biomed Pharmacother. 2022;149: 112924.

    Article  CAS  PubMed  Google Scholar 

  7. Kim TW, Koo SY, Studer L. Pluripotent stem cell therapies for Parkinson disease: present challenges and future opportunities. Front Cell Dev Biol. 2020;8:729.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Slanzi A, Iannoto G, Rossi B, Zenaro E, Constantin G. In vitro models of neurodegenerative diseases. Front Cell Dev Biol. 2020;8:328.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Shen XN, Niu LD, Wang YJ, Cao XP, Liu Q, Tan L, et al. Inflammatory markers in Alzheimer’s disease and mild cognitive impairment: a meta-analysis and systematic review of 170 studies. J Neurol Neurosurg Psychiatry. 2019;90(5):590–8.

    Article  PubMed  Google Scholar 

  10. King E, O’Brien J, Donaghy P, Williams-Gray CH, Lawson RA, Morris CM, et al. Inflammation in mild cognitive impairment due to Parkinson’s disease, Lewy body disease, and Alzheimer’s disease. Int J Geriatr Psychiatry. 2019;34(8):1244–50.

    Article  PubMed  Google Scholar 

  11. Degerskär ANW, Englund EM. Cause of death in autopsy-confirmed dementia disorders. Eur J Neurol. 2020;27(12):2415–21.

    Article  PubMed  Google Scholar 

  12. Centeno EGZ, Cimarosti H, Bithell A. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol Neurodegener. 2018;13(1):27.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Mockett BG, Richter M, Abraham WC, Müller UC. Therapeutic potential of secreted amyloid precursor protein APPsα. Front Mol Neurosci. 2017;10.

  14. Zaretsky DV, Zaretskaia M. Degradation products of amyloid protein: are they the culprits? Curr Alzheimer Res. 2020;17(10):869–80.

    Article  CAS  PubMed  Google Scholar 

  15. Rischel EB, Gejl M, Brock B, Rungby J, Gjedde A. In Alzheimer’s disease, amyloid beta accumulation is a protective mechanism that ultimately fails. Alzheimer’s & Dementia. 2022;19(3):771–83.

    Article  Google Scholar 

  16. National Institute on Aging. Alzheimer’s Disease Genetics Fact Sheet [Internet]. [cited 2023 Aug 31]. Available from: https://www.nia.nih.gov/health/alzheimers-disease-genetics-fact-sheet.

  17. Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet. 2009;41(10):1094–9.

    Article  CAS  PubMed  Google Scholar 

  18. Ulland TK, Colonna M. TREM2—a key player in microglial biology and Alzheimer disease. Nat Rev Neurol. 2018;14(11):667–75.

    Article  CAS  PubMed  Google Scholar 

  19. Sinsky J, Majerova P, Kovac A, Kotlyar M, Jurisica I, Hanes J. Physiological tau interactome in brain and its link to tauopathies. J Proteome Res. 2020;19(6):2429–42.

    Article  CAS  PubMed  Google Scholar 

  20. Trojanowski JQ, Shin RW, Schmidt ML, Lee VMY. Relationship between plaques, tangles, and dystrophic processes in Alzheimer’s disease. Neurobiol Aging. 1995;16(3):335–40.

    Article  CAS  PubMed  Google Scholar 

  21. Ferguson AC, Tank R, Lyall LM, Ward J, Celis-Morales C, Strawbridge R, et al. Alzheimer’s disease susceptibility gene apolipoprotein E (APOE) and blood biomarkers in UK biobank (N = 395,769). Beason-Held L, editor. J Alzheimer’s Dis. 2020;1–11.

  22. Van Oostveen WM, De Lange ECM, Orzechowski A. Molecular sciences imaging techniques in Alzheimer’s disease: a review of applications in early diagnosis and longitudinal monitoring. Int J Mol Sci. 2021;22(4):2110.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Sutherland K, Li T, Cao C. Alzheimer’s disease and the immune system. SOJ Neurol. 2015;2(1).

  24. Tan AH, Hor JW, Chong CW, Lim SY. Probiotics for Parkinson’s disease: current evidence and future directions. JGH Open. 2021;5(4):414–9.

    Article  PubMed  Google Scholar 

  25. Bettcher BM, Tansey MG, Dorothée G, Heneka MT. Peripheral and central immune system crosstalk in Alzheimer disease—a research prospectus. Nat Rev Neurol. 2021;17(11):689–701.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Piao J, Zabierowski S, Dubose BN, Hill EJ, Navare M, Claros N, et al. Preclinical efficacy and safety of a human embryonic stem cell-derived midbrain dopamine progenitor product, MSK-DA01. Cell Stem Cell. 2021;28(2):217–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Qiu B, Bessler N, Figler K, Buchholz MB, Rios AC, Malda J, et al. Bioprinting neural systems to model central nervous system diseases. Adv Funct Mater. 2020;30(44):1910250.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jasmin M, Ahn EH, Voutilainen MH, Fombonne J, Guix C, Viljakainen T, et al. Netrin‐1 and its receptor DCC modulate survival and death of dopamine neurons and Parkinson’s disease features. EMBO J. 2020;40(3).

  29. Fouke KE, Wegman ME, Weber SA, Brady EB, Román-Vendrell C, Morgan JR. Synuclein regulates synaptic vesicle clustering and docking at a vertebrate synapse. Front Cell Dev Biol. 2021;26:9.

    Google Scholar 

  30. Yoo G, Shin YK, Lee NK. The role of α-synuclein in SNARE-mediated synaptic vesicle fusion. J Mol Biol. 2023;435(1):167775.

    Article  CAS  PubMed  Google Scholar 

  31. Li Y, Zhu Z, Chen J, Zhang M, Yang Y, Huang P. Dilated perivascular space in the midbrain may reflect dopamine neuronal degeneration in Parkinson’s disease. Front Aging Neurosci. 2020;12.

  32. Oliveira FPM, Walker Z, Walker RWH, Attems J, Castanheira JC, Silva Â, et al. 123I-FP-CIT SPECT in dementia with Lewy bodies, Parkinson’s disease and Alzheimer’s disease: a new quantitative analysis of autopsy confirmed cases. J Neurol Neurosurg Psychiatry. 2021;92(6):662–7.

    Article  Google Scholar 

  33. Johns Hopkins Medicine. The Genetic Link to Parkinson’s Disease [Internet]. [cited 2023 Aug 31]. Available from: https://www.hopkinsmedicine.org/health/conditions-and-diseases/parkinsons-disease/the-genetic-link-to-parkinsons-disease.

  34. Hatton C, Reeve A, Lax NZ, Blain A, Ng YS, El-Agnaf O, et al. Complex I reductions in the nucleus basalis of Meynert in Lewy body dementia: the role of Lewy bodies. Acta Neuropathol Commun. 2020;8(1).

  35. Kalia LV, Lang AE. Parkinson’s disease. The Lancet. 2015;386(9996):896–912.

    Article  CAS  Google Scholar 

  36. Han JW, Ahn YD, Kim WS, Shin CM, Jeong SJ, Song YS, et al. Psychiatric manifestation in patients with Parkinson’s disease. J Korean Med Sci. 2018;33(47).

  37. Lin WC, Lee PL, Lu CH, Lin CP, Chou KH. Linking stage-specific plasma biomarkers to gray matter atrophy in Parkinson disease. Am J Neuroradiol. 2021;42(8):1444–51.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Chen Z, Wu B, Li G, Zhou L, Zhang L, Liu J. Age and sex differentially shape brain networks in Parkinson’s disease. CNS Neurosci Ther. 2023;29:1907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Picard K, Bisht K, Poggini S, Garofalo S, Golia MT, Basilico B, et al. Microglial-glucocorticoid receptor depletion alters the response of hippocampal microglia and neurons in a chronic unpredictable mild stress paradigm in female mice. Brain Behav Immun. 2021;97:423–39.

    Article  CAS  PubMed  Google Scholar 

  40. Basilico B, Ferrucci L, Ratano P, Golia MT, Grimaldi A, Rosito M, et al. Microglia control glutamatergic synapses in the adult mouse hippocampus. Glia. 2022;70(1):173–95.

    Article  CAS  PubMed  Google Scholar 

  41. Wang S, Sudan R, Peng V, Zhou Y, Du S, Yuede CM, et al. TREM2 drives microglia response to amyloid-β via SYK-dependent and -independent pathways. Cell. 2022;185(22):4153-4169.e19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yanguas-Casás N, Crespo-Castrillo A, Arevalo MA, Garcia-Segura LM. Aging and sex: impact on microglia phagocytosis. Aging Cell. 2020;19(8): e13182.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Devanney NA, Stewart AN, Gensel JC. Microglia and macrophage metabolism in CNS injury and disease: the role of immunometabolism in neurodegeneration and neurotrauma. Exp Neurol. 2020;329: 113310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Verdon DJ, Mulazzani M, Jenkins MR. Cellular and molecular mechanisms of CD8+ T cell differentiation, dysfunction and exhaustion. Int J Mol Sci. 2020;21(19):7357.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rocamora-Reverte L, Melzer FL, Würzner R, Weinberger B. The complex role of regulatory T cells in immunity and aging. Front Immunol. 2021;11.

  46. Kim K, Wang X, Ragonnaud E, Bodogai M, Illouz T, DeLuca M, et al. Therapeutic B-cell depletion reverses progression of Alzheimer’s disease. Nat Commun. 2021;12(1):2185.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s & Dementia. 2018;4(1):575–90.

    Google Scholar 

  48. Paolicelli RC, Sierra A, Stevens B, Tremblay ME, Aguzzi A, Ajami B, et al. Microglia states and nomenclature: a field at its crossroads. Neuron. 2022;110(21):3458–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gross AL, Walker KA, Moghekar AR, Pettigrew C, Soldan A, Albert MS, et al. Plasma markers of inflammation linked to clinical progression and decline during preclinical AD. Front Aging Neurosci. 2019;11.

  50. Morgan AR, Touchard S, Leckey C, O’Hagan C, Nevado-Holgado AJ, et al. Inflammatory biomarkers in Alzheimer’s disease plasma. Alzheimer’s & Dementia. 2019;15(6):776–87.

    Article  Google Scholar 

  51. Shabestari SK, Morabito S, Danhash EP, McQuade A, Sanchez JR, Miyoshi E, et al. Absence of microglia promotes diverse pathologies and early lethality in Alzheimer’s disease mice. Cell Rep. 2022;39(11):110961.

    Article  PubMed Central  Google Scholar 

  52. St-Pierre MK, VanderZwaag J, Loewen S, Tremblay MÈ. All roads lead to heterogeneity: the complex involvement of astrocytes and microglia in the pathogenesis of Alzheimer’s disease. Front Cell Neurosci. 2022;16.

  53. Bennett FC, Liddelow SA. Microglia metabolic breakdown drives Alzheimer’s pathology. Cell Metab. 2019;30(3):405–6.

    Article  CAS  PubMed  Google Scholar 

  54. De Schepper S, Ge JZ, Crowley G, Ferreira LSS, Garceau D, Toomey CE, et al. Perivascular cells induce microglial phagocytic states and synaptic engulfment via SPP1 in mouse models of Alzheimer’s disease. Nat Neurosci. 2023;26(3):406–15.

    PubMed  PubMed Central  Google Scholar 

  55. Bisht K, Sharma KP, Lecours C, Sánchez MG, El Hajj H, Milior G, et al. Dark microglia: a new phenotype predominantly associated with pathological states. Glia. 2016;64(5):826–39.

    Article  PubMed  PubMed Central  Google Scholar 

  56. St-Pierre MK, Šimončičová E, Bögi E, Tremblay MÈ. Shedding light on the dark side of the microglia. ASN Neuro. 2020;12:1759091420925335.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yan P, Kim KW, Xiao Q, Ma X, Czerniewski LR, Liu H, et al. Peripheral monocyte-derived cells counter amyloid plaque pathogenesis in a mouse model of Alzheimer’s disease. J Clin Invest. 2022;132(11): e152565.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Munawara U, Catanzaro M, Xu W, Tan C, Hirokawa K, Bosco N, et al. Hyperactivation of monocytes and macrophages in MCI patients contributes to the progression of Alzheimer’s disease. Immunity & Ageing. 2021;18(1):29.

    Article  CAS  Google Scholar 

  59. Sanchez-Sanchez JL, Giudici KV, Guyonnet S, Delrieu J, Li Y, Bateman RJ, et al. Plasma MCP-1 and changes on cognitive function in community-dwelling older adults. Alzheimer’s Res Ther. 2022;14(1):5.

    Article  CAS  Google Scholar 

  60. Machhi J, Yeapuri P, Lu Y, Foster E, Chikhale R, Herskovitz J, et al. CD4+ effector T cells accelerate Alzheimer’s disease in mice. J Neuroinflammation. 2021;18(1):272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhou J, Geng Y, Su T, Wang Q, Ren Y, Zhao J, et al. NMDA receptor-dependent prostaglandin-endoperoxide synthase 2 induction in neurons promotes glial proliferation during brain development and injury. Cell Rep. 2022;38(13):110557.

    Article  CAS  PubMed  Google Scholar 

  62. Batista AF, Rody T, Forny-Germano L, Cerdeiro S, Bellio M, Ferreira ST, et al. Interleukin-1β mediates alterations in mitochondrial fusion/fission proteins and memory impairment induced by amyloid-β oligomers. J Neuroinflammation. 2021;18(1):54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Plescher M, Seifert G, Hansen JN, Bedner P, Steinhäuser C, Halle A. Plaque-dependent morphological and electrophysiological heterogeneity of microglia in an Alzheimer’s disease mouse model. Glia. 2018;66(7):1464–80.

    Article  PubMed  Google Scholar 

  64. Franco-Bocanegra DK, Gourari Y, McAuley C, Chatelet DS, Johnston DA, Nicoll JAR, et al. Microglial morphology in Alzheimer’s disease and after Aβ immunotherapy. Sci Rep. 2021;11(1):15955.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Lee WJ, Liao YC, Wang YF, Lin IF, Wang SJ, Fuh JL. Plasma MCP-1 and cognitive decline in patients with Alzheimer’s disease and mild cognitive impairment: a two-year follow-up study. Sci Rep. 2018;8(1):1280.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Dai L, Wang Q, Lv X, Gao F, Chen Z, Shen Y. Elevated β-secretase 1 expression mediates CD4+ T cell dysfunction via PGE2 signalling in Alzheimer’s disease. Brain Behav Immun. 2021;98:337–48.

    Article  CAS  PubMed  Google Scholar 

  67. Unger MS, Li E, Scharnagl L, Poupardin R, Altendorfer B, Mrowetz H, et al. CD8+ T-cells infiltrate Alzheimer’s disease brains and regulate neuronal- and synapse-related gene expression in APP-PS1 transgenic mice. Brain Behav Immun. 2020;89:67–86.

    Article  CAS  PubMed  Google Scholar 

  68. Williams-Gray CH, Foltynie T, Brayne CEG, Robbins TW, Barker RA. Evolution of cognitive dysfunction in an incident Parkinson’s disease cohort. Brain. 2007;130(7):1787–98.

    Article  CAS  PubMed  Google Scholar 

  69. Williams-Gray CH, Mason SL, Evans JR, Foltynie T, Brayne C, Robbins TW, et al. The CamPaIGN study of Parkinson’s disease: 10-year outlook in an incident population-based cohort. J Neurol Neurosurg Psychiatry. 2013;84(11):1258–64.

    Article  PubMed  Google Scholar 

  70. Williams-Gray CH, Evans JR, Goris A, Foltynie T, Ban M, Robbins TW, et al. The distinct cognitive syndromes of Parkinson’s disease: 5 year follow-up of the CamPaIGN cohort. Brain. 2009;132(11):2958–69.

    Article  PubMed  Google Scholar 

  71. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat Genet. 2010;42(9):781–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hill-Burns EM, Factor SA, Zabetian CP, Thomson G, Payami H. Evidence for more than one Parkinson’s disease-associated variant within the HLA region. PLoS ONE. 2011;6(11): e27109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Pierce S, Coetzee GA. Parkinson’s disease-associated genetic variation is linked to quantitative expression of inflammatory genes. PLoS ONE. 2017;12(4): e0175882.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Witoelar A, Jansen IE, Wang Y, Desikan RS, Gibbs JR, Blauwendraat C, et al. Genome-wide pleiotropy between Parkinson disease and autoimmune diseases. JAMA Neurol. 2017;74(7):780.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Li M, Wan J, Xu Z, Tang B. The association between Parkinson’s disease and autoimmune diseases: a systematic review and meta-analysis. Front Immunol. 2023;14:1103053.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Tansey MG, Wallings RL, Houser MC, Herrick MK, Keating CE, Joers V. Inflammation and immune dysfunction in Parkinson disease. Nat Rev Immunol. 2022;22(11):657–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Awogbindin IO, Ishola IO, St-Pierre MK, Carrier M, Savage JC, Di Paolo T, et al. Remodeling microglia to a protective phenotype in Parkinson’s disease? Neurosci Lett. 2020;735: 135164.

    Article  CAS  PubMed  Google Scholar 

  78. De Biase LM, Schuebel KE, Fusfeld ZH, Jair K, Hawes IA, Cimbro R, et al. Local cues establish and maintain region-specific phenotypes of basal ganglia microglia. Neuron. 2017;95(2):341–56.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Tang Y, Li T, Li J, Yang J, Liu H, Zhang XJ, et al. Jmjd3 is essential for the epigenetic modulation of microglia phenotypes in the immune pathogenesis of Parkinson’s disease. Cell Death Differ. 2014;21(3):369–80.

    Article  CAS  PubMed  Google Scholar 

  80. Smajić S, Prada-Medina CA, Landoulsi Z, Ghelfi J, Delcambre S, Dietrich C, et al. Single-cell sequencing of human midbrain reveals glial activation and a Parkinson-specific neuronal state. Brain. 2022;145(3):964–78.

    Article  PubMed  Google Scholar 

  81. Kouli A, Camacho M, Allinson K, Williams-Gray CH. Neuroinflammation and protein pathology in Parkinson’s disease dementia. Acta Neuropathol Commun. 2020;8:1–19.

    Article  Google Scholar 

  82. Chaudhuri KR, Schapira AHV. Non-motor symptoms of Parkinson’s disease: dopaminergic pathophysiology and treatment. Lancet Neurol. 2009;8(5):464–74.

    Article  CAS  PubMed  Google Scholar 

  83. Pellegrini C, D’Antongiovanni V, Miraglia F, Rota L, Benvenuti L, Di Salvo C, et al. Enteric α-synuclein impairs intestinal epithelial barrier through caspase-1-inflammasome signaling in Parkinson’s disease before brain pathology. NPJ Parkinsons Dis. 2022;8(1):9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Sulzer D, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, et al. T cells from patients with Parkinson’s disease recognize α-synuclein peptides. Nature. 2017;546(7660):656–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Grozdanov V, Bliederhaeuser C, Ruf WP, Roth V, Fundel-Clemens K, Zondler L, et al. Inflammatory dysregulation of blood monocytes in Parkinson’s disease patients. Acta Neuropathol. 2014;128:651–63.

  86. Williams-Gray CH, Wijeyekoon R, Yarnall AJ, Lawson RA, Breen DP, Evans JR, et al. Serum immune markers and disease progression in an incident Parkinson’s disease cohort (ICICLE-PD). Mov Disord. 2016;31(7):995–1003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Chen X, Hu Y, Cao Z, Liu Q, Cheng Y. Cerebrospinal fluid inflammatory cytokine aberrations in Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Front Immunol. 2018;9:2122.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Yan Z, Yang W, Wei H, Dean MN, Standaert DG, Cutter GR, et al. Dysregulation of the adaptive immune system in patients with early-stage Parkinson disease. Neurol Neuroimmunol Neuroinflamm. 2021;8(5).

  89. Contaldi E, Magistrelli L, Comi C. T lymphocytes in Parkinson’s disease. J Parkinson’s Dis. 2022;12(s1):S65–74.

    Article  CAS  Google Scholar 

  90. Shalash A, Salama M, Makar M, Roushdy T, Elrassas HH, Mohamed W, et al. Elevated serum α-synuclein autoantibodies in patients with Parkinson’s disease relative to Alzheimer’s disease and controls. Front Neurol. 2017;8:720.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Li R, Tropea TF, Baratta LR, Zuroff L, Diaz-Ortiz ME, Zhang B, et al. Abnormal B-cell and Tfh-cell profiles in patients with Parkinson disease: a cross-sectional study. Neurol Neuroimmunol Neuroinflamm. 2022;9(2).

  92. Wang P, Luo M, Zhou W, Jin X, Xu Z, Yan S, et al. Global characterization of peripheral B cells in Parkinson’s disease by single-cell RNA and BCR sequencing. Front Immunol. 2022;13: 814239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Gray MT, Woulfe JM. Striatal blood–brain barrier permeability in Parkinson’s disease. J Cereb Blood Flow Metab. 2015;35(5):747–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, et al. Alzheimer’s disease. The Lancet. 2021;397(10284):1577–90.

    Article  CAS  Google Scholar 

  95. Khachaturian ZS. The ‘aducanumab story’: will the last chapter spell the end of the ‘amyloid hypothesis’ or mark a new beginning? J Prevent Alzheimer’s Dis. 2022;9(2):190–2.

    CAS  Google Scholar 

  96. Baruch K, Kertser A, Matalon O, Forsht O, Braiman S, Shochat E, et al. IBC-Ab002, an anti-PD-L1 monoclonal antibody tailored for treating Alzheimer’s disease. Alzheimer’s & Dementia. 2020;16(S9): e042978.

    Article  Google Scholar 

  97. Kang BW, Kim F, Cho JY, Kim SY, Rhee J, Choung JJ. Phosphodiesterase 5 inhibitor mirodenafil ameliorates Alzheimer-like pathology and symptoms by multimodal actions. Alzheimer’s Res Ther. 2022;14(1):1–17.

    Google Scholar 

  98. Koh SH, Kwon HS, Choi SH, Jeong JH, Na HR, Lee CN, et al. Efficacy and safety of GV1001 in patients with moderate-to-severe Alzheimer’s disease already receiving donepezil: a phase 2 randomized, double-blind, placebo-controlled, multicenter clinical trial. Alzheimer’s Res Ther. 2021;13(1):1–11.

    Google Scholar 

  99. Kaeberlein M, Galvan V. Rapamycin and Alzheimer’s disease: time for a clinical trial? Sci Transl Med. 2019;11(476).

  100. McShane R, Westby MJ, Roberts E, Minakaran N, Schneider L, Farrimond LE, et al. Memantine for dementia. Cochrane Database Syst Rev. 2019;2019(3):1–446.

    Google Scholar 

  101. Nørgaard CH, Friedrich S, Hansen CT, Gerds T, Ballard C, Møller DV, et al. Treatment with glucagon‐like peptide‐1 receptor agonists and incidence of dementia: data from pooled double‐blind randomized controlled trials and nationwide disease and prescription registers. Alzheimer’s & Dementia. 2022;8(1).

  102. Hua X, Church K, Walker W, L’Hostis P, Viardot G, Danjou P, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of the positive modulator of HGF/MET, fosgonimeton, in healthy volunteers and subjects with Alzheimer’s disease: randomized, placebo-controlled, double-blind, phase I clinical trial. J Alzheimer’s Dis. 2022;86(3):1399.

    Article  CAS  Google Scholar 

  103. Alzheimer Disease—ClinicalTrials.gov [Internet]. [cited 2023 Apr 5]. Available from: https://clinicaltrials.gov/ct2/results?term=drugs&cond=Alzheimer+Disease&Search=Apply&recrs=b&recrs=a&recrs=f&recrs=d&age_v=&gndr=&type=&rslt=.

  104. Mehanna R, Lai EC. Deep brain stimulation in Parkinson’s disease. Transl Neurodegener. 2013;2(1):22.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Mehanna R, Bajwa JA, Fernandez H, Wagle Shukla AA, others. Cognitive impact of deep brain stimulation on Parkinson’s disease patients. Parkinson’s Dis. 2017;2017.

  106. Stoker TB, Barker RA. Recent developments in the treatment of Parkinson’s disease. F1000Res. 2020;9:862.

    Article  CAS  Google Scholar 

  107. Marucci G, Buccioni M, Ben DD, Lambertucci C, Volpini R, Amenta F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology. 2021;1(190): 108352.

    Article  Google Scholar 

  108. van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, et al. Lecanemab in early Alzheimer’s disease. N Engl J Med. 2023;388(1):9–21.

    Article  PubMed  Google Scholar 

  109. FDA Grants Accelerated Approval for Alzheimer’s Disease Treatment | FDA [Internet]. [cited 2023 Mar 23]. Available from: https://www.fda.gov/news-events/press-announcements/fda-grants-accelerated-approval-alzheimers-disease-treatment.

  110. Larkin HD. Lecanemab gains FDA approval for early Alzheimer disease. JAMA. 2023;329(5):363–363.

    PubMed  Google Scholar 

  111. Tolar M, Abushakra S, Hey JA, Porsteinsson A, Sabbagh M. Aducanumab, gantenerumab, BAN2401, and ALZ-801—the first wave of amyloid-targeting drugs for Alzheimer’s disease with potential for near term approval. Alzheimers Res Ther. 2020;12(1).

  112. ANAVEX®2-73 (BLARCAMESINE) PHASE 2B/3 STUDY MET PRIMARY AND KEY SECONDARY ENDPOINTS, [Internet]. [cited 2023 Mar 22]. Available from: https://www.anavex.com/post/anavex-2-73-blarcamesine-phase-2b-3-study-met-primary-and-key-secondary-endpoints.

  113. Yiannopoulou KG, Papageorgiou SG. Current and future treatments in Alzheimer disease: an update. J Cent Nerv Syst Dis. 2020;29:12.

    Google Scholar 

  114. Tran TN, Vo TNN, Frei K, Truong DD. Levodopa-induced dyskinesia: clinical features, incidence, and risk factors. J Neural Transm. 2018;125:1109–17.

    Article  CAS  PubMed  Google Scholar 

  115. Turcano P, Mielke MM, Bower JH, Parisi JE, Cutsforth-Gregory JK, Ahlskog JE, et al. Levodopa-induced dyskinesia in Parkinson disease: a population-based cohort study. Neurology. 2018;91(24):e2238–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Carbone F, Djamshidian A, Seppi K, Poewe W. Apomorphine for Parkinson’s disease: efficacy and safety of current and new formulations. CNS Drugs. 2019;33(9):905–18.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Khalifeh M, Read MI, Barreto GE, Sahebkar A. Trehalose against Alzheimer’s disease: insights into a potential therapy. BioEssays. 2020;42(8):1900195.

    Article  CAS  Google Scholar 

  118. Jin LW, Di LJ, Nguyen HM, Singh V, Singh L, Chavez M, et al. Repurposing the KCa3.1 inhibitor senicapoc for Alzheimer’s disease. Ann Clin Transl Neurol. 2019;6(4):723–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Cummings J, Lee G, Nahed P, Kambar MEZN, Zhong K, Fonseca J, et al. Alzheimer’s disease drug development pipeline: 2022. Alzheimer’s and Dementia. 2022;8(1).

  120. Patel AG, Nehete PN, Krivoshik SR, Pei X, Cho EL, Nehete BP, et al. Innate immunity stimulation via CpG oligodeoxynucleotides ameliorates Alzheimer’s disease pathology in aged squirrel monkeys. Brain. 2021;144(7):2146.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Wang YL, Zhang Y, Xu J. Sodium oligomannate combined with rivastigmine may improve cerebral blood flow and cognitive impairment following CAR-T cell therapy: a case report. Front Oncol. 2022;18:12.

    Google Scholar 

  122. Potter H, Woodcock JH, Boyd TD, Coughlan CM, O’Shaughnessy JR, Borges MT, et al. Safety and efficacy of sargramostim (GM-CSF) in the treatment of Alzheimer’s disease. Alzheimer’s & Dementia. 2021;7(1): e12158.

    Google Scholar 

  123. Ward M, Long H, Schwabe T, Rhinn H, Tassi I, Salazar SV, et al. A phase 1 study of AL002 in healthy volunteers. Alzheimer’s & Dementia. 2021;17(S9): e054669.

    Article  Google Scholar 

  124. Torres-Acosta N, O’Keefe JH, O’Keefe EL, Isaacson R, Small G. Therapeutic potential of TNF-α inhibition for Alzheimer’s disease prevention. J Alzheimer’s Dis. 2020;78(2):619.

    Article  CAS  Google Scholar 

  125. MacPherson KP, Sompol P, Kannarkat GT, Chang J, Sniffen L, Wildner ME, et al. Peripheral administration of the soluble TNF inhibitor XPro1595 modifies brain immune cell profiles, decreases beta-amyloid plaque load, and rescues impaired long-term potentiation in 5xFAD mice. Neurobiol Dis. 2017;1(102):81.

    Article  Google Scholar 

  126. Lennard L. The clinical pharmacology of 6-mercaptopurine. Eur J Clin Pharmacol. 1992;43(4):329–39.

    Article  CAS  PubMed  Google Scholar 

  127. Labanieh L, Majzner RG, Mackall CL. Programming CAR-T cells to kill cancer. Nat Biomed Eng. 2018;2(6):377–91.

    Article  CAS  PubMed  Google Scholar 

  128. Nickerson P, Jeffery J, Rush D. Long-term allograft surveillance: the role of protocol biopsies. Curr Opin Urol. 2001;11(2):133–7.

    Article  CAS  PubMed  Google Scholar 

  129. Candini O, Grisendi G, Foppiani EM, Brogli M, Aramini B, Masciale V, et al. A novel 3D in vitro platform for pre-clinical investigations in drug testing, gene therapy, and immuno-oncology. Sci Rep. 2019;9(1):1–12.

    Article  CAS  Google Scholar 

  130. Jin Z, Li X, Zhang X, Desousa P, Xu T, Wu A. Engineering the fate and function of human T-Cells via 3D bioprinting. Biofabrication. 2021;13(3): 035016.

    Article  CAS  Google Scholar 

  131. Gray JI, Westerhof LM, MacLeod MKL. The roles of resident, central and effector memory CD4 T-cells in protective immunity following infection or vaccination. Immunology. 2018;154(4):574–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Snow AL, Oliveira JB, Zheng L, Dale JK, Fleisher TA, Lenardo MJ. Critical role for BIM in T cell receptor restimulation-induced death. Biol Direct. 2008;3(1):1–17.

    Article  Google Scholar 

  133. Leach DG, Young S, Hartgerink JD. Advances in immunotherapy delivery from implantable and injectable biomaterials. Acta Biomater. 2019;1(88):15–31.

    Article  Google Scholar 

  134. Lu W, Wei Y, Cao Y, Xiao X, Li Q, Lyu H, et al. CD19 CAR-T cell treatment conferred sustained remission in B-ALL patients with minimal residual disease. Cancer Immunol Immunother. 2021;70(12):3501–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Goodridge JP, Reiser JW, Bjordahl R, Mandal M, Chang C, Clarck R, et al. Combinational strategy targeting B cell malignancy using iPSC engineered CAR-NK (FT596) and CAR-T cell (FT819) platforms with therapeutic antibody to achieve an effective deep and durable response. Cancer Res. 2020;80(16_Supplement):2216–2216.

    Article  Google Scholar 

  136. Egrilmez MY, Karabay U, Aydemir S, Baykara B, Husemoglu RB. The cellular responses of human macrophages seeded on 3D printed thermoplastic polyurethane scaffold. J Med Innovat Technol. 2021;3(2):40–5.

    Article  Google Scholar 

  137. Xiao J, Gao Y. The manufacture of 3D printing of medical grade TPU. Prog Addit Manuf. 2017;2(3):117–23.

    Article  Google Scholar 

  138. Huang X, He C, Lin G, Lu L, Xing K, Hua X, et al. Induced CD10 expression during monocyte-to-macrophage differentiation identifies a unique subset of macrophages in pancreatic ductal adenocarcinoma. Biochem Biophys Res Commun. 2020;524(4):1064–71.

    Article  CAS  PubMed  Google Scholar 

  139. Aldrich A, Kuss MA, Duan B, Kielian T. 3D bioprinted scaffolds containing viable macrophages and antibiotics promote clearance of Staphylococcus aureus craniotomy-associated biofilm infection. ACS Appl Mater Interfaces. 2019;11(13):12298–307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Lefebvre M, Jacqueline C, Amador G, Le Mabecque V, Miegeville A, Potel G, et al. Efficacy of daptomycin combined with rifampicin for the treatment of experimental methicillin-resistant Staphylococcus aureus (MRSA) acute osteomyelitis. Int J Antimicrob Agents. 2010;36(6):542–4.

    Article  CAS  PubMed  Google Scholar 

  141. Woitschach F, Kloss M, Schlodder K, Borck A, Grabow N, Reisinger EC, et al. In vitro study of the interaction of innate immune cells with liquid silicone rubber coated with Zwitterionic methyl methacrylate and thermoplastic polyurethanes. Materials. 2021;14(20):5972.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. de Medeiros LM, De Bastiani MA, Rico EP, Schonhofen P, Pfaffenseller B, Wollenhaupt-Aguiar B, et al. Cholinergic differentiation of human neuroblastoma SH-SY5Y cell line and its potential use as an in vitro model for Alzheimer’s disease studies. Mol Neurobiol. 2019;56(11):7355–67.

    Article  PubMed  Google Scholar 

  143. Gan L, Cookson MR, Petrucelli L, La Spada AR. Converging pathways in neurodegeneration, from genetics to mechanisms. Nat Neurosci. 2018;21(10):1300–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Kovalevich J, Langford D. Considerations for the use of SH-SY5Y neuroblastoma cells in neurobiology. Methods Mol Biol. 2013;1078:9–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhao J, Liu X, Xia W, Zhang Y, Wang C. Targeting amyloidogenic processing of APP in Alzheimer’s disease. Front Mol Neurosci. 2020;4:13.

    Google Scholar 

  146. Xicoy H, Wieringa B, Martens GJM. The SH-SY5Y cell line in Parkinson’s disease research: a systematic review. Mol Neurodegener. 2017;12(1):1–11.

    Article  Google Scholar 

  147. Korecka JA, van Kesteren RE, Blaas E, Spitzer SO, Kamstra JH, Smit AB, et al. Phenotypic characterization of retinoic acid differentiated SH-SY5Y cells by transcriptional profiling. PLoS ONE. 2013;8(5): e63862.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Brown DG, Wobst HJ. Opportunities and challenges in phenotypic screening for neurodegenerative disease research. J Med Chem. 2019;63(5):1823–40.

    Article  PubMed  Google Scholar 

  149. Cooper O, Hargus G, Deleidi M, Blak A, Osborn T, Marlow E, et al. Differentiation of human ES and Parkinson’s disease iPS cells into ventral midbrain dopaminergic neurons requires a high activity form of SHH, FGF8a and specific regionalization by retinoic acid. Mol Cell Neurosci. 2010;45(3):258–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Hargus G, Cooper O, Deleidi M, Levy A, Lee K, Marlow E, et al. Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proc Natl Acad Sci. 2010;107(36):15921–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Ochalek A, Szczesna K, Petazzi P, Kobolak J, Dinnyes A. Generation of cholinergic and dopaminergic interneurons from human pluripotent stem cells as a relevant tool for in vitro modeling of neurological disorders and therapy. Stem Cells Int. 2016;2016.

  152. Agholme L, Lindström T, Kågedal K, Marcusson J, Hallbeck M. An in vitro model for neuroscience: differentiation of SH-SY5Y cells into cells with morphological and biochemical characteristics of mature neurons. J Alzheimer’s Dis. 2010;20(4):1069–82.

    Article  CAS  Google Scholar 

  153. Hossini AM, Megges M, Prigione A, Lichtner B, Toliat MR, Wruck W, et al. Induced pluripotent stem cell-derived neuronal cells from a sporadic Alzheimer’s disease donor as a model for investigating AD-associated gene regulatory networks. BMC Genomics. 2015;16:1–22.

    CAS  Google Scholar 

  154. Zhang D, Pekkanen-Mattila M, Shahsavani M, Falk A, Teixeira AI, Herland A. A 3D Alzheimer’s disease culture model and the induction of P21-activated kinase mediated sensing in iPSC derived neurons. Biomaterials. 2014;35(5):1420–8.

    Article  CAS  PubMed  Google Scholar 

  155. Choi SH, Kim YH, Quinti L, Tanzi RE, Kim DY. 3D culture models of Alzheimer’s disease: a road map to a “cure-in-a-dish.” Mol Neurodegener. 2016;11:1–11.

    Article  Google Scholar 

  156. Sharma NS, Karan A, Lee D, Yan Z, Xie J. Advances in modeling Alzheimer’s disease in vitro. Adv Nanobiomed Res. 2021;1(12):2100097.

    Article  CAS  Google Scholar 

  157. Lee HK, Velazquez Sanchez C, Chen M, Morin PJ, Wells JM, Hanlon EB, et al. Three dimensional human neuro-spheroid model of Alzheimer’s disease based on differentiated induced pluripotent stem cells. PLoS ONE. 2016;11(9): e0163072.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Hernández-Sapiéns MA, Reza-Zaldívar EE, Cevallos RR, Márquez-Aguirre AL, Gazarian K, Canales-Aguirre AA. A three-dimensional Alzheimer’s disease cell culture model using iPSC-derived neurons carrying A246E mutation in PSEN1. Front Cell Neurosci. 2020;14:151.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Lomoio S, Pandey RS, Rouleau N, Menicacci B, Kim W, Cantley WL, et al. 3D bioengineered neural tissue generated from patient-derived iPSCs mimics time-dependent phenotypes and transcriptional features of Alzheimer’s disease. Mol Psychiatry. 2023;1–12.

  160. Kwak SS, Washicosky KJ, Brand E, Von Maydell D, Aronson J, Kim S, et al. Amyloid-β42/40 ratio drives tau pathology in 3D human neural cell culture models of Alzheimer’s disease. Nat Commun. 2020;11(1):1377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Taylor-Whiteley TR, Le Maitre CL, Duce JA, Dalton CF, Smith DP. Recapitulating Parkinson’s disease pathology in a three-dimensional human neural cell culture model. Dis Model Mech. 2019;12(4):dmm038042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Moreno EL, Hachi S, Hemmer K, Trietsch SJ, Baumuratov AS, Hankemeier T, et al. Differentiation of neuroepithelial stem cells into functional dopaminergic neurons in 3D microfluidic cell culture. Lab Chip. 2015;15(11):2419–28.

    Article  CAS  PubMed  Google Scholar 

  163. Fiore NJ, Ganat YM, Devkota K, Batorsky R, Lei M, Lee K, et al. Bioengineered models of Parkinson’s disease using patient-derived dopaminergic neurons exhibit distinct biological profiles in a 3D microenvironment. Cell Mol Life Sci. 2022;79(2):1–20.

    Article  Google Scholar 

  164. Abdelrahman S, Alsanie WF, Khan ZN, Albalawi HI, Felimban RI, Moretti M, et al. A Parkinson’s disease model composed of 3D bioprinted dopaminergic neurons within a biomimetic peptide scaffold. Biofabrication. 2022;14(4): 044103.

    Article  Google Scholar 

  165. Saylam E, Akkaya Y, Ilhan E, Cesur S, Guler E, Sahin A, et al. Levodopa-loaded 3D-printed poly (Lactic) acid/chitosan neural tissue scaffold as a promising drug delivery system for the treatment of Parkinson’s disease. Appl Sci. 2021;11(22):10727.

    Article  CAS  Google Scholar 

  166. Cuní-López C, Quek H, Oikari LE, Stewart R, Nguyen TH, Sun Y, et al. 3D models of Alzheimer’s disease patient microglia recapitulate disease phenotype and show differential drug responses compared to 2D. bioRxiv. 2021;2021–3.

  167. Sabate-Soler S, Louise Nickels S, Saraiva C, Berger E, Dubonyte U, Barmpa K, et al. Microglia integration into human midbrain organoids leads to increased neuronal maturation and functionality. Wiley Online Library. 2022;70(7):1267–88.

    CAS  Google Scholar 

  168. Markus B, Anna-Sophie S, Simon G, Dominik L, Alice K, Marvin R, et al. Incorporation of stem cell-derived astrocytes into neuronal organoids to allow neuro-glial interactions in toxicological studies. Altex. 2020;37(3):409–28.

    Google Scholar 

  169. Xu R, Boreland AJ, Li X, Erickson C, Jin M, Atkins C, et al. Developing human pluripotent stem cell-based cerebral organoids with a controllable microglia ratio for modeling brain development and pathology. Stem Cell Reports. 2021;16(8):1923–37.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Song L, Yuan X, Jones Z, Vied C, Miao Y, Marzano M, et al. Functionalization of brain region-specific spheroids with isogenic microglia-like cells. Sci Rep. 2019;9(1).

  171. Cai H, Ao Z, Hu L, Moon Y, Wu Z, Lu HC, et al. Acoustofluidic assembly of 3D neurospheroids to model Alzheimer’s disease. Analyst. 2020;145(19):6243–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Ilaria R, Marta T, Gianluigi F, Diego A, Carmen G. 3D brain tissue physiological model with co-cultured primary neurons and glial cells in hydrogels. J Tissue Eng. 2020;11:1–13.

    Google Scholar 

  173. Chemmarappally JM, Pegram HCN, Abeywickrama N, Fornari E, Hargreaves AJ, De Girolamo LA, et al. A co-culture nanofibre scaffold model of neural cell degeneration in relevance to Parkinson’s disease. Sci Rep. 2020;10(1):1–14.

    Article  Google Scholar 

  174. Rueda-Gensini L, Serna JA, Rubio D, Camilo Orozco J, Bolaños NI, Cruz JC, et al. Three-dimensional neuroimmune co-culture system for modeling Parkinson’s disease microenvironments in vitro. Biofabrication. 2023;15:45001.

    Article  Google Scholar 

  175. Drummond E, Wisniewski T. Alzheimer’s disease: experimental models and reality. Acta Neuropathol. 2017;133(2):155–75.

    Article  CAS  PubMed  Google Scholar 

  176. Sundaramoorthy TH, Castanho I. The Neuroepigenetic landscape of vertebrate and invertebrate models of neurodegenerative diseases. Epigenet Insights. 2022;4:15.

    Google Scholar 

  177. Fernandez-Funez P, de Mena L, Rincon-Limas DE. Modeling the complex pathology of Alzheimer’s disease in Drosophila. Exp Neurol. 2015;1(274):58–71.

    Article  Google Scholar 

  178. Alexander AG, Marfil V, Li C. Use of C. elegans as a model to study Alzheimer’s disease and other neurodegenerative diseases. Front Genet. 2014;5:95895.

    Article  Google Scholar 

  179. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spatzle/Toll/Cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86(6):973–83.

    Article  CAS  PubMed  Google Scholar 

  180. Tassetto M, Kunitomi M, Andino R. Circulating immune cells mediate a systemic RNAi-based adaptive antiviral response in drosophila. Cell. 2017;169(2):314-325.e13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Dhankhar J, Agrawal N, Shrivastava A. An interplay between immune response and neurodegenerative disease progression: an assessment using Drosophila as a model. J Neuroimmunol. 2020;15(346): 577302.

    Article  Google Scholar 

  182. Nagai J, Yu X, Papouin T, Cheong E. Behaviorally consequential astrocytic regulation of neural circuits. Neuron. 2021;109(4):576–96.

    Article  CAS  PubMed  Google Scholar 

  183. Sarparast M, Hinman J, Pourmand E, Vonarx D, Ramirez L, Ma W, et al. Cytochrome P450 and epoxide hydrolase metabolites in Aβ and tau-induced neurodegeneration: insights from Caenorhabditis elegans. bioRxiv. 2023;2023.10.02.560527.

  184. Stewart CR, Stuart LM, Wilkinson K, Van Gils JM, Deng J, Halle A, et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol. 2010;11(2):155.

    Article  CAS  PubMed  Google Scholar 

  185. Tan L, Schedl P, Song HJ, Garza D, Konsolaki M. The Toll–>NFkappaB signaling pathway mediates the neuropathological effects of the human Alzheimer’s Abeta42 polypeptide in Drosophila. PLoS ONE. 2008;3(12):e3966.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Zarini-Gakiye E, Sanadgol N, Parivar K, Vaezi G. Alpha-lipoic acid ameliorates tauopathy-induced oxidative stress, apoptosis, and behavioral deficits through the balance of DIAP1/DrICE ratio and redox homeostasis: age is a determinant factor. Metab Brain Dis. 2021;36(4):669–83.

    Article  CAS  PubMed  Google Scholar 

  187. Cowan CM, Sealey MA, Mudher A. Suppression of tau-induced phenotypes by vitamin E demonstrates the dissociation of oxidative stress and phosphorylation in mechanisms of tau toxicity. J Neurochem. 2021;157(3):684–94.

    Article  CAS  PubMed  Google Scholar 

  188. Lohr KM, Frost B, Scherzer C, Feany MB. Biotin rescues mitochondrial dysfunction and neurotoxicity in a tauopathy model. Proc Natl Acad Sci U S A. 2020;117(52):33608–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Siddique YH, Rahul A, Ara G, Afzal M, Varshney H, Gaur K, et al. Beneficial effects of apigenin on the transgenic Drosophila model of Alzheimer’s disease. Chem Biol Interact. 2022;366:110120.

    Article  CAS  PubMed  Google Scholar 

  190. Barati A, Masoudi R, Yousefi R, Monsefi M, Mirshafiey A. Tau and amyloid beta differentially affect the innate immune genes expression in Drosophila models of Alzheimer’s disease and β- D Mannuronic acid (M2000) modulates the dysregulation. Gene. 2022;15(808): 145972.

    Article  Google Scholar 

  191. Yang Y, Gehrke S, Imai Y, Huang Z, Ouyang Y, Wang JW, et al. Mitochondrial pathology and muscle and dopaminergic neuron degeneration caused by inactivation of Drosophila Pink1 is rescued by Parkin. Proc Natl Acad Sci U S A. 2006;103(28):10793–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Nayak N, Mishra M. Drosophila melanogaster as a model to understand the mechanisms of infection mediated neuroinflammation in neurodegenerative diseases. J Integr Neurosci. 2022;21(2):66.

    Article  PubMed  Google Scholar 

  193. Olsen AL, Feany MB. Parkinson’s disease risk genes act in glia to control neuronal α-synuclein toxicity. Neurobiol Dis. 2021;1(159): 105482.

    Article  Google Scholar 

  194. Ranjan VD, Qiu L, Tan EK, Zeng L, Zhang Y. Modelling Alzheimer’s disease: insights from in vivo to in vitro three-dimensional culture platforms. J Tissue Eng Regen Med. 2018;12(9):1944–58.

    Article  CAS  PubMed  Google Scholar 

  195. Doncheva NT, Palasca O, Yarani R, Litman T, Anthon C, Groenen MAM, et al. Human pathways in animal models: possibilities and limitations. Nucleic Acids Res. 2021;49(4):1859–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Spangenberg EE, Lee RJ, Najafi AR, Rice RA, Elmore MRP, Blurton-Jones M, et al. Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-β pathology. Brain. 2016;139(Pt 4):1265–81.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Dietrich K, Bouter Y, Müller M, Bayer TA. Synaptic alterations in mouse models for Alzheimer disease—a special focus on N-truncated abeta 4–42. Molecules. 2018;23(4):718.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Roda AR, Esquerda-Canals G, Martí-Clúa J, Villegas S. Cognitive impairment in the 3xTg-AD mouse model of Alzheimer’s disease is affected by Aβ-immunotherapy and cognitive stimulation. Pharmaceutics. 2020;12(10):944.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Li S, Jin M, Liu L, Dang Y, Ostaszewski BL, Selkoe DJ. Decoding the synaptic dysfunction of bioactive human AD brain soluble Aβ to inspire novel therapeutic avenues for Alzheimer’s disease. Acta Neuropathol Commun. 2018;6(1):121.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Weintraub MK, Kranjac D, Eimerbrink MJ, Pearson SJ, Vinson BT, Patel J, et al. Peripheral administration of poly I: C leads to increased hippocampal amyloid-beta and cognitive deficits in a non-transgenic mouse. Behav Brain Res. 2014;266:183–7.

    Article  CAS  PubMed  Google Scholar 

  201. Loewen SM, Chavesa AM, Murray CJ, Traetta ME, Burns SE, Pekarik KH, et al. The outcomes of maternal immune activation induced with the viral mimetic poly I:C on microglia in exposed rodent offspring. Dev Neurosci. 2023;45(4):191–209.

    Article  CAS  PubMed  Google Scholar 

  202. Cheng-Hathaway PJ, Reed-Geaghan EG, Jay TR, Casali BT, Bemiller SM, Puntambekar SS, et al. The Trem2 R47H variant confers loss-of-function-like phenotypes in Alzheimer’s disease. Mol Neurodegener. 2018;13(1):29.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA, Blumenthal PD, et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron. 2016;89(1):37–53.

    Article  CAS  PubMed  Google Scholar 

  204. Meeuwsen S, Bsibsi M, Persoon-Deen C, Ravid R, van Noort JM. Cultured human adult microglia from different donors display stable cytokine, chemokine and growth factor gene profiles but respond differently to a pro-inflammatory stimulus. NeuroImmunoModulation. 2005;12(4):235–45.

    Article  CAS  PubMed  Google Scholar 

  205. Stojkovska I, Wagner BM, Morrison BE. Parkinson’s disease and enhanced inflammatory response. Exp Biol Med. 2015;240(11):1387–95.

    Article  CAS  Google Scholar 

  206. Deng I, Corrigan F, Zhai G, Zhou XF, Bobrovskaya L. Lipopolysaccharide animal models of Parkinson’s disease: recent progress and relevance to clinical disease. Brain Behavior Immunity-Health. 2020;4: 100060.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Brundin P, Melki R, Kopito R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat Rev Mol Cell Biol. 2010;11(4):301–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Cameron T, Bennet T, Rowe EM, Anwer M, Wellington CL, Cheung KC. Review of design considerations for brain-on-a-chip models. Micromachines (Basel). 2021;12(4):441.

    Article  PubMed  Google Scholar 

  209. Cameron TC, Randhawa A, Grist SM, Bennet T, Hua J, Alde LG, et al. PDMS organ-on-chip design and fabrication: strategies for improving fluidic integration and chip robustness of rapidly prototyped microfluidic in vitro models. Micromachines (Basel). 2022;13(10):1573.

    Article  PubMed  Google Scholar 

  210. Campbell SB, Wu Q, Yazbeck J, Liu C, Okhovatian S, Radisic M. Beyond polydimethylsiloxane: alternative materials for fabrication of organ-on-a-chip devices and microphysiological systems. ACS Biomater Sci Eng. 2020;7(7):2880–99.

    Article  PubMed  Google Scholar 

  211. Wu Q, Liu J, Wang X, Feng L, Wu J, Zhu X, et al. Organ-on-a-chip: recent breakthroughs and future prospects. Biomed Eng Online. 2020;19:1–19.

    Article  CAS  Google Scholar 

  212. Leung CM, De Haan P, Ronaldson-Bouchard K, Kim GA, Ko J, Rho HS, et al. A guide to the organ-on-a-chip. Nat Rev Methods Primers. 2022;2(1):33.

    Article  CAS  Google Scholar 

  213. Piergiovanni M, Leite SB, Corvi R, Whelan M. Standardisation needs for organ on chip devices. Lab Chip. 2021;21(15):2857–68.

    Article  CAS  PubMed  Google Scholar 

  214. Sima F, Sugioka K, Vázquez RM, Osellame R, Kelemen L, Ormos P. Three-dimensional femtosecond laser processing for lab-on-a-chip applications. Nanophotonics. 2018;7(3):613–34.

    Article  CAS  Google Scholar 

  215. Paoli R, Di Giuseppe D, Badiola-Mateos M, Martinelli E, Lopez-Martinez MJ, Samitier J. Rapid manufacturing of multilayered microfluidic devices for organ on a chip applications. Sensors. 2021;21(4):1382.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Ko J, Park D, Lee S, Gumuscu B, Jeon NL. Engineering organ-on-a-chip to accelerate translational research. Micromachines (Basel). 2022;13(8):1200.

    Article  PubMed  Google Scholar 

  217. Puryear JR III, Yoon JK, Kim Y. Advanced fabrication techniques of microengineered physiological systems. Micromachines (Basel). 2020;11(8):730.

    Article  Google Scholar 

  218. Kim J, Yoon T, Kim P, Bekhbat M, Kang SM, Rho HS, et al. Manufactured tissue-to-tissue barrier chip for modeling the human blood–brain barrier and regulation of cellular trafficking. Lab Chip. 2023;23:2990–3001.

    Article  CAS  PubMed  Google Scholar 

  219. Convery N, Samardzhieva I, Stormonth-Darling JM, Harrison S, Sullivan GJ, Gadegaard N. 3D printed tooling for injection molded microfluidics. Macromol Mater Eng. 2021;306(11):2100464.

    Article  CAS  Google Scholar 

  220. Kane KIW, Jarazo J, Moreno EL, Fleming RMT, Schwamborn JC. Passive controlled flow for Parkinson’s disease neuronal cell culture in 3D microfluidic devices. Organs-on-a-Chip. 2020;2: 100005.

    Article  Google Scholar 

  221. Zhou Y, Qiao H, Xu F, Zhao W, Wang J, Gu L, et al. Bioengineering of a human physiologically relevant microfluidic blood–cerebrospinal fluid barrier model. Lab Chip. 2023;23:3002–15.

    Article  CAS  PubMed  Google Scholar 

  222. Brown JA, Codreanu SG, Shi M, Sherrod SD, Markov DA, Neely MD, et al. Metabolic consequences of inflammatory disruption of the blood-brain barrier in an organ-on-chip model of the human neurovascular unit. J Neuroinflammation. 2016;13(1):1–17.

    Article  Google Scholar 

  223. Salmon I, Grebenyuk S, Fattah ARA, Rustandi G, Pilkington T, Verfaillie C, et al. Engineering neurovascular organoids with 3D printed microfluidic chips. Lab Chip. 2022;22(8):1615–29.

    Article  CAS  PubMed  Google Scholar 

  224. Kang YJ, Diep YN, Tran M, Tran VTA, Ambrin G, Ngo H, et al. Three-dimensional human neural culture on a chip recapitulating neuroinflammation and neurodegeneration. Nat Protocols. 2023;18(9):2838–67.

    Article  CAS  PubMed  Google Scholar 

  225. Park J, Wetzel I, Marriott I, Dréau D, D’Avanzo C, Kim DY, et al. A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat Neurosci. 2018;21(7):941–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Park J, Baik SH, Mook-Jung I, Irimia D, Cho H. Mimicry of central-peripheral immunity in Alzheimer’s disease and discovery of neurodegenerative roles in neutrophil. Front Immunol. 2019;10:480449.

    Article  Google Scholar 

  227. Seo S, Choi CH, Yi KS, Kim SU, Lee K, Choi N, et al. An engineered neurovascular unit for modeling neuroinflammation. Biofabrication. 2021;13(3): 035039.

    Article  CAS  Google Scholar 

  228. Seo S, Jang M, Kim H, Sung JH, Choi N, Lee K, et al. Neuro-glia-vascular-on-a-chip system to assess aggravated neurodegeneration via brain endothelial cells upon exposure to diesel exhaust particles. Adv Funct Mater. 2023;33(12):2210123.

    Article  CAS  Google Scholar 

  229. Jorfi M, D’Avanzo C, Kim DY, Irimia D. Three-dimensional models of the human brain development and diseases. Adv Healthc Mater. 2018;7(1).

  230. Jorfi M, D’avanzo C, Tanzi RE, Kim DY, Irimia D. Human neurospheroid arrays for in vitro studies of Alzheimer’s disease. Sci Rep. 2018;8(1):2450.

    Article  PubMed  PubMed Central  Google Scholar 

  231. Fernandes JTS, Chutna O, Chu V, Conde JP, Outeiro TF. A novel microfluidic cell co-culture platform for the study of the molecular mechanisms of Parkinson’s disease and other synucleinopathies. Front Neurosci. 2016;10:227609.

    Article  Google Scholar 

  232. de Rus JA, Alpaugh M, Denis HL, Tancredi JL, Boutin M, Decaestecker J, et al. The contribution of inflammatory astrocytes to BBB impairments in a brain-chip model of Parkinson’s disease. Nat Commun. 2023;14(1):3651.

    Article  Google Scholar 

  233. Pediaditakis I, Kodella KR, Manatakis DV, Hinojosa CD, Manolakos ES, Rubin LL, et al. Modeling Alpha-synuclein pathology in a human brain-chip to assess blood-brain barrier disruption in Parkinson’s disease. BioRxiv. 2021;2020–7.

Download references

Acknowledgements

This collaboration was supported by the Stem Cell Network and the Medical Research Council through the “MRC-SCN UK–Canada Regenerative Medicine Exchange Program Awards” (MR/X503125/1). We also thank Nottingham Trent University and the University of Victoria for hosting the research exchange.

Funding

The project was funded by “MRC-SCN UK-Canada Regenerative Medicine Exchange Program Awards” (MR/X503125/1).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: WB, RS, JJE, SMW, and YR; project administration: WB, YR; writing original draft: WB, RS, VAS, BCB, AC, VK, FK, MVH, MET, and JJE; figures design: VAS. All authors reviewed and approved of the manuscript.

Corresponding authors

Correspondence to Stephanie M. Willerth or Yvonne Reinwald.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors have no competing financial or non-financial interests to disclose.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Balestri, W., Sharma, R., da Silva, V.A. et al. Modeling the neuroimmune system in Alzheimer’s and Parkinson’s diseases. J Neuroinflammation 21, 32 (2024). https://0-doi-org.brum.beds.ac.uk/10.1186/s12974-024-03024-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12974-024-03024-8

Keywords