Next Article in Journal
Dynamic Neuro-Glial-Vascular Responses in a Mouse Model of Vascular Cognitive Impairment
Previous Article in Journal
Characterizing Secondary and Atypical Parkinsonisms: Defining Features and Clinical Variability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neuroglia in Neurodegeneration: Exploring Glial Dynamics in Brain Disorders

1
CNB-Salk Institute of Biological Studies, La Jolla, San Diego, CA 92037, USA
2
Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA
3
School of Studies in Neuroscience, Jiwaji University, Gwalior 474001, India
4
Faculty of Life Sciences, Department of Zoology, Aligarh Muslim University, Aligarh 202001, India
*
Author to whom correspondence should be addressed.
Neuroglia 2024, 5(4), 488-504; https://doi.org/10.3390/neuroglia5040031
Submission received: 23 October 2024 / Revised: 22 November 2024 / Accepted: 27 November 2024 / Published: 5 December 2024

Abstract

:
Neurodegenerative diseases represent a significant global health burden, characterized by progressive loss of neuronal function and structure. While traditionally viewed as primarily neuronal disorders, recent research has highlighted the crucial roles of neuroglia-astrocytes, microglia, and oligodendrocytes in the pathogenesis and progression of these diseases. This review explores the dual nature of glial cells in neurodegenerative processes, focusing on their protective and potentially harmful functions in Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and other neurodegenerative disorders. We examine the complex interactions between different glial cell types and neurons, highlighting recent discoveries in glial-neuronal metabolic coupling, neuroinflammation, and protein aggregation. Advanced technologies, such as single-cell RNA sequencing and spatial transcriptomics, have revealed unprecedented glial heterogeneity and disease-specific glial states, reshaping our understanding of these cells’ roles in health and disease. The review also discusses emerging concepts in neuroglial research, including the role of extracellular vesicles in disease propagation, epigenetic regulation of glial function, and the application of artificial intelligence in glial biology. Finally, we explore the therapeutic implications of targeting glia in neurodegenerative diseases, addressing both the promising avenues and challenges in developing glial-focused interventions. By integrating recent advances in neuroglial research, this review provides a comprehensive overview of the field and highlights future directions for research and therapeutic development. Understanding the complex roles of neuroglia in neurodegenerative diseases is crucial for developing more effective treatments and ultimately improving patient outcomes.

1. Introduction

Neurodegenerative diseases represent a group of disorders characterized by the progressive degeneration of the nervous system, posing significant challenges to global health. Conditions such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple sclerosis (MS) have complex pathophysiologies that extend beyond neuronal dysfunction, implicating various cellular and molecular mechanisms [1]. Recent research has shifted focus towards neuroglia—the non-neuronal cells of the central nervous system (CNS)—which play critical roles in the initiation and progression of these disorders. Among neuroglial cells, astrocytes have emerged as essential regulators of neuronal health and CNS function. These star-shaped cells, the most abundant glial cells in the CNS, are integral to maintaining homeostasis, providing metabolic support to neurons, and regulating neurotransmitter levels [2]. However, in the context of neurodegenerative diseases, astrocytes demonstrate a duality in function. They can adopt both neuroprotective and reactive states, the latter potentially contributing to disease progression. This dual behavior complicates our understanding of astrocytes’ roles in neurodegeneration [3]. Similarly, microglia, the resident immune cells of the CNS, exhibit a spectrum of activation states that play a pivotal role in disease outcomes. Microglia are vital for responding to neuroinflammation and clearing cellular debris, essential for maintaining CNS health. However, chronic microglial activation can result in neurotoxic effects, thus contributing to the pathogenesis of neurodegenerative diseases such as AD and PD [4]. Another key player is the oligodendrocyte, responsible for myelin production within the CNS. Dysfunction in oligodendrocytes leads to demyelination, which is a hallmark of MS and is also implicated in the progression of other neurodegenerative disorders [5]. We conducted a narrative review to provide an integrative overview of the complex roles of neuroglial cells across various neurodegenerative diseases. The literature search was performed across databases including PubMed, Google Scholar, Web of Science, and Scopus. We used keywords such as “neuroglia”, “neurodegeneration”, “astrocytes”, “microglia”, “oligodendrocytes”, “Alzheimer’s disease”, “Parkinson’s disease”, and “multiple sclerosis” to identify relevant studies. We included original research articles and reviews published in English, mostly over the last decade (2013–2023), that focused on the roles of glial cells in neurodegenerative diseases. Studies not directly addressing glial cells or neurodegenerative conditions, case reports, and non-peer-reviewed articles were excluded. This approach allowed us to consolidate findings from diverse sources and provide a comprehensive perspective on the dual roles of glial cells, with particular attention to recent technological advances such as single-cell RNA sequencing, which have greatly enhanced our understanding of glial heterogeneity. Given the diverse roles of these neuroglial cells, it is crucial to understand their dual contributions to both neuroprotection and neurodegeneration. This review will explore three key aspects: (1) the neuroprotective and inflammatory roles of astrocytes, (2) the impact of microglial activation states in AD and PD, and (3) the consequences of oligodendrocyte dysfunction in MS. By elucidating these complex interactions, we aim to identify potential therapeutic targets for mitigating the effects of these debilitating diseases, thereby paving the way for novel treatment strategies.

2. Astrocytes: Neuroprotectors or Neuroinflammatory Agents?

Astrocytes, the most abundant glial cells in the CNS, play a pivotal role in maintaining neuronal health and function. However, in the context of neurodegenerative diseases, astrocytes exhibit a complex dual nature, acting as both neuroprotectors and potential contributors to disease progression. This duality has been the subject of intense research in recent years, revealing a nuanced understanding of astrocytic function in pathological conditions. In their neuroprotective capacity, astrocytes provide essential support to neurons through various mechanisms. They regulate extracellular ion concentrations, particularly potassium, which is crucial for maintaining proper neuronal excitability [6]. Astrocytes also play a vital role in neurotransmitter uptake and recycling, particularly for glutamate, preventing excitotoxicity and supporting synaptic function [7]. Furthermore, these cells contribute to metabolic support by providing energy substrates to neurons through the Astrocyte-neuron lactate shuttle (ANLS) [8].
In the context of AD, astrocytes have been shown to participate in the clearance of Amyloid-β (Aβ) through various mechanisms, including enzymatic degradation and phagocytosis [9]. A recent study by Iram et al. (2020) demonstrated that astrocytes can internalize and degrade tau seeds, potentially limiting the spread of tau pathology in tauopathies such asAD [10]. However, astrocytes can also adopt reactive states that may contribute to neuroinflammation and exacerbate neurodegenerative processes. Liddelow et al. (2017) identified two distinct types of reactive astrocytes: A1 astrocytes, which are neurotoxic, and A2 astrocytes, which are potentially neuroprotective [11]. A1 astrocytes, induced by activated microglia, lose many normal astrocytic functions and gain neurotoxic properties, including the ability to rapidly kill neurons and oligodendrocytes. In PD, reactive astrocytes have been implicated in both neuroprotection and neurotoxicity. While some studies have shown that astrocytes can protect dopaminergic neurons through the release of glial cell line-derived neurotrophic factor (GDNF) [12], others have demonstrated that astrocytic activation can contribute to neuroinflammation and oxidative stress, exacerbating dopaminergic neuron loss [13].
Recent advances in single-cell RNA sequencing have revealed unprecedented heterogeneity among astrocytes in neurodegenerative diseases. Habib et al. (2020) identified multiple astrocyte states in AD mouse models and AD human brains, including a disease-associated astrocyte (DAA) population with a distinct transcriptional signature [14]. Similarly, Wheeler et al. (2020) described region-specific astrocyte subtypes in the healthy and the diseased human brain, highlighting the complexity of astrocytic responses in different CNS regions and disease contexts [15]. Understanding the molecular mechanisms underlying the transition between neuroprotective and neurotoxic astrocyte states is crucial for developing targeted therapeutic approaches. Recent work by Guttenplan et al. (2021) demonstrated that blocking the formation of A1 astrocytes through genetic or pharmacological means could prevent neurodegeneration in mouse models of PD and ALS, highlighting the potential of astrocyte-targeted therapies [16]. While the dual nature of astrocytes in neurodegenerative diseases presents both challenges and opportunities for therapeutic interventions, future research should focus on elucidating the factors that determine astrocyte phenotypes in different disease contexts and developing strategies to promote neuroprotective astrocytic functions while mitigating their neurotoxic potential (Figure 1).

3. Microglia: The Brain’s Immune Cells at a Crossroads

Microglia, the resident immune cells of the CNS, play a crucial role in maintaining brain homeostasis and responding to pathological conditions. In neurodegenerative diseases, microglia exhibit a complex array of activation states that can either contribute to neuroprotection or exacerbate neurodegeneration. This section will focus on the diverse roles of microglia in AD and PD, highlighting recent advances in our understanding of microglial function in these disorders.
In AD, microglia have been implicated in both protective and detrimental roles. On the protective side, microglia are capable of phagocytosing and degrading Aβ plaques, a hallmark of AD pathology [17]. Recent studies have identified a subset of disease-associated microglia (DAM) that cluster around Aβ plaques and express genes associated with phagocytosis and lipid metabolism [18]. These DAM are thought to play a protective role by containing Aβ spread and limiting its neurotoxic effects. However, chronic microglial activation in AD can also contribute to neuroinflammation and neurodegeneration. Prolonged exposure to Aβ can lead to a dysfunctional microglial phenotype characterized by reduced phagocytic capacity and increased production of pro-inflammatory cytokines [19]. This inflammatory state can further damage neurons and exacerbate AD pathology. The role of TREM2 (triggering receptor expressed on myeloid cells 2) in microglial function has gained significant attention in AD research. TREM2 is a microglial surface receptor that regulates phagocytosis and inflammatory responses. Variants in the TREM2 gene have been associated with increased AD risk, highlighting the importance of microglial function in disease progression [20]. Recent work by Zhou et al. (2020) used single-nucleus RNA sequencing to identify TREM2-dependent and TREM2-independent microglial responses in AD, revealing complex microglial activation patterns that go beyond simple pro- and anti-inflammatory states [21]. In PD, microglial activation is a key feature of the neuroinflammatory process accompanying dopaminergic neuron loss. Positron emission tomography (PET) imaging studies have demonstrated increased microglial activation in the brains of PD patients, correlating with disease severity [22]. While acute microglial activation can be neuroprotective through the clearance of cellular debris and release of neurotrophic factors, chronic activation can lead to sustained production of pro-inflammatory mediators and reactive oxygen species, contributing to dopaminergic neuron death [23].
Recent research has shed light on the role of α-synuclein, the primary component of Lewy bodies in PD in microglial activation. Aggregated α-synuclein can activate microglia through Toll-like receptors (TLRs) and other pattern recognition receptors, leading to the production of pro-inflammatory cytokines and oxidative stress [24]. Interestingly, microglia have also been implicated in the spread of α-synuclein pathology through the release of exosomes containing misfolded α-synuclein [25]. The concept of microglial priming has gained attention in both AD and PD research. Primed microglia, which have been exposed to an initial inflammatory stimulus, exhibit an exaggerated response to subsequent stimuli. This heightened reactivity can contribute to chronic neuroinflammation and disease progression [26]. Factors such as aging, systemic inflammation, and genetic predisposition can contribute to microglial priming, potentially explaining the increased susceptibility to neurodegenerative diseases with age. Recent technological advances, particularly in single-cell RNA sequencing, have revealed unprecedented heterogeneity in microglial populations in neurodegenerative diseases. Masuda et al. (2019) identified disease-specific microglial subpopulations in AD, PD, and MS, challenging the traditional M1/M2 polarization model and suggesting a more nuanced understanding of microglial activation states [27]. Microglia play complex and multifaceted roles in AD and PD, with the potential to both protect against and contribute to neurodegeneration. Understanding the factors that regulate microglial activation states and developing strategies to promote beneficial microglial functions while mitigating harmful ones represent promising avenues for therapeutic intervention in neurodegenerative diseases.

4. Oligodendrocytes: Myelination Maestros in Multiple Sclerosis

Oligodendrocytes are specialized glial cells responsible for producing myelin in the CNS. Their primary function is to form and maintain the myelin sheath, which is crucial for rapid saltatory conduction of action potentials along axons. In MS, a chronic inflammatory demyelinating disease of the CNS, oligodendrocyte dysfunction plays a central role in the pathogenesis and progression of the disease. MS is characterized by the formation of demyelinating lesions, which result from the immune-mediated destruction of myelin and oligodendrocytes. The loss of myelin not only impairs axonal conduction but also leaves axons vulnerable to degeneration [28]. While the initial stages of MS are marked by inflammation and demyelination, the later stages are characterized by chronic neurodegeneration and failure of remyelination, leading to progressive disability [29]. Recent research has shed light on the complex roles of oligodendrocytes and their precursor cells (OPCs) in MS pathology. OPCs are present in the adult CNS and can differentiate into mature oligodendrocytes to replace those lost during disease progression. However, in MS, this process is often impaired, leading to incomplete or failed remyelination [30].
Several factors contribute to the dysfunction of oligodendrocytes and OPCs in MS:
Inflammation: The inflammatory environment in MS lesions can inhibit OPC differentiation and oligodendrocyte maturation. Pro-inflammatory cytokines such as TNF-α and IFN-γ have been shown to impair OPC differentiation and induce oligodendrocyte apoptosis [31].
  • Oxidative stress: Increased levels of reactive oxygen species in MS lesions can damage oligodendrocytes and myelin. Oligodendrocytes are particularly vulnerable to oxidative stress due to their high metabolic rate and iron content [32].
  • Epigenetic changes: Recent studies have identified epigenetic alterations in oligodendrocytes from MS patients, which may contribute to their dysfunction. For example, Moyon et al. (2017) demonstrated that changes in histone deacetylase activity can impair OPC differentiation and remyelination [33].
  • Altered signaling pathways: Dysregulation of signaling pathways important for oligodendrocyte differentiation and myelination, such as the Wnt and Notch pathways, has been observed in MS lesions [34].
Despite these challenges, there is growing evidence that promoting remyelination could be a viable therapeutic strategy in MS. Several approaches are being investigated:
  • Enhancing OPC differentiation: Compounds that promote OPC differentiation, such as clemastine and bazedoxifene, have shown promise in preclinical studies and early clinical trials [35,36].
  • Modulating the immune response: Therapies that target specific aspects of the immune response, such as anti-CD20 antibodies, have shown efficacy in reducing inflammation and potentially promoting a more favorable environment for remyelination [37].
  • Neuroprotective strategies: Approaches aimed at protecting oligodendrocytes and axons from damage, such as antioxidants or mitochondrial stabilizers, are being explored [38].
  • Cell replacement therapies: While still in its early stages, the transplantation of OPCs or other stem cells to promote remyelination is an area of active research [39].
Recent technological advances, particularly in single-cell RNA sequencing, have revealed previously unrecognized heterogeneity among oligodendrocyte lineage cells in MS. Falcão et al. (2018) identified distinct populations of OPCs and oligodendrocytes with varying degrees of maturation in MS lesions, providing new insights into the dynamics of oligodendrocyte differentiation and remyelination in the disease context [40]. Oligodendrocyte dysfunction is a central feature of MS pathology, contributing to both the initial demyelination and the failure of remyelination in chronic disease. Understanding the complex interplay between oligodendrocytes, the immune system, and the CNS microenvironment is crucial for developing effective therapies to promote remyelination and halt disease progression in MS.

5. Glial-Glial Interactions in Neurodegenerative Processes

The complex interplay between different glial cell types is increasingly recognized as a crucial factor in the progression of neurodegenerative diseases. While individual glial cell types have been extensively studied, recent research has highlighted the importance of glial-glial interactions in shaping the neuroinflammatory environment and influencing disease outcomes. This section will focus on the intricate communication between astrocytes, microglia, and oligodendrocytes in the context of neurodegenerative processes.

6. Astrocyte-Microglial Interactions

Astrocytes and microglia engage in a highly dynamic interplay that orchestrates neuroinflammatory responses and neuronal health. In Alzheimer’s disease (AD), activated microglia release IL-1α, TNF, and C1q, which convert astrocytes into a neurotoxic A1 phenotype. A1 astrocytes lose their homeostatic functions, such as synaptic support and antioxidant defense, while gaining neurotoxic properties that induce neuronal death [11]. Emerging research highlights the complexity of this interaction. For instance, astrocyte-derived IL-33 has been implicated in the regulation of microglial phagocytic activity, particularly in synaptic pruning during development. Dysregulation of this pathway in neurodegeneration may contribute to aberrant synaptic loss and cognitive decline [14,41]. Similarly, Yun et al. (2018) demonstrated that the astrocytic release of complement protein C3 activates microglia, promoting neuroinflammation in tauopathies, highlighting how specific signaling molecules mediate these interactions [42]. Astrocytes can also act as modulators of microglial states. Factors such as TGF-β and IL-10 secreted by astrocytes have been shown to promote a neuroprotective microglial phenotype, dampening neuroinflammation and potentially slowing disease progression [41,43]. These findings suggest that targeting astrocyte-microglial communication could restore balance to the neuroimmune environment.

7. Oligodendrocyte-Microglial Interactions

The interaction between oligodendrocytes and microglia is crucial in both demyelinating diseases, such asmultiple sclerosis (MS), and neurodegenerative diseases with myelin disruption, such as Parkinson’s disease (PD) and AD. Activated microglia release pro-inflammatory cytokines, including IL-6 and TNF-α, and reactive oxygen species, which induce oligodendrocyte apoptosis and impair myelination [44]. Conversely, microglia can facilitate remyelination by clearing myelin debris and secreting growth factors such as activin-A, which enhances oligodendrocyte precursor cell (OPC) differentiation [45,46]. Microglia displaying a pro-regenerative phenotype further support remyelination through the secretion of insulin-like growth factor-1 (IGF-1) and brain-derived neurotrophic factor (BDNF), which promote OPC survival and differentiation [47]. However, chronic microglial activation, such as that observed in aging or prolonged neuroinflammation, results in sustained production of inflammatory mediators, impairing their ability to clear myelin debris effectively and to provide a supportive environment for remyelination [48]. Oligodendrocytes also play an active role in modulating microglial activation through the release of exosomes. These oligodendrocyte-derived exosomes contain bioactive molecules, including heat-shock proteins and myelin-associated proteins, which influence microglial states. They can reduce inflammatory responses in microglia and promote a shift towards a repair-associated phenotype [49]. The term “bidirectional”, in this context, refers to the dynamic, reciprocal communication between microglia and oligodendrocytes. On the one hand, microglia influence oligodendrocyte survival, differentiation, and myelin repair through their secretory profiles, which can vary from pro-inflammatory to regenerative, depending on their activation state. On the other hand, oligodendrocytes, through mechanisms such as exosome release, actively regulate microglial activation and function, thus shaping the neuroinflammatory environment. These interactions highlight their potential as therapeutic targets in diseases characterized by demyelination, with strategies aiming to restore the balance between pro-repair and pro-inflammatory states in both cell types.

8. Astrocyte-Oligodendrocyte Interactions

Astrocytes play a multifaceted role in supporting oligodendrocyte function, particularly in myelination and remyelination. Reactive astrocytes, however, display dual roles depending on the disease context and the specific factors they secrete. For instance, astrocyte-derived ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor (LIF) have been shown to promote OPC differentiation and remyelination [50]. In contrast, endothelin-1, another astrocyte-derived factor, inhibits OPC maturation, thereby impairing remyelination [51]. Recent studies highlight the metabolic coupling between astrocytes and oligodendrocytes. Astrocytes supply lactate to oligodendrocytes as an energy source during myelination. Dysregulation of astrocytic metabolic pathways in diseases such asMS can deprive oligodendrocytes of this crucial support, exacerbating myelin loss [52]. Additionally, astrocytes expressing the transcription factor MAFG in MS lesions promote chronic inflammation, further disrupting the remyelination process [15]. These interactions are also influenced by the extracellular matrix (ECM). For example, astrocyte-driven changes in ECM composition can affect OPC migration and differentiation, either facilitating or hindering remyelination [53]. Therapeutic strategies targeting astrocyte metabolism and ECM remodeling may offer novel approaches to enhancing remyelination.

9. Tripartite Glial Interactions

The interaction among astrocytes, microglia, and oligodendrocytes forms a complex and dynamic network that plays a pivotal role in shaping the progression of neurodegenerative diseases. In Alzheimer’s disease (AD), amyloid-β (Aβ) accumulation activates microglia, which in turn induces neurotoxic astrocytes. These activated astrocytes impair oligodendrocyte function and myelination, creating a self-perpetuating cycle of neuroinflammation and neuronal damage [11,54]. Recent advances, particularly through single-cell and single-nucleus RNA sequencing studies, have provided detailed maps of glial interactions within the AD brain, identifying specific molecular pathways that could be targeted to slow or halt disease progression [55,56]. The nature of these interactions is multifaceted and bidirectional. For example, astrocytes play a key role in amplifying microglial activation through the release of pro-inflammatory molecules such as S100B. This amplification not only increases neuroinflammation but also affects oligodendrocyte survival and the integrity of myelin, further propagating the disease cycle [57]. In this context, astrocytes do not simply act as bystanders; they actively influence the inflammatory response by modifying microglial phenotypes. This can result in either pro-inflammatory or neuroprotective responses, depending on the cytokines and other factors released by astrocytes. On the other hand, microglia also shape the astrocyte response. Under certain conditions, microglia can drive astrocytes toward a more neurotoxic phenotype by secreting inflammatory mediators such as IL-6 and TNF-α. However, when microglia adopt a pro-regenerative phenotype, they release factors such as insulin-like growth factor-1 (IGF-1) and brain-derived neurotrophic factor (BDNF), which help support oligodendrocyte precursor cell (OPC) survival and differentiation, facilitating remyelination [47]. Moreover, oligodendrocytes themselves are active participants in regulating microglial activation. Recent studies have shown that oligodendrocyte-derived exosomes, which contain heat-shock proteins and myelin-associated proteins, can modulate microglial states, often reducing inflammation and promoting repair mechanisms. These interactions underscore the bidirectional nature of the glial network, where each cell type not only responds to signals but also sends feedback that can either promote or inhibit disease progression. Understanding these intricate and context-dependent glial-glial interactions is essential for developing targeted therapies aimed at restoring glial homeostasis and mitigating neurodegeneration. Future research should focus on the temporal and spatial dynamics of these interactions—particularly how changes in metabolic, inflammatory, and structural factors at different stages of disease contribute to the overall neurodegenerative process [58] (Figure 2).

10. Neuroglia in Other Neurodegenerative Diseases

While AD, PD, and MS have been extensively studied in terms of glial involvement, other neurodegenerative disorders also show significant glial contributions to their pathogenesis. This section explores the roles of neuroglia in amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), and frontotemporal dementia (FTD). In ALS, characterized by progressive motor neuron degeneration, glial cells play crucial roles. Astrocytes adopt a reactive phenotype that can be both neuroprotective and neurotoxic. A1 astrocytes, induced by activated microglia, are abundant in ALS and contribute to motor neuron death [11]. However, some astrocyte populations may provide neuroprotection through glutamate uptake and neurotrophic factor release [59]. Microglial activation, a hallmark of ALS pathology, can release pro-inflammatory factors damaging motor neurons, but may also play protective roles in early disease stages [60]. Recent single-cell RNA sequencing has identified disease-specific microglial subtypes in ALS, revealing potential therapeutic targets [61]. Oligodendrocyte dysfunction and demyelination are also observed in ALS, with OPCs failing to mature properly, potentially contributing to motor neuron vulnerability [62]. HD, a genetic disorder causing progressive brain damage, shows significant glial cell contributions to its pathology. Reactive astrocytes in HD exhibit impaired glutamate uptake, potentially contributing to excitotoxicity [63]. However, astrocyte-mediated release of BDNF could be neuroprotective in HD models [64]. Chronic microglial activation in HD contributes to neuroinflammation, with mutant huntingtin expression in microglia enhancing pro-inflammatory responses and exacerbating neuronal damage [65]. Oligodendrocytes in HD show impaired maturation and myelination capacity due to mutant huntingtin expression, potentially contributing to neuronal dysfunction [66]. In FTD, characterized by progressive neuronal loss in the frontal and temporal lobes, glial involvement has gained increasing attention. Reactive astrocytes are prominent in FTD and may contribute to neurodegeneration through various mechanisms. Astrocytes in FTD patients with C9orf72 mutations exhibit altered gene expression profiles, potentially affecting their supportive functions [63]. Microglial activation is observed in FTD and may contribute to disease progression. Progranulin deficiency, a common cause of FTD, leads to lysosomal dysfunction in microglia and exacerbates neuroinflammation [67]. White matter pathology is increasingly recognized in FTD, with oligodendroglial inclusions and myelin abnormalities suggesting a potential role for oligodendrocyte dysfunction in the disease process [68]. These findings highlight the complex and multifaceted roles of neuroglia in various neurodegenerative diseases, emphasizing the need for further research to develop targeted therapeutic strategies addressing glial dysfunction in these disorders.

11. Emerging Themes

Across various neurodegenerative diseases, several key themes highlight the involvement of glial cells. First, the idea of diverse glial activation states is now widely recognized, moving beyond the simple notion of “activated” or “resting” glial phenotypes [69]. Additionally, disruptions in the metabolic support that glia provide to neurons are observed in multiple disorders, which may increase neuronal vulnerability [70]. Another significant factor is the accumulation of disease-related proteins, such as superoxide dismutase 1 (SOD1) in ALS and huntingtin in HD, within glial cells. This protein aggregation can impair glial function and drive disease progression [71]. Emerging evidence also suggests that glial cells may contribute to the spread of protein aggregates, further exacerbating neurodegenerative pathology [72]. Recent studies have brought attention to the dual role of extracellular vesicles (EVs) in neurodegeneration, where they can both exacerbate and mitigate disease progression. The mechanisms underlying this duality are complex and context-dependent. On one hand, EVs can exacerbate disease by promoting inflammation, protein aggregation, and glial dysfunction. For instance, in Alzheimer’s disease (AD), EVs carrying amyloid-β (Aβ) can transport these toxic proteins between cells, facilitating the spread of neurodegenerative pathology. This process is further amplified by microglial activation induced by the inflammatory content of EVs, such as pro-inflammatory cytokines and complement proteins. These factors can create a vicious cycle of inflammation and neurodegeneration, particularly in diseases such as AD and Parkinson’s disease (PD). On the other hand, EVs can also mitigate disease by promoting repair processes, such as clearing toxic aggregates and supporting tissue regeneration. EVs containing neurotrophic factors such asbrain-derived neurotrophic factor (BDNF) and insulin-like growth factor 1 (IGF-1) can facilitate neuroprotection by promoting neuronal survival, differentiation, and synaptic plasticity. Additionally, oligodendrocyte-derived EVs may aid remyelination in demyelinating diseases such asmultiple sclerosis (MS) by delivering proteins that support oligodendrocyte precursor cell differentiation and myelin repair. This protective effect is often observed when EVs are involved in reparative microglial responses, where microglia shift toward a neuroprotective phenotype in response to specific EV cargo. Thus, the dual role of EVs in neurodegeneration is regulated by factors such as EV content, the state of glial activation, and the disease context. While pro-inflammatory EVs can fuel neurodegeneration, neuroprotective EVs offer potential therapeutic benefits. Understanding the specific pathways that drive these opposing effects could lead to the development of targeted therapeutic strategies aimed at manipulating EV function to promote repair and reduce disease progression. A deeper understanding of these complex roles of glia and EVs across neurodegenerative diseases is essential for developing therapies aimed at modulating glial function to promote neuroprotection and slow disease progression.

12. Emerging Concepts in Neuroglial Research

Recent technological advancements and novel research approaches have led to several emerging concepts in the field of neuroglial research. These new insights are reshaping our understanding of glial biology and its roles in neurodegenerative diseases. This section will explore five key emerging concepts: glial-neuronal metabolic coupling, single-cell technologies revealing glial heterogeneity, extracellular vesicles (EVs) and protein aggregation, glial epigenetics, and artificial intelligence (AI) in glial research.
A.
Glial-Neuronal Metabolic Coupling
The concept of glial-neuronal metabolic coupling has gained substantial recognition in recent years, highlighting the intricate relationship between glial cells and neurons. Rather than serving merely as support cells, glia actively participate in maintaining neuronal function. One notable example is the ANLS, where astrocytes supply lactate to neurons as an energy source during periods of heightened activity. This process, detailed by Magistretti and Allaman (2018), highlights the critical role astrocytes play in sustaining neuronal energy demands [8]. Similarly, research by Saab et al. (2016) has revealed that oligodendrocytes, traditionally seen as myelin producers, also provide direct metabolic support to axons through the transport of lactate via monocarboxylate transporters [73]. This discovery reshapes the view of oligodendrocytes as more than just insulators. Additionally, microglia contribute to brain energy regulation. Bernier et al. (2020) demonstrated that microglia modulate both neuronal activity and astrocytic glycolysis, thereby maintaining energy homeostasis in the brain [74].
B.
Single-Cell Technologies Revealing Glial Heterogeneity
Advanced single-cell technologies have revolutionized our understanding of glial cell diversity, revealing unprecedented heterogeneity among glial populations. Single-cell RNA sequencing, as demonstrated by studies from Mathys et al. (2019) and Zhou et al. (2020), has been instrumental in identifying distinct glial subtypes, such as DAM and astrocytes in AD [21,75]. These findings have deepened our understanding of the specialized roles glial cells play in neurodegeneration. Spatial transcriptomics, employed by Chen et al. (2020), has mapped gene expression in glial cells in relation to specific brain regions, providing insights into region-specific functions in both healthy and diseased states [76]. Furthermore, multiomics approaches by Mrdjen et al. (2018) have combined single-cell proteomics and transcriptomics to characterize diverse myeloid populations within the CNS [77]. These multi-layered insights have unveiled new glial subsets with potential implications in neurodegenerative disease mechanisms Figure 3.
C.
Extracellular Vesicles (EVs) and Protein Aggregation
EVs have emerged as crucial mediators of intercellular communication, particularly in the context of neurodegenerative diseases. These vesicles are now understood to play a role in the spread of pathogenic proteins, contributing to disease progression. Research by Asai et al. (2015) revealed that microglia-derived EVs can promote the propagation of tau pathology in AD models, suggesting a harmful role in spreading neurotoxic proteins [78]. Conversely, Men et al. (2019) demonstrated that astrocyte-derived EVs possess potential neuroprotective effects, transferring beneficial proteins to neurons and possibly mitigating neuronal damage [79]. Furthermore, oligodendrocyte-derived EVs have been implicated in myelin maintenance and axonal support, as shown by Frühbeis et al. (2020) [80]. These findings point to a dual role of EVs in both exacerbating and mitigating disease processes, depending on the glial cell type and the context.
D.
Glial Epigenetics
The role of epigenetic regulation in glial cells has gained increasing attention, particularly for its implications in neurodegenerative diseases. Microglial epigenetics, reviewed by Cheray and Joseph (2018), has highlighted how epigenetic mechanisms govern microglial activation and memory, influencing their response to inflammation and injury [81]. Understanding these mechanisms could lead to therapeutic interventions aimed at modulating neuroinflammation. In the context of AD, astrocyte epigenetics have also been studied, with Neal and Richardson (2018) demonstrating that epigenetic changes in astrocytes contribute to their reactive phenotypes [82]. This epigenetic regulation influences the astrocytes’ ability to respond to injury and inflammation. Similarly, oligodendrocyte epigenetics, as reviewed by Emery and Lu (2015), plays a critical role in oligodendrocyte differentiation and myelination [83]. Epigenetic mechanisms may also be crucial for developing therapies aimed at promoting remyelination in demyelinating disorders.
E.
Artificial Intelligence in Glial Research
The integration of AI into glial research is opening new frontiers for understanding and manipulating glial function. AI-assisted image analysis, as demonstrated by Heiland et al. (2019), enables high-throughput analysis of microglial morphology, allowing researchers to assess glial activation states more efficiently and accurately [84]. This technology facilitates the study of glial responses on a large scale, which is essential for understanding their roles in neurodegeneration. Additionally, predictive modeling of glial responses using machine learning, as shown by Zeisel et al. (2018), allows for the prediction of glial cell types and states based on single-cell RNA sequencing data [85]. This has provided new insights into glial heterogeneity and the identification of disease-associated subtypes. AI is also transforming the landscape of drug discovery. AI-driven approaches to drug discovery have been applied to identify novel compounds targeting specific glial cell types, accelerating the development of therapies for neurodegenerative diseases (Figure 3) [86]. These emerging concepts and technologies are reshaping our understanding of glial biology and their critical roles in neurodegenerative diseases. As we integrate these insights into future research, we move closer to developing more effective therapies that target glial function and improve outcomes for those suffering from these devastating conditions.
Figure 3. New Discoveries and Future Directions in Neuroglial Research. This multi-part figure presents the latest advances in neuroglial research. Panel (A) shows how single-cell RNA sequencing reveals different types of glial cells. Panel (B) highlights the use of spatial mapping to study glial gene activity in brain tissue. Panel (C) shows how AI is being used to discover drugs targeting glial cells. Panel (D) illustrates how epigenetic changes regulate glial activity. The figure ends with a diagram of personalized therapies, combining genetic data to create tailored treatments for neurodegenerative diseases.
Figure 3. New Discoveries and Future Directions in Neuroglial Research. This multi-part figure presents the latest advances in neuroglial research. Panel (A) shows how single-cell RNA sequencing reveals different types of glial cells. Panel (B) highlights the use of spatial mapping to study glial gene activity in brain tissue. Panel (C) shows how AI is being used to discover drugs targeting glial cells. Panel (D) illustrates how epigenetic changes regulate glial activity. The figure ends with a diagram of personalized therapies, combining genetic data to create tailored treatments for neurodegenerative diseases.
Neuroglia 05 00031 g003

13. Therapeutic Implications

The growing understanding of neuroglia in neurodegenerative diseases has created new possibilities for therapeutic intervention, particularly by targeting glial cells. This section delves into various strategies for addressing specific glial populations, the challenges in developing glia-focused therapies, and future directions for research and drug development.
Targeting specific glial populations has emerged as a promising approach to combating neurodegenerative diseases. Astrocyte-targeted therapies focus on modulating astrocyte reactivity and enhancing their neuroprotective roles. Research by Liddelow et al. (2017) suggests that blocking the formation of neurotoxic A1 astrocytes could protect neurons [11], while targeting pathways such as NF-κB or the complement system could prevent harmful astrocyte activation [16]. Another promising avenue involves enhancing astrocytic glutamate uptake to reduce excitotoxicity, a process in which excess glutamate damages neurons. The β-lactam antibiotic ceftriaxone has been shown to upregulate the glutamate transporter GLT-1, offering potential therapeutic benefits, as highlighted by Rothstein et al. (2005) [59].
Microglial-targeted therapies focus on modulating microglial activation and reducing pro-inflammatory signaling. Shifting microglial activity toward a more protective phenotype could mitigate neuroinflammation, with anti-TREM2 antibodies, such as the one developed by Schlepckow et al. (2020), showing promise in enhancing microglial function in AD models [87]. Additionally, inhibiting pro-inflammatory pathways in microglia, such as the p38 MAPK pathway, has shown potential in preclinical studies, as evidenced by Gannon et al. (2019) [88]. Oligodendrocyte-targeted therapies are also gaining attention, particularly in the context of remyelination and oligodendrocyte protection. Compounds such as clemastine, which promotes OPC differentiation, have shown potential in MS trials [35]. Protecting oligodendrocytes from oxidative stress and mitochondrial dysfunction may also play a key role in preventing demyelination, as discussed by Rao et al. (2021) [89].
Modulating glial-glial and glial-neuronal interactions represents another innovative therapeutic strategy. Enhancing metabolic coupling between neurons and glial cells, such as through the ANLS or by improving oligodendrocyte-axon support, could provide neuroprotection, according to Magistretti and Allaman (2018) [8]. This approach aims to maintain energy balance in the brain, protecting neurons from metabolic stress. Another emerging area of interest is the modulation of glial EVs, which play a role in both the propagation of disease-related proteins and the transport of neuroprotective molecules. Therapies designed to inhibit pathogenic EV release or enhance the production of protective EVs could influence disease progression, as noted by You and Ikezu (2019) [90]. Additionally, targeting neuroinflammatory signaling cascades shared by various glial cell types could provide a more comprehensive approach to reducing neuroinflammation. Hammond et al. (2019) emphasized the potential of therapies that inhibit shared inflammatory pathways in slowing disease progression [91]. As we continue to explore these therapeutic strategies, the dynamic role of glial cells in neurodegenerative diseases offers hope for more effective treatments. However, translating these preclinical findings into clinical therapies remains a challenge, underscoring the need for continued research and development of glial-focused interventions.

14. Challenges and Future Directions

Despite significant progress in developing glial-targeted therapies for neurodegenerative diseases, several key challenges need to be addressed to enhance their effectiveness. One major challenge is improving glial cell-type specificity in treatments. With growing evidence of the distinct roles that astrocytes, microglia, and oligodendrocytes play in neurodegenerative diseases, it is clear that therapies must target specific subpopulations rather than broad glial categories. This will help minimize off-target effects and ensure that interventions act only on the intended cell types, thereby improving therapeutic outcomes.
Another critical hurdle is determining the optimal therapeutic window for glial-targeted interventions. Timing plays a pivotal role in the success of these therapies, as glial cells can have both beneficial and harmful effects, depending on the stage of the disease. For instance, in the early stages of diseases such as Alzheimer’s, glial activation may support neuronal survival and repair, but if this activation persists, it can lead to chronic inflammation, contributing to neurodegeneration. Therapies will need to balance these opposing effects, with a focus on determining when glial activation shifts from neuroprotective to neurotoxic. This requires a precise understanding of the temporal dynamics of glial activation, with a need for ongoing research into how these processes unfold across different stages of disease progression.
Combination therapies are another promising approach to managing neurodegenerative diseases. Given the multifactorial nature of these disorders, targeting multiple glial subtypes or combining glial-focused interventions with traditional neuron-targeted therapies could provide synergistic effects, improving both disease management and patient outcomes. For instance, combining therapies that modulate both microglial activity and oligodendrocyte function may not only reduce neuroinflammation but also promote tissue repair and regeneration. Clinical strategies should therefore focus on integrating glial-targeted therapies with existing approaches, creating a more comprehensive and effective treatment regimen that can address the diverse pathogenic mechanisms at play.
Furthermore, developing reliable biomarkers to indicate specific glial activation states is crucial for improving patient selection and monitoring treatment responses. Biomarkers that can track changes in glial activity, such as inflammatory markers or myelin integrity, would enable clinicians to identify the most appropriate therapies for individual patients and provide real-time insights into treatment efficacy.
The integration of AI and machine learning technologies also holds promise for accelerating the development of personalized glial-targeted therapies. By harnessing large datasets, these technologies can help identify novel therapeutic targets, predict patient responses to treatments, and streamline drug design processes. AI-driven models could predict which therapies are most likely to succeed based on individual patients’ genetic and disease profiles, allowing for more precise, tailored interventions.
Looking ahead, personalized medicine will be a key pillar of future glial-targeted therapies. Combining genetic, transcriptomic, and proteomic data will allow for the development of highly specific treatments that are tailored to the unique disease mechanisms of individual patients. Understanding how glial activation profiles differ from one patient to another and how these differences influence disease progression will be essential in designing treatments that maximize therapeutic benefit.
In conclusion, while advances in understanding glial biology in neurodegenerative diseases have laid a strong foundation, numerous challenges remain. Future research should focus on refining the specificity of glial-targeted therapies, optimizing the timing of interventions, integrating combination strategies, and developing reliable biomarkers. By addressing these challenges and leveraging emerging technologies, there is great potential to develop more effective, personalized therapies that can slow or even reverse the progression of these debilitating diseases.

Author Contributions

N.J.D. and S.A.B.: conceptualization, methodology, U.J. and J.A.B.: literature search, writing the original manuscript draft, and illustration preparation; N.J.D. and S.A.B.: editing, reviewing, and finalizing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AD–Alzheimer’s disease; AI—Artificial intelligence; ALS—Amyotrophic lateral sclerosis; ANLS—Astrocyte-neuron lactate shuttle; —Amyloid-β; CNS—Central nervous system; DAM—Disease-associated microglia; EVs—Extracellular vesicles; FTD—Frontotemporal dementia; HD—Huntington’s disease; MS—Multiple sclerosis; OPCs—Oligodendrocyte precursor cells; PD—Parkinson’s disease; TREM2—Triggering receptor expressed on myeloid cells 2.

References

  1. Dugger, B.N.; Dickson, D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef]
  2. Sofroniew, M.V.; Vinters, H.V. Astrocytes: Biology and pathology. Acta Neuropathol. 2010, 119, 7–35. [Google Scholar] [CrossRef] [PubMed]
  3. Liddelow, S.A.; Barres, B.A. Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity 2017, 46, 957–967. [Google Scholar] [CrossRef] [PubMed]
  4. Salter, M.W.; Stevens, B. Microglia emerge as central players in brain disease. Nat. Med. 2017, 23, 1018–1027. [Google Scholar] [CrossRef] [PubMed]
  5. Bankston, A.N.; Mandler, M.D.; Feng, Y. Oligodendroglia and neurotrophic factors in neurodegeneration. Neurosci. Bull. 2019, 35, 459–468. [Google Scholar] [CrossRef]
  6. Verkhratsky, A.; Nedergaard, M. Physiology of astroglia. Physiol. Rev. 2018, 98, 239–389. [Google Scholar] [CrossRef]
  7. Schousboe, A.; Bak, L.K.; Waagepetersen, H.S. Astrocytic control of biosynthesis and turnover of the neurotransmitters glutamate and GABA. Front. Endocrinol. 2013, 4, 102. [Google Scholar] [CrossRef]
  8. Magistretti, P.J.; Allaman, I. Lactate in the brain: From metabolic end-product to signalling molecule. Nat. Rev. Neurosci. 2018, 19, 235–249. [Google Scholar] [CrossRef]
  9. Ries, M.; Sastre, M. Mechanisms of Aβ clearance and degradation by glial cells. Front. Aging Neurosci. 2016, 8, 160. [Google Scholar] [CrossRef]
  10. Iram, T.; Ramirez-Ortiz, Z.; Byrne, M.H.; Coleman, U.A.; Kingery, N.D.; Means, T.K.; Frenkel, D.; El Khoury, J. Megf10 is a receptor for C1Q that mediates clearance of apoptotic cells by astrocytes. J. Neurosci. 2016, 36, 5185–5192. [Google Scholar] [CrossRef]
  11. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
  12. Rocha, S.M.; Cristovão, A.C.; Campos, F.L.; Fonseca, C.P.; Baltazar, G. Astrocyte-derived GDNF is a potent inhibitor of microglial activation. Neurobiol. Dis. 2012, 47, 407–415. [Google Scholar] [CrossRef] [PubMed]
  13. Booth, H.D.E.; Hirst, W.D.; Wade-Martins, R. The role of astrocyte dysfunction in Parkinson’s disease pathogenesis. Trends Neurosci. 2017, 40, 358–370. [Google Scholar] [CrossRef] [PubMed]
  14. Habib, N.; McCabe, C.; Medina, S.; Varshavsky, M.; Kitsberg, D.; Dvir-Szternfeld, R.; Green, G.; Dionne, D.; Nguyen, L.; Marshall, J.L. Disease-associated astrocytes in Alzheimer’s disease and aging. Nat. Neurosci. 2020, 23, 701–706. [Google Scholar] [CrossRef]
  15. Wheeler, M.A.; Clark, I.C.; Tjon, E.C.; Li, Z.; Zandee, S.E.J.; Couturier, C.P.; Watson, B.R.; Scalisi, G.; Alkwai, S.; Rothhammer, V. MAFG-driven astrocytes promote CNS inflammation. Nature 2020, 578, 593–599. [Google Scholar] [CrossRef]
  16. Guttenplan, K.A.; Weigel, M.K.; Prakash, P.; Wijewardhane, P.R.; Hasel, P.; Rufen-Blanchette, U.; Münch, A.E.; Blum, J.A.; Fine, J.; Neal, M.C. Neurotoxic reactive astrocytes induce cell death via saturated lipids. Nature 2021, 599, 102–107. [Google Scholar] [CrossRef]
  17. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef]
  18. Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 2017, 169, 1276–1290.e17. [Google Scholar] [CrossRef]
  19. Hickman, S.; Izzy, S.; Sen, P.; Morsett, L.; El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 2018, 21, 1359–1369. [Google Scholar] [CrossRef]
  20. Ulland, T.K.; Colonna, M. TREM2—A key player in microglial biology and Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 667–675. [Google Scholar] [CrossRef]
  21. Zhou, Y.; Song, W.M.; Andhey, P.S.; Swain, A.; Levy, T.; Miller, K.R.; Poliani, P.L.; Cominelli, M.; Grover, S.; Gilfillan, S. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nat. Med. 2020, 26, 131–142. [Google Scholar] [CrossRef] [PubMed]
  22. Ouchi, Y.; Yoshikawa, E.; Sekine, Y.; Futatsubashi, M.; Kanno, T.; Ogusu, T.; Torizuka, T. Microglial activation and dopamine terminal loss in early Parkinson’s disease. Ann. Neurol. 2005, 57, 168–175. [Google Scholar] [CrossRef] [PubMed]
  23. Subramaniam, S.R.; Federoff, H.J. Targeting microglial activation states as a therapeutic avenue in Parkinson’s disease. Front. Aging Neurosci. 2017, 9, 176. [Google Scholar] [CrossRef] [PubMed]
  24. Grozdanov, V.; Bousset, L.; Hoffmeister, M.; Bliederhaeuser, C.; Meier, C.; Madiona, K.; Pieri, L.; Kiechle, M.; McLean, P.J.; Kassubek, J. Increased immune activation by pathologic α-synuclein in Parkinson’s disease. Ann. Neurol. 2019, 86, 593–606. [Google Scholar] [CrossRef]
  25. Xia, Y.; Zhang, G.; Han, C.; Ma, K.; Guo, X.; Wan, F.; Kou, L.; Yin, S.; Liu, L.; Huang, J. Microglia as modulators of exosomal alpha-synuclein transmission. Cell Death Dis. 2019, 10, 174. [Google Scholar] [CrossRef]
  26. Perry, V.H.; Holmes, C. Microglial priming in neurodegenerative disease. Nat. Rev. Neurol. 2014, 10, 217–224. [Google Scholar] [CrossRef]
  27. Masuda, T.; Sankowski, R.; Staszewski, O.; Böttcher, C.; Amann, L.; Sagar, N.; Scheiwe, C.; Nessler, S.; Kunz, P.; van Loo, G. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature 2019, 566, 388–392. [Google Scholar] [CrossRef]
  28. Trapp, B.D.; Nave, K.-A. Multiple sclerosis: An immune or neurodegenerative disorder? Annu. Rev. Neurosci. 2008, 31, 247–269. [Google Scholar] [CrossRef]
  29. Lubetzki, C.; Zalc, B.; Williams, A.; Stadelmann, C.; Stankoff, B. Remyelination in multiple sclerosis: From basic science to clinical translation. Lancet Neurol. 2020, 19, 678–688. [Google Scholar] [CrossRef]
  30. Franklin, R.J.M.; Ffrench-Constant, C. Regenerating CNS myelin—From mechanisms to experimental medicines. Nat. Rev. Neurosci. 2017, 18, 753–769. [Google Scholar] [CrossRef]
  31. Cui, L.-Y.; Chu, S.-F.; Chen, N.-H. The role of chemokines and chemokine receptors in multiple sclerosis. Int. Immunopharmacol. 2020, 83, 106314. [Google Scholar] [CrossRef] [PubMed]
  32. Lassmann, H.; van Horssen, J. Oxidative stress and its impact on neurons and glia in multiple sclerosis lesions. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2016, 1862, 506–510. [Google Scholar] [CrossRef] [PubMed]
  33. Moyon, S.; Huynh, J.L.; Dutta, D.; Zhang, F.; Ma, D.; Yoo, S.; Lawrence, R.; Wegner, M.; John, G.R.; Emery, B. Functional characterization of DNA methylation in the oligodendrocyte lineage. Cell Rep. 2016, 15, 748–760. [Google Scholar] [CrossRef] [PubMed]
  34. Fancy, S.P.J.; Baranzini, S.E.; Zhao, C.; Yuk, D.-I.; Irvine, K.-A.; Kaing, S.; Sanai, N.; Franklin, R.J.M.; Rowitch, D.H. Dysregulation of the Wnt pathway inhibits timely myelination and remyelination in the mammalian CNS. Genes Dev. 2009, 23, 1571–1585. [Google Scholar] [CrossRef]
  35. Green, A.J.; Gelfand, J.M.; Cree, B.A.; Bevan, C.; Boscardin, W.J.; Mei, F.; Inman, J.; Arnow, S.; Devereux, M.; Abounasr, A. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): A randomised, controlled, double-blind, crossover trial. Lancet 2017, 390, 2481–2489. [Google Scholar] [CrossRef]
  36. Rankin, K.A.; Mei, F.; Kim, K.; Shen, Y.-A.A.; Mayoral, S.R.; Desponts, C.; Lorrain, D.S.; Green, A.J.; Baranzini, S.E.; Chan, J.R. Selective estrogen receptor modulators enhance CNS remyelination independent of estrogen receptors. J. Neurosci. 2019, 39, 2184–2194. [Google Scholar] [CrossRef]
  37. Bar-Or, A.; Calkwood, J.C.; Chognot, C.; Evershed, J.; Fox, E.J.; Herman, A.; Manfrini, M.; McNamara, J.; Robertson, D.S.; Stokmaier, D. Effect of ocrelizumab on vaccine responses in patients with multiple sclerosis: The VELOCE study. Neurology 2020, 95, e1999–e2008. [Google Scholar] [CrossRef]
  38. Mahad, D.H.; Trapp, B.D.; Lassmann, H. Pathological mechanisms in progressive multiple sclerosis. Lancet Neurol. 2015, 14, 183–193. [Google Scholar] [CrossRef]
  39. Goldman, S.A.; Nedergaard, M.; Windrem, M.S. Glial progenitor cell–based treatment and modeling of neurological disease. Science 2012, 338, 491–495. [Google Scholar] [CrossRef]
  40. Falcão, A.M.; van Bruggen, D.; Marques, S.; Meijer, M.; Jäkel, S.; Agirre, E.; Samudyata, N.; Floriddia, E.M.; Vanichkina, D.P.; Ffrench-Constant, C. Disease-specific oligodendrocyte lineage cells arise in multiple sclerosis. Nat. Med. 2018, 24, 1837–1844. [Google Scholar] [CrossRef]
  41. Jha, M.K.; Jo, M.; Kim, J.H.; Suk, K. Microglia-Astrocyte Crosstalk: An Intimate Molecular Conversation. Neurosci. 2019, 25, 227–240. [Google Scholar] [CrossRef] [PubMed]
  42. Yun, S.P.; Kam, T.I.; Panicker, N.; Kim, S.; Oh, Y.; Park, J.S.; Kwon, S.H.; Park, Y.J.; Karuppagounder, S.S.; Park, H.; et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nat. Med. 2018, 24, 931–938. [Google Scholar] [CrossRef] [PubMed]
  43. Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar] [CrossRef] [PubMed]
  44. Peferoen, L.; Kipp, M.; van der Valk, P.; van Noort, J.M.; Amor, S. Oligodendrocyte-microglia cross-talk in the central nervous system. Immunol. 2014, 141, 302–313. [Google Scholar] [CrossRef]
  45. Lloyd, A.F.; Miron, V.E. The pro-remyelination properties of microglia in the central nervous system. Nat. Rev. Neurol. 2019, 15, 447–458. [Google Scholar] [CrossRef]
  46. Hagemeyer, N.; Hanft, K.M.; Akriditou, M.A.; Unger, N.; Park, E.S.; Stanley, E.R.; Staszewski, O.; Dimou, L.; Prinz, M. Microglia contribute to normal myelinogenesis and to oligodendrocyte progenitor maintenance during adulthood. Acta Neuropathol. 2017, 134, 441–458. [Google Scholar] [CrossRef]
  47. Moyon, S.; Frawley, R.; Marechal, D.; Huang, D.; Marshall-Phelps, K.L.; Kegel, L.; Bøstrand, S.M.; Sadowski, B.; Jiang, Y.-H.; Lyons, D.A.; et al. TET1-mediated DNA hydroxymethylation regulates adult remyelination in mice. Nat. Commun. 2021, 12, 3359. [Google Scholar] [CrossRef]
  48. Gao, C.; Jiang, J.; Tan, Y.; Chen, S. Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 359. [Google Scholar] [CrossRef]
  49. Xiao, L.; Hareendran, S.; Loh, Y.P. Function of exosomes in neurological disorders and brain tumors. Extracell. Vesicles Circ. Nucleic Acids 2021, 2, 55. [Google Scholar] [CrossRef]
  50. Stankoff, B.; Aigrot, M.S.; Noël, F.; Wattilliaux, A.; Zalc, B.; Lubetzki, C. Ciliary neurotrophic factor (CNTF) enhances myelin formation: A novel role for CNTF and CNTF-related molecules. J. Neurosci. 2002, 22, 9221–9227. [Google Scholar] [CrossRef]
  51. Hammond, T.R.; Gadea, A.; Dupree, J.; Kerninon, C.; Nait-Oumesmar, B.; Aguirre, A.; Gallo, V. Astrocyte-derived endothelin-1 inhibits remyelination through notch activation. Neuron 2014, 81, 588–602. [Google Scholar] [CrossRef] [PubMed]
  52. Barateiro, A.; Brites, D.; Fernandes, A. Oligodendrocyte development and myelination in neurodevelopment: Molecular mechanisms in health and disease. Curr. Pharm. Des. 2016, 22, 656–679. [Google Scholar] [CrossRef] [PubMed]
  53. Sofroniew, M.V. Astrocyte reactivity: Subtypes, states, and functions in CNS innate immunity. Trends Immunol. 2020, 41, 758–770. [Google Scholar] [CrossRef] [PubMed]
  54. Desai, M.K.; Mastrangelo, M.A.; Ryan, D.A.; Sudol, K.L.; Narrow, W.C.; Bowers, W.J. Early oligodendrocyte/myelin pathology in Alzheimer’s disease mice constitutes a novel therapeutic target. Am. J. Pathol. 2010, 177, 1422–1435. [Google Scholar] [CrossRef]
  55. Mathys, H.; Boix, C.A.; Akay, L.A.; Xia, Z.; Davila-Velderrain, J.; Ng, A.P.; Jiang, X.; Abdelhady, G.; Galani, K.; Mantero, J.J.N. Single-cell multiregion dissection of Alzheimer’s disease. Nature 2024, 632, 858–868. [Google Scholar] [CrossRef]
  56. Grubman, A.; Chew, G.; Ouyang, J.F.; Sun, G.; Choo, X.Y.; McLean, C.; Simmons, R.K.; Buckberry, S.; Vargas-Landin, D.B.; Poppe, D.; et al. A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation. Nat. Neurosci. 2021, 24, 595–610. [Google Scholar] [CrossRef]
  57. López-Muguruza, E.; Matute, C. Alterations of oligodendrocyte and myelin energy metabolism in multiple sclerosis. Int. J. Mol. Sci. 2023, 24, 12912. [Google Scholar] [CrossRef]
  58. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of neuroinflammation in neurodegeneration development. Signal Transduct. Target. Ther. 2023, 8, 267. [Google Scholar] [CrossRef]
  59. Rothstein, J.D.; Patel, S.; Regan, M.R.; Haenggeli, C.; Huang, Y.H.; Bergles, D.E.; Jin, L.; Dykes Hoberg, M.; Vidensky, S.; Chung, D.S. β-Lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature 2005, 433, 73–77. [Google Scholar] [CrossRef]
  60. Geloso, M.C.; Corvino, V.; Marchese, E.; Serrano, A.; Michetti, F.; D’Ambrosi, N. The dual role of microglia in ALS: Mechanisms and therapeutic approaches. Front. Aging Neurosci. 2017, 9, 242. [Google Scholar] [CrossRef]
  61. Spiller, K.J.; Restrepo, C.R.; Khan, T.; Dominique, M.A.; Fang, T.C.; Canter, R.G.; Roberts, C.J.; Miller, K.R.; Ransohoff, R.M.; Trojanowski, J.Q. Microglia-mediated recovery from ALS-relevant motor neuron degeneration in a mouse model of TDP-43 proteinopathy. Nat. Neurosci. 2018, 21, 329–340. [Google Scholar] [CrossRef] [PubMed]
  62. Kang, S.H.; Li, Y.; Fukaya, M.; Lorenzini, I.; Cleveland, D.W.; Ostrow, L.W.; Rothstein, J.D.; Bergles, D.E. Degeneration and impaired regeneration of gray matter oligodendrocytes in amyotrophic lateral sclerosis. Nat. Neurosci. 2013, 16, 571–579. [Google Scholar] [CrossRef] [PubMed]
  63. Hallmann, A.-L.; Araúzo-Bravo, M.J.; Mavrommatis, L.; Ehrlich, M.; Röpke, A.; Brockhaus, J.; Missler, M.; Sterneckert, J.; Schöler, H.R.; Kuhlmann, T. Astrocyte pathology in a human neural stem cell model of frontotemporal dementia caused by mutant TAU protein. Sci. Rep. 2017, 7, 42991. [Google Scholar] [CrossRef] [PubMed]
  64. Benraiss, A.; Wang, S.; Herrlinger, S.; Li, X.; Chandler-Militello, D.; Mauceri, J.; Burm, H.B.; Toner, M.; Osipovitch, M.; Jim Xu, Q. Human glia can both induce and rescue aspects of disease phenotype in Huntington disease. Nat. Commun. 2016, 7, 11758. [Google Scholar] [CrossRef]
  65. Crotti, A.; Benner, C.; Kerman, B.E.; Gosselin, D.; Lagier-Tourenne, C.; Zuccato, C.; Cattaneo, E.; Gage, F.H.; Cleveland, D.W.; Glass, C.K. Mutant Huntingtin promotes autonomous microglia activation via myeloid lineage-determining factors. Nat. Neurosci. 2014, 17, 513–521. [Google Scholar] [CrossRef]
  66. Jin, J.; Peng, Q.; Hou, Z.; Jiang, M.; Wang, X.; Langseth, A.J.; Tao, M.; Barker, P.B.; Mori, S.; Bergles, D.E. Early white matter abnormalities, progressive brain pathology and motor deficits in a novel knock-in mouse model of Huntington’s disease. Hum. Mol. Genet. 2015, 24, 2508–2527. [Google Scholar] [CrossRef]
  67. Lui, H.; Zhang, J.; Makinson, S.R.; Cahill, M.K.; Kelley, K.W.; Huang, H.-Y.; Shang, Y.; Oldham, M.C.; Martens, L.H.; Gao, F. Progranulin deficiency promotes circuit-specific synaptic pruning by microglia via complement activation. Cell 2016, 165, 921–935. [Google Scholar] [CrossRef]
  68. Ferrer, I.; Andrés-Benito, P.; Zelaya, M.V.; Aguirre, M.E.E.; Carmona, M.; Ausín, K.; Lachén-Montes, M.; Fernández-Irigoyen, J.; Santamaría, E.; Del Rio, J.A. Familial globular glial tauopathy linked to MAPT mutations: Molecular neuropathology and seeding capacity of a prototypical mixed neuronal and glial tauopathy. Acta Neuropathol. 2020, 139, 735–771. [Google Scholar] [CrossRef]
  69. Ransohoff, R.M. A polarizing question: Do M1 and M2 microglia exist? Nat. Neurosci. 2016, 19, 987–991. [Google Scholar] [CrossRef]
  70. Barros, L.F.; Brown, A.; Swanson, R.A. Glia in brain energy metabolism: A perspective. Glia 2018, 66, 1134–1137. [Google Scholar] [CrossRef]
  71. Radford, R.A.; Morsch, M.; Rayner, S.L.; Cole, N.J.; Pountney, D.L.; Chung, R.S. The established and emerging roles of astrocytes and microglia in amyotrophic lateral sclerosis and frontotemporal dementia. Front. Cell. Neurosci. 2015, 9, 414. [Google Scholar] [CrossRef] [PubMed]
  72. Perea, J.R.; Llorens-Martín, M.; Ávila, J.; Bolós, M. The Role of Microglia in the Spread of Tau: Relevance for Tauopathies. Front. Cell. Neurosci. 2018, 12, 172. [Google Scholar] [CrossRef] [PubMed]
  73. Saab, A.S.; Tzvetavona, I.D.; Trevisiol, A.; Baltan, S.; Dibaj, P.; Kusch, K.; Möbius, W.; Goetze, B.; Jahn, H.M.; Huang, W. Oligodendroglial NMDA receptors regulate glucose import and axonal energy metabolism. Neuron 2016, 91, 119–132. [Google Scholar] [CrossRef] [PubMed]
  74. Bernier, L.-P.; York, E.M.; Kamyabi, A.; Choi, H.B.; Weilinger, N.L.; MacVicar, B.A. Microglial metabolic flexibility supports immune surveillance of the brain parenchyma. Nat. Commun. 2020, 11, 1559. [Google Scholar] [CrossRef]
  75. Mathys, H.; Davila-Velderrain, J.; Peng, Z.; Gao, F.; Mohammadi, S.; Young, J.Z.; Menon, M.; He, L.; Abdurrob, F.; Jiang, X. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 2019, 570, 332–337. [Google Scholar] [CrossRef]
  76. Chen, W.-T.; Lu, A.; Craessaerts, K.; Pavie, B.; Frigerio, C.S.; Corthout, N.; Qian, X.; Laláková, J.; Kühnemund, M.; Voytyuk, I. Spatial transcriptomics and in situ sequencing to study Alzheimer’s disease. Cell 2020, 182, 976–991.e19. [Google Scholar] [CrossRef]
  77. Mrdjen, D.; Pavlovic, A.; Hartmann, F.J.; Schreiner, B.; Utz, S.G.; Leung, B.P.; Lelios, I.; Heppner, F.L.; Kipnis, J.; Merkler, D. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity 2018, 48, 380–395.e6. [Google Scholar] [CrossRef]
  78. Asai, H.; Ikezu, S.; Tsunoda, S.; Medalla, M.; Luebke, J.; Haydar, T.; Wolozin, B.; Butovsky, O.; Kügler, S.; Ikezu, T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015, 18, 1584–1593. [Google Scholar] [CrossRef]
  79. Men, Y.; Yelick, J.; Jin, S.; Tian, Y.; Chiang, M.S.R.; Higashimori, H.; Brown, E.; Jarvis, R.; Yang, Y. Exosome reporter mice reveal the involvement of exosomes in mediating neuron to astroglia communication in the CNS. Nat. Commun. 2019, 10, 4136. [Google Scholar] [CrossRef]
  80. Frühbeis, C.; Kuo-Elsner, W.P.; Müller, C.; Barth, K.; Peris, L.; Tenzer, S.; Möbius, W.; Werner, H.B.; Nave, K.-A.; Fröhlich, D. Oligodendrocytes support axonal transport and maintenance via exosome secretion. PLoS Biol. 2020, 18, e3000621. [Google Scholar] [CrossRef]
  81. Cheray, M.; Joseph, B. Epigenetics control microglia plasticity. Front. Cell. Neurosci. 2018, 12, 243. [Google Scholar] [CrossRef] [PubMed]
  82. Neal, M.; Richardson, J.R. Epigenetic regulation of astrocyte function in neuroinflammation and neurodegeneration. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2017, 1864, 432–443. [Google Scholar] [CrossRef] [PubMed]
  83. Emery, B.; Lu, Q.R. Transcriptional and epigenetic regulation of oligodendrocyte development and myelination in the central nervous system. Cold Spring Harb. Perspect. Biol. 2015, 7, a020461. [Google Scholar] [CrossRef]
  84. Heiland, D.H.; Wörner, J.; Haaker, J.G.; Delev, D.; Pompe, N.; Mercas, B.; Franco, P.; Gäbelein, A.; Heynckes, S.; Pfeifer, D. The integrative metabolomic-transcriptomic landscape of glioblastome multiforme. Oncotarget 2017, 8, 49178. [Google Scholar] [CrossRef]
  85. Zeisel, A.; Hochgerner, H.; Lönnerberg, P.; Johnsson, A.; Memic, F.; Van Der Zwan, J.; Häring, M.; Braun, E.; Borm, L.E.; La Manno, G. Molecular architecture of the mouse nervous system. Cell 2018, 174, 999–1014.e22. [Google Scholar] [CrossRef]
  86. Qiu, Y.; Cheng, F. Artificial intelligence for drug discovery and development in Alzheimer’s disease. Curr. Opin. Struct. Biol. 2024, 85, 102776. [Google Scholar] [CrossRef]
  87. Schlepckow, K.; Monroe, K.M.; Kleinberger, G.; Cantuti-Castelvetri, L.; Parhizkar, S.; Xia, D.; Willem, M.; Werner, G.; Pettkus, N.; Brunner, B. Enhancing protective microglial activities with a dual function TREM 2 antibody to the stalk region. EMBO Mol. Med. 2020, 12, e11227. [Google Scholar] [CrossRef]
  88. Gannon, P.J.; Akay-Espinoza, C.; Yee, A.C.; Briand, L.A.; Erickson, M.A.; Gelman, B.B.; Gao, Y.; Haughey, N.J.; Zink, M.C.; Clements, J.E. HIV protease inhibitors alter amyloid precursor protein processing via β-site amyloid precursor protein cleaving enzyme-1 translational up-regulation. Am. J. Pathol. 2017, 187, 91–109. [Google Scholar] [CrossRef]
  89. Rao, V.T.S.; Fuh, S.-C.; Karamchandani, J.R.; Woulfe, J.M.J.; Munoz, D.G.; Ellezam, B.; Blain, M.; Ho, M.-K.; Bedell, B.J.; Antel, J.P. Astrocytes in the pathogenesis of multiple sclerosis: An in situ MicroRNA study. J. Neuropathol. Exp. Neurol. 2019, 78, 1130–1146. [Google Scholar] [CrossRef]
  90. You, Y.; Ikezu, T. Emerging roles of extracellular vesicles in neurodegenerative disorders. Neurobiol. Dis. 2019, 130, 104512. [Google Scholar] [CrossRef]
  91. Hammond, T.R.; Marsh, S.E.; Stevens, B. Immune signaling in neurodegeneration. Immunity 2019, 50, 955–974. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The Dual Role of Glial Cells in Neurodegenerative Diseases. This figure represents how astrocytes, microglia, and oligodendrocytes can play both protective and harmful roles in neurodegenerative diseases. On the left, glial cells are shown supporting neurons and protecting the brain. On the right, they contribute to damage and disease progression. Key processes such asA1/A2 astrocyte states, microglial activation, and oligodendrocytes myelination are highlighted. Arrows indicate how these functions affect neurons and the course of disease.
Figure 1. The Dual Role of Glial Cells in Neurodegenerative Diseases. This figure represents how astrocytes, microglia, and oligodendrocytes can play both protective and harmful roles in neurodegenerative diseases. On the left, glial cells are shown supporting neurons and protecting the brain. On the right, they contribute to damage and disease progression. Key processes such asA1/A2 astrocyte states, microglial activation, and oligodendrocytes myelination are highlighted. Arrows indicate how these functions affect neurons and the course of disease.
Neuroglia 05 00031 g001
Figure 2. Glial-Glial and Glial-Neuron Interactions in Neurodegeneration. This figure illustrates the interactions between different glial cells and neurons in neurodegenerative diseases. It highlights important communication signals, metabolites, and extracellular vesicles that mediate these interactions. Key features include how astrocytes provide energy to neurons, microglia convert astrocytes into harmful A1 types, oligodendrocytes support axons, and vesicles spread toxic proteins. These interactions are shown as interconnected processes that drive neurodegeneration.
Figure 2. Glial-Glial and Glial-Neuron Interactions in Neurodegeneration. This figure illustrates the interactions between different glial cells and neurons in neurodegenerative diseases. It highlights important communication signals, metabolites, and extracellular vesicles that mediate these interactions. Key features include how astrocytes provide energy to neurons, microglia convert astrocytes into harmful A1 types, oligodendrocytes support axons, and vesicles spread toxic proteins. These interactions are shown as interconnected processes that drive neurodegeneration.
Neuroglia 05 00031 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dar, N.J.; Bhat, J.A.; John, U.; Bhat, S.A. Neuroglia in Neurodegeneration: Exploring Glial Dynamics in Brain Disorders. Neuroglia 2024, 5, 488-504. https://doi.org/10.3390/neuroglia5040031

AMA Style

Dar NJ, Bhat JA, John U, Bhat SA. Neuroglia in Neurodegeneration: Exploring Glial Dynamics in Brain Disorders. Neuroglia. 2024; 5(4):488-504. https://doi.org/10.3390/neuroglia5040031

Chicago/Turabian Style

Dar, Nawab John, Javeed Ahmad Bhat, Urmilla John, and Shahnawaz Ali Bhat. 2024. "Neuroglia in Neurodegeneration: Exploring Glial Dynamics in Brain Disorders" Neuroglia 5, no. 4: 488-504. https://doi.org/10.3390/neuroglia5040031

APA Style

Dar, N. J., Bhat, J. A., John, U., & Bhat, S. A. (2024). Neuroglia in Neurodegeneration: Exploring Glial Dynamics in Brain Disorders. Neuroglia, 5(4), 488-504. https://doi.org/10.3390/neuroglia5040031

Article Metrics

Back to TopTop