Skip to content

This report is written by MaltSci based on the latest literature and research findings


What is the role of microglia in neurodegenerative diseases?

Abstract

Microglia, the resident immune cells of the central nervous system (CNS), are critical in maintaining homeostasis and responding to pathological changes in neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Recent research has revealed that microglia exhibit a dual nature, with the ability to adopt both protective and detrimental roles depending on their activation state. In healthy conditions, microglia contribute to synaptic plasticity, clearance of cellular debris, and support neuronal health. However, during neurodegeneration, they can become activated, leading to the release of pro-inflammatory cytokines and neurotoxic factors that exacerbate neuronal damage. This review systematically explores the complex biology of microglia, detailing their origin, development, and homeostatic functions, alongside their contributions to the pathogenesis of AD, PD, and MS. We examine the molecular pathways involved in microglial activation, highlighting the significance of neuroinflammation, cytokine release, and phagocytosis of protein aggregates. Furthermore, emerging therapeutic strategies aimed at modulating microglial activity to enhance neuroprotection while minimizing neurotoxicity are discussed. By synthesizing recent findings, this review aims to illuminate the intricate dynamics of microglial function and identify promising avenues for future research and therapeutic intervention in neurodegeneration.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Overview of Microglial Biology
    • 2.1 Origin and Development of Microglia
    • 2.2 Homeostatic Functions of Microglia
  • 3 Microglia in Neurodegenerative Diseases
    • 3.1 Role in Alzheimer's Disease
    • 3.2 Role in Parkinson's Disease
    • 3.3 Role in Multiple Sclerosis
  • 4 Mechanisms of Microglial Activation
    • 4.1 Neuroinflammation and Cytokine Release
    • 4.2 Phagocytosis and Clearance of Protein Aggregates
    • 4.3 Interaction with Neuronal Cells
  • 5 Therapeutic Implications
    • 5.1 Modulating Microglial Activity
    • 5.2 Potential Drug Targets
    • 5.3 Future Directions in Microglial Research
  • 6 Conclusion

1 Introduction

Microglia, the resident immune cells of the central nervous system (CNS), play a pivotal role in maintaining homeostasis and responding to pathological changes within the brain. As the primary defenders against injury and disease, these macrophage-like cells are integral to both neurodevelopment and neurodegeneration. In recent years, there has been a surge of interest in understanding the dual nature of microglia in neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). This growing body of research highlights not only the protective functions of microglia, including the clearance of cellular debris and promotion of neuronal survival, but also their potential to contribute to neuroinflammation and neuronal damage under pathological conditions [1][2].

The significance of studying microglia in the context of neurodegenerative diseases cannot be overstated. Neurodegenerative disorders are characterized by progressive neuronal loss and associated cognitive and motor dysfunctions, representing a substantial burden on healthcare systems worldwide. Understanding the role of microglia in these diseases is essential for developing targeted therapeutic strategies aimed at modulating their activity to either enhance neuroprotection or mitigate neurotoxicity [2][3]. Furthermore, elucidating the mechanisms underlying microglial activation states and their interactions with neuronal cells is critical for identifying potential biomarkers and therapeutic targets in neurodegeneration [4][5].

Current research indicates that microglia can adopt various activation states, ranging from pro-inflammatory (M1) to anti-inflammatory (M2) phenotypes, each associated with distinct functions in the CNS [1][6]. While M1 microglia are implicated in promoting neuroinflammation and exacerbating neuronal injury, M2 microglia are thought to facilitate tissue repair and neuroprotection [2]. This dichotomy underscores the complexity of microglial roles in neurodegenerative diseases, as their activation can be influenced by a multitude of factors, including the nature of the pathological stimulus and the microenvironment [3][7].

This review will systematically explore the multifaceted roles of microglia in neurodegenerative diseases, beginning with an overview of microglial biology, including their origin, development, and homeostatic functions. We will then delve into the specific contributions of microglia to the pathogenesis of AD, PD, and MS, highlighting the mechanisms through which they influence disease progression [2][4]. The review will further examine the molecular pathways involved in microglial activation, including neuroinflammation, cytokine release, and phagocytosis of protein aggregates [2][8].

In addition to discussing the detrimental aspects of microglial activation, we will also address emerging therapeutic strategies aimed at modulating microglial activity to enhance their protective functions while minimizing neurotoxic effects. Potential drug targets and innovative approaches, such as microglial replacement therapy and the use of small molecules to influence microglial activation states, will be evaluated [3][7].

By synthesizing recent findings from experimental models and clinical studies, this review aims to provide a comprehensive overview of the complex interplay between microglia and neurodegenerative diseases. In doing so, we hope to illuminate the intricate dynamics of microglial function and identify promising avenues for future research and therapeutic intervention in the realm of neurodegeneration.

2 Overview of Microglial Biology

2.1 Origin and Development of Microglia

Microglia are the resident immune cells of the central nervous system (CNS) and play critical roles in both physiological and pathological conditions, particularly in neurodegenerative diseases. They originate from early erythromyeloid progenitors in the yolk sac during embryonic development and migrate into the developing brain, where they contribute to the establishment of a fully mature blood-brain barrier [9].

In their healthy state, microglia are involved in maintaining CNS homeostasis, regulating synaptic plasticity, and supporting neuronal health. They perform essential functions such as the clearance of cellular debris, dead neurons, and redundant synapses, which is crucial for normal brain development and function [10]. Microglia exhibit a unique homeostatic phenotype, which is tightly regulated by the microenvironment of the CNS [10].

However, under pathological conditions, microglia undergo activation and morphological changes, transitioning from a surveillant state to an activated phenotype. This activation can be triggered by various stimuli, including neuronal injury, neuroinflammation, and the presence of pathological protein aggregates [11]; [2]. The activated microglia can exhibit dual roles: they can adopt a neuroprotective phenotype that aids in tissue repair and regeneration, or a neurotoxic phenotype that exacerbates neurodegeneration [11]; [1].

In neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), microglial activation is a common feature. Evidence suggests that activated microglia secrete pro-inflammatory cytokines and neurotoxic factors, contributing to neuronal death and the progression of these diseases [12]; [13]. Conversely, microglia can also engage in protective functions, such as phagocytosing harmful aggregates and supporting neuronal survival [8].

Recent advances in single-cell technologies have revealed significant heterogeneity among microglia in terms of their states and functions during neurodegeneration. Some microglial states correlate with specific pathological hallmarks, indicating that their roles are not strictly beneficial or detrimental but rather context-dependent [2]. This complexity highlights the need for a nuanced understanding of microglial biology in the context of neurodegenerative diseases.

Moreover, the metabolic reprogramming of microglia has been identified as a critical factor influencing their function during neurodegeneration. Alterations in metabolic pathways can affect microglial activation states, further complicating their role in disease progression [14].

In summary, microglia are pivotal in the pathogenesis of neurodegenerative diseases, serving both protective and harmful roles depending on their activation state and the surrounding environment. Understanding the intricate biology of microglia, including their origin, development, and functional plasticity, is essential for developing effective therapeutic strategies targeting these cells in neurodegenerative disorders [10]; [9].

2.2 Homeostatic Functions of Microglia

Microglia are the resident immune cells of the central nervous system (CNS) and play a crucial role in maintaining homeostasis and responding to pathological changes. In healthy brains, microglia contribute to tissue homeostasis, synaptic plasticity, and the regulation of neural networks. They are involved in various essential functions such as the clearance of cellular debris, the elimination of redundant synapses during development, and the response to injuries or infections by adopting an activated state [10][12].

Under physiological conditions, microglia continuously survey the CNS environment through their dynamic processes, ensuring proper neuronal function and health. They are integral to neurogenesis and the maintenance of synaptic integrity, supporting neuronal proliferation and connectivity [9]. Microglia also modulate inflammation and repair mechanisms, balancing pro-inflammatory and anti-inflammatory responses to maintain CNS homeostasis [1].

However, during neurodegenerative diseases, microglia undergo significant changes in their morphology and function, often leading to a dysregulated inflammatory response. This transition is characterized by the activation of microglia, which can become polarized into distinct phenotypes. The M1 phenotype is associated with pro-inflammatory cytokine production and neurotoxicity, while the M2 phenotype is linked to anti-inflammatory effects and tissue repair [12][15].

In neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS), microglial activation is observed, and their homeostatic functions can be compromised. Activated microglia secrete various pro-inflammatory cytokines and neurotoxic factors that exacerbate neuronal damage and contribute to disease progression [2][11]. For instance, in AD, microglia can phagocytose synaptic structures, leading to synapse loss, which correlates with cognitive decline [16].

Moreover, microglia exhibit high spatial and temporal heterogeneity, and specific states of microglia correlate with pathological hallmarks of neurodegenerative diseases [8]. They not only play a protective role by clearing pathological proteins but can also contribute to neuroinflammation and neuronal degeneration if their activation becomes excessive [2][14].

The duality of microglial function—acting as both protectors and potential mediators of neurodegeneration—highlights their complex biology and the need for a nuanced understanding of their role in CNS health and disease. Research into the modulation of microglial activity is ongoing, with the aim of developing therapeutic strategies that can enhance their protective functions while mitigating their harmful effects in neurodegenerative diseases [15][17].

3 Microglia in Neurodegenerative Diseases

3.1 Role in Alzheimer's Disease

Microglia, the resident immune cells of the central nervous system (CNS), play multifaceted roles in neurodegenerative diseases, particularly Alzheimer's disease (AD). Their functions can be broadly categorized into protective and detrimental effects, which significantly influence the progression of neurodegeneration.

In Alzheimer's disease, microglia are known to be involved in the pathological processes associated with the disease, including the clearance of amyloid-beta (Aβ) plaques and the management of neuroinflammation. Evidence suggests that microglia can exhibit a neuroprotective role by phagocytosing Aβ and other debris, thereby contributing to the maintenance of brain homeostasis [18]. However, the activation of microglia can also lead to the release of pro-inflammatory cytokines and neurotoxic factors, exacerbating neuronal damage and contributing to disease progression [19].

Research has shown that activated microglia are present in close association with Aβ plaques, where they demonstrate a pattern of activation that correlates with the severity of neurodegeneration. For instance, microglia exhibit a dynamic response characterized by changes in their numbers, activation states, and cytokine expression during the progression of AD [20]. Furthermore, microglial activation has been implicated in synapse loss, a critical aspect of cognitive decline in AD [16]. Studies indicate that microglia can directly mediate synaptic degeneration, a process that may occur independently of amyloid pathology, highlighting their central role in neurodegeneration [16].

Moreover, microglia are not only involved in the response to Aβ accumulation but also participate in the regulation of tau pathology and neuronal loss. They are implicated in processes such as tau phosphorylation and the disintegration of synaptic structures, which are key features of Alzheimer's pathology [18]. This dual role of microglia, acting both as protectors and aggressors, complicates their overall impact on neuronal health and disease progression.

Recent studies have further explored the concept of microglial dysfunction in the context of aging and AD. Aging appears to alter microglial function, leading to a state of chronic neuroinflammation that exacerbates neurodegenerative processes [21]. This shift from a protective to a detrimental phenotype in aging microglia underscores the importance of understanding microglial biology in developing therapeutic strategies aimed at modulating their activity in AD.

Furthermore, genetic studies have identified polymorphisms in genes such as TREM2, APOE, and CD33 that significantly influence microglial function and their response to neurodegenerative processes. These findings suggest that microglial activity could serve as a therapeutic target, potentially allowing for interventions that enhance their beneficial functions while mitigating their harmful effects [19].

In summary, microglia are pivotal in the pathogenesis of Alzheimer's disease, where they can both protect against and contribute to neurodegeneration. Understanding the nuanced roles of microglia in AD pathology is essential for developing effective therapeutic approaches aimed at restoring microglial function and improving clinical outcomes in patients with neurodegenerative diseases.

3.2 Role in Parkinson's Disease

Microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in the pathogenesis of neurodegenerative diseases, particularly in Parkinson's disease (PD). These cells exhibit a complex dual nature, capable of both protective and detrimental functions depending on their activation state and the surrounding microenvironment.

In the context of Parkinson's disease, microglia are activated in response to dopaminergic neuronal degeneration in the substantia nigra pars compacta. This activation is characterized by the release of pro-inflammatory mediators such as cytokines, nitric oxide, and reactive oxygen species, which can exacerbate neuroinflammation and neuronal damage (Gao et al. 2023; Isik et al. 2023). Sustained neuroinflammation, primarily mediated by microglia, is recognized as a significant contributing factor to the progression of PD (Trainor et al. 2024).

Recent studies have shown that microglia can adopt different phenotypes during the disease process. The M1 phenotype is pro-inflammatory and associated with the release of neurotoxic factors, while the M2 phenotype is anti-inflammatory and linked to neuroprotection (Isik et al. 2023). In PD, a persistent M1 activation is observed, leading to a cycle of inflammation that further damages dopaminergic neurons (Gao et al. 2023). Furthermore, microglia have been implicated in the propagation of α-synuclein aggregates, a hallmark of PD, which can further drive neuroinflammation and neuronal death (Zhao et al. 2020).

Microglia also play a role in the phagocytosis of misfolded proteins and cellular debris. Interestingly, while monomeric α-synuclein enhances microglial phagocytosis, aggregated forms of this protein can inhibit this process, indicating that the state of α-synuclein significantly influences microglial function (Park et al. 2008). This dynamic underscores the importance of microglial activation states in the progression of PD.

Moreover, microglia's response to neuronal injury is not static; they exhibit temporal and spatial heterogeneity. For instance, in the early stages of degeneration, microglia may initially adopt a protective role, but as neurodegeneration progresses, they can shift towards a more neurotoxic phenotype (Ayerra et al. 2024). This transition is critical, as it reflects the changing landscape of neuroinflammation in PD.

The infiltration of peripheral immune cells also modifies microglial function, pushing them towards a pro-inflammatory state that can accelerate disease progression (Gao et al. 2023). Consequently, targeting microglial activation and their inflammatory responses presents a promising therapeutic strategy for managing Parkinson's disease and potentially other neurodegenerative disorders (Yu et al. 2023).

In summary, microglia serve as key players in the neuroinflammatory processes associated with Parkinson's disease. Their activation states, influenced by various pathological stimuli, dictate their role in either promoting neuroprotection or exacerbating neurodegeneration. Understanding these mechanisms is vital for developing targeted therapies aimed at modulating microglial activity to mitigate the progression of neurodegenerative diseases.

3.3 Role in Multiple Sclerosis

Microglia, the resident immune cells of the central nervous system (CNS), play a complex and dual role in neurodegenerative diseases, particularly in the context of multiple sclerosis (MS). Their involvement is characterized by both neurotoxic and neuroprotective functions, which can vary depending on their activation state and the surrounding microenvironment.

In multiple sclerosis, microglia are crucial in mediating inflammation and tissue damage. They are activated in response to inflammatory signals, leading to the production of pro-inflammatory cytokines, reactive oxygen species, and proteolytic enzymes, which contribute to neurodegeneration and the development of cortical lesions [22]. The inflammatory response driven by microglia can exacerbate neuronal damage, leading to progressive neurodegeneration [23]. For instance, polarized M1 microglia are associated with the production of neurotoxic molecules that impair neural networks and perpetuate inflammation [1].

Conversely, microglia also possess neuroprotective capabilities. They can adopt an M2 phenotype, secreting anti-inflammatory mediators and neurotrophic factors that promote tissue repair and homeostasis [1]. Recent studies suggest that microglia are involved in remyelination processes, aiding in the recovery of myelin after demyelination [24]. This regenerative aspect highlights their potential as therapeutic targets in MS, where modulation of microglial activation could enhance their protective functions while mitigating their harmful effects [25].

The heterogeneity of microglial activation is a critical factor in their role in MS. Advances in single-cell technologies have revealed that microglia exhibit high spatial and temporal heterogeneity, with specific states correlating with distinct pathological features and functions [2]. This complexity underscores the need for a nuanced understanding of microglial roles, as they can influence disease progression through both damaging and reparative mechanisms [26].

Moreover, the aging process impacts microglial function, leading to a decline in their ability to repair CNS damage, which may exacerbate the vulnerability of axons and neurons [22]. Therefore, the modulation of microglial activity is a promising strategy for therapeutic intervention in MS, aiming to balance their neurotoxic and neuroprotective roles [23].

In summary, microglia are pivotal in the pathophysiology of multiple sclerosis, exhibiting dual roles that can either promote neurodegeneration or facilitate repair. Understanding these mechanisms is essential for developing targeted therapies that harness the beneficial properties of microglia while minimizing their detrimental effects in neurodegenerative diseases.

4 Mechanisms of Microglial Activation

4.1 Neuroinflammation and Cytokine Release

Microglia, the resident immune cells of the central nervous system (CNS), play a pivotal role in the pathogenesis of neurodegenerative diseases through their activation and the subsequent release of pro-inflammatory cytokines. In response to various stimuli such as injury, infection, or the presence of misfolded proteins, microglia become activated and can adopt different phenotypes that influence their functional outcomes in neurodegenerative conditions.

Upon activation, microglia can polarize into two main states: M1 and M2. M1 microglia are associated with a pro-inflammatory response, characterized by the production of cytokines such as IL-1, IL-6, and TNF-α, which can lead to neurotoxicity and exacerbate neuronal damage. These pro-inflammatory cytokines contribute to the inflammatory milieu within the CNS, potentially promoting neuronal degeneration and dysfunction of neural networks [1][27][28]. Conversely, M2 microglia are typically associated with anti-inflammatory responses and tissue repair, secreting neurotrophic factors that support neuronal survival and homeostasis [1].

The dysregulation of microglial activation is a common feature in various neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). In these conditions, chronic activation of microglia leads to a sustained release of inflammatory cytokines, which can create a vicious cycle of neuroinflammation and neuronal damage [17][29]. This overactivation is thought to result from the accumulation of neurotoxic factors such as amyloid-beta (Aβ) peptides and cellular debris, which further stimulate microglial responses and exacerbate the inflammatory response [13][30].

Moreover, microglia's role in neuroinflammation extends beyond mere cytokine release; they are involved in the phagocytosis of apoptotic neurons and the clearance of pathological aggregates, which is essential for maintaining CNS homeostasis. However, when microglial function is compromised, or when they become excessively activated, their protective roles can turn detrimental, leading to increased neuronal loss and the progression of neurodegenerative diseases [31][32].

In summary, microglia serve as a double-edged sword in neurodegenerative diseases. While they are crucial for immune surveillance and tissue repair, their dysregulated activation and the subsequent release of pro-inflammatory cytokines can significantly contribute to the progression of neurodegeneration. Understanding the mechanisms of microglial activation and the modulation of their responses presents a promising avenue for therapeutic interventions aimed at mitigating neuroinflammation and its detrimental effects on neuronal health [17][33].

4.2 Phagocytosis and Clearance of Protein Aggregates

Microglia, the resident immune cells of the central nervous system (CNS), play a critical role in the pathogenesis of neurodegenerative diseases through various mechanisms, particularly in their capacity for phagocytosis and clearance of protein aggregates. These processes are essential for maintaining neural homeostasis and mitigating neuroinflammation, which is a hallmark of many neurodegenerative conditions.

In neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), microglia are activated in response to pathological changes, including the accumulation of misfolded proteins and cellular debris. This activation can lead to a range of functional states in microglia, which are characterized by their ability to phagocytose harmful entities, including protein aggregates like amyloid-beta in AD and alpha-synuclein in PD.

The phagocytic activity of microglia is crucial for clearing these aggregates from the extracellular space, thereby preventing further neuronal damage and promoting a healthier microenvironment. For instance, studies have shown that microglia can engulf and eliminate synaptic structures, a process that, while potentially detrimental in excess, can be protective when regulated appropriately [16]. The clearance of these aggregates is facilitated by the secretion of pro-inflammatory cytokines, which can recruit additional immune cells to the site of pathology, amplifying the inflammatory response [13].

However, the dual nature of microglial activation complicates their role in neurodegenerative diseases. While they can exhibit protective functions through phagocytosis, chronic activation can lead to detrimental effects, including the release of neurotoxic factors that exacerbate neuronal injury and inflammation [11]. This phenomenon has been observed where microglia transition from a neuroprotective to a neurotoxic phenotype, contributing to the progression of neurodegenerative diseases [8].

Moreover, the ability of microglia to clear protein aggregates can be impaired under certain conditions, such as excessive accumulation of these aggregates, which can lead to microglial dysfunction. This impairment may result in a vicious cycle where ineffective clearance leads to increased neuroinflammation and further neuronal degeneration [2]. Thus, the balance between the protective and harmful roles of microglia is critical in determining the outcome of neurodegenerative diseases.

In summary, microglia play a vital role in the pathogenesis of neurodegenerative diseases through their mechanisms of activation, phagocytosis, and clearance of protein aggregates. Their ability to adapt to different functional states underscores their complexity as both protectors and potential contributors to neurodegeneration. Understanding these dynamics is essential for developing therapeutic strategies aimed at modulating microglial activity to enhance their protective functions while minimizing their neurotoxic effects [9][34].

4.3 Interaction with Neuronal Cells

Microglia, the resident immune cells of the central nervous system (CNS), play multifaceted roles in neurodegenerative diseases through their activation mechanisms and interactions with neuronal cells. Their activation is often associated with changes in their structure, signaling, and function, which can either contribute to neurodegeneration or provide neuroprotection, depending on the context and state of activation.

Upon stimulation, microglia transition from a surveillant state to an activated phenotype. This activation is characterized by their ability to produce pro-inflammatory cytokines and neurotoxic molecules, particularly in the M1 polarized state, which can lead to neuronal dysfunction and promote neuroinflammatory responses. Conversely, in the M2 polarized state, microglia secrete anti-inflammatory mediators and neurotrophic factors that are crucial for restoring homeostasis and promoting repair processes in the CNS (Du et al., 2017; Gao et al., 2023).

The interactions between microglia and neuronal cells are critical for maintaining brain homeostasis. Microglia communicate with neurons, influencing their health and function. This communication allows microglia to maintain a surveillance state, enabling them to respond to environmental changes and potential threats effectively. Disruptions in these interactions can lead to impaired microglial function and contribute to the pathogenesis of neurodegenerative diseases (Harry, 2021).

Moreover, microglia play a role in synaptic pruning, a process essential for normal brain development and function. In Alzheimer's disease, for example, microglia are implicated in the phagocytosis of synaptic structures, which may lead to synapse loss and contribute to the neurodegenerative process (Rajendran & Paolicelli, 2018). This dual role of microglia—acting as both protectors and potential initiators of neurodegeneration—underscores the complexity of their function in various neurodegenerative conditions (Cao et al., 2025).

The dynamics of microglial activation are influenced by several factors, including systemic immune responses and environmental factors, which can shape their phenotype and function. Understanding these mechanisms is crucial for developing therapeutic strategies aimed at modulating microglial activity to promote neuroprotection while mitigating their potentially harmful effects (Gao et al., 2023; Xu et al., 2023).

In summary, microglia serve as key players in the pathophysiology of neurodegenerative diseases through their activation mechanisms and interactions with neuronal cells. Their ability to shift between protective and detrimental roles highlights the importance of targeting microglial functions for therapeutic interventions in neurodegenerative disorders.

5 Therapeutic Implications

5.1 Modulating Microglial Activity

Microglia, the resident immune cells of the central nervous system (CNS), play a dual role in neurodegenerative diseases, exhibiting both protective and detrimental effects. Understanding the complexities of microglial activation and their functional states is crucial for developing therapeutic strategies aimed at modulating their activity.

Microglia are involved in maintaining homeostasis within the CNS and responding to pathological insults. Upon activation, they can adopt different phenotypes, primarily classified into pro-inflammatory (M1) and anti-inflammatory (M2) states. M1 microglia produce pro-inflammatory cytokines and neurotoxic molecules that can exacerbate neurodegeneration, while M2 microglia secrete neurotrophic factors and anti-inflammatory mediators that support neuronal survival and tissue repair [1]. The balance between these states is critical; excessive M1 activation contributes to neuroinflammation and neuronal damage, while M2 activation promotes neuroprotection and repair [35].

Recent research emphasizes the potential of immunomodulation as a therapeutic approach in neurodegenerative diseases. Rather than solely focusing on anti-inflammatory strategies, there is a growing recognition of the importance of actively modulating microglial responses to enhance their protective functions. For instance, targeting specific molecular pathways involved in microglial activation can promote the transition from a neurotoxic to a neuroprotective phenotype, thereby reducing neuroinflammation and facilitating neuroregeneration [35].

The complexity of microglial responses necessitates a nuanced understanding of their roles in various neurodegenerative conditions. In diseases such as Alzheimer's, Parkinson's, and multiple sclerosis, microglia have been shown to engage in both neuroprotective and neurotoxic activities. They can phagocytose pathological protein aggregates, but excessive activation can lead to impaired phagocytic function and contribute to neuroinflammation [2]. Therapeutic strategies that enhance microglial phagocytosis while minimizing neuroinflammatory responses hold promise for mitigating disease progression [3].

Furthermore, recent advancements in understanding the molecular mechanisms regulating microglial activation have opened new avenues for therapy. For example, modulating glutamate signaling and exploring the role of microRNAs in microglial function are areas of active research. Dysregulated microRNA expression can influence microglial activation and cytokine production, which are critical in the context of neurodegenerative diseases [8].

Additionally, the emerging concept of microglial replacement therapy, which involves depleting dysfunctional microglia and repopulating the CNS with new, healthy microglia, presents a novel therapeutic strategy. This approach aims to restore the balance of microglial functions, enhancing neuroprotection while reducing neurotoxicity [36].

In conclusion, the modulation of microglial activity represents a promising therapeutic target in neurodegenerative diseases. By understanding the dual roles of microglia and developing strategies to enhance their protective functions while mitigating harmful inflammatory responses, it may be possible to create effective treatments that improve outcomes for patients suffering from these debilitating conditions. Continued research into the mechanisms governing microglial behavior and the development of targeted therapies will be essential in addressing the challenges posed by neurodegenerative diseases.

5.2 Potential Drug Targets

Microglia, the resident immune cells of the central nervous system (CNS), play a pivotal role in both the progression and potential treatment of neurodegenerative diseases. Their functions are multifaceted, ranging from homeostatic maintenance of neuronal networks to mediating neuroinflammation, which can lead to neuronal damage and degeneration. Understanding the dual roles of microglia—both protective and detrimental—has significant therapeutic implications.

In the context of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), microglia can exhibit both neuroprotective and neurotoxic properties. On one hand, microglia are involved in the clearance of pathological protein aggregates and dead cells, thus exerting protective effects through phagocytosis and the release of neurotrophic factors [2]. Conversely, their activation can lead to the release of pro-inflammatory cytokines and neurotoxic substances, contributing to neuroinflammation and neuronal loss [1]. This dichotomy underscores the complexity of microglial functions and their potential as therapeutic targets.

Recent advances in understanding microglial biology have highlighted several potential drug targets aimed at modulating their activity. Strategies include enhancing microglial phagocytosis, reducing neuroinflammation, and promoting a protective phenotype. For instance, therapies that target the modulation of microglial immune responses rather than merely inhibiting their inflammatory actions are being explored. This approach aims to retain their protective functions while mitigating their neurotoxic effects [35].

Furthermore, microglia-targeted interventions are being investigated, such as hematopoietic stem cell gene therapy, which has shown promise in replacing dysfunctional microglia with engineered cells capable of exerting beneficial effects in neurodegenerative conditions [37]. Additionally, the use of microglial inhibitors and activators, such as PLX3397 and lipopolysaccharide, has been studied for their effects on disease progression, suggesting that fine-tuning microglial activity could be a viable therapeutic avenue [36].

Another exciting area of research involves the modulation of microglial activity through microRNAs (miRNAs), which have been shown to regulate various aspects of microglial function, including their activation state and cytokine production. Dysregulated miRNA expression in microglia has been implicated in the pathogenesis of neurodegenerative diseases, presenting an opportunity for novel therapeutic strategies that target these regulatory pathways [8].

In summary, the role of microglia in neurodegenerative diseases is complex, encompassing both protective and detrimental effects. The therapeutic implications of this duality are profound, as they suggest that microglia can be targeted for intervention in neurodegeneration. Future therapeutic strategies may focus on enhancing the protective functions of microglia while inhibiting their neurotoxic responses, utilizing various approaches such as immunomodulation, gene therapy, and miRNA-based interventions to develop effective treatments for neurodegenerative diseases.

5.3 Future Directions in Microglial Research

Microglia, the resident immune cells of the central nervous system (CNS), play multifaceted roles in neurodegenerative diseases, exhibiting both protective and detrimental effects. They are crucial in maintaining CNS homeostasis, responding to injury, and mediating inflammatory responses. In neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis, microglia become activated and can adopt various phenotypes that influence their functional outcomes.

Microglia are involved in several key processes during neurodegeneration. They can phagocytose and clear pathological protein aggregates, contributing to neuroprotection. However, excessive activation can lead to neuroinflammation, characterized by the release of pro-inflammatory cytokines and neurotoxic molecules, which can exacerbate neuronal damage and promote disease progression (Gao et al. 2023; Du et al. 2017). The dual role of microglia in neurodegenerative diseases is underscored by their ability to shift between pro-inflammatory (M1) and anti-inflammatory (M2) states, which is critical in determining their impact on neuronal health and disease outcomes (Colonna & Butovsky 2017).

Therapeutically, targeting microglia has emerged as a promising strategy for treating neurodegenerative diseases. Recent research suggests that modulating microglial activity rather than simply inhibiting their inflammatory responses could enhance their protective functions while minimizing neurotoxicity. This approach includes strategies to promote the conversion of microglia into a protective phenotype, enhance their phagocytic capacity, and reduce neuroinflammation (Peña-Altamira et al. 2016; Zhang et al. 2023). Additionally, the development of novel microglial-targeted therapies, such as hematopoietic stem cell gene therapy, holds potential for restoring microglial function and promoting neuroprotection in various neurodegenerative conditions (Biffi 2024).

Future directions in microglial research should focus on elucidating the complex mechanisms underlying microglial activation and their diverse roles in neurodegenerative diseases. Advances in single-cell sequencing technologies have revealed significant heterogeneity among microglial populations, indicating that different microglial states may correlate with specific disease phenotypes and functional outcomes (Amor et al. 2022). Understanding these variations can inform the development of more targeted and effective therapeutic interventions. Moreover, investigating the interplay between microglia and other cell types in the CNS, including neurons and astrocytes, will be essential to comprehensively address the multifactorial nature of neurodegenerative diseases (Wang et al. 2023).

In conclusion, microglia are pivotal in the pathogenesis of neurodegenerative diseases, serving as both potential therapeutic targets and key players in disease progression. Continued exploration of their biology, activation states, and interactions within the CNS will be crucial for advancing therapeutic strategies aimed at mitigating neurodegeneration and enhancing neuroprotection.

6 Conclusion

Microglia are central to the pathogenesis of neurodegenerative diseases, exhibiting a dual role that can be both protective and detrimental. Their ability to switch between pro-inflammatory (M1) and anti-inflammatory (M2) states underscores the complexity of their functions in the central nervous system (CNS). Current research highlights the importance of understanding microglial biology, activation mechanisms, and interactions with neuronal cells to develop targeted therapeutic strategies. Modulating microglial activity presents a promising avenue for enhancing neuroprotection while minimizing neurotoxicity. Future research should focus on elucidating the diverse states of microglia, their heterogeneity, and their interactions with other CNS cell types to identify effective interventions for neurodegenerative diseases. The integration of novel therapeutic approaches, including gene therapy and microRNA modulation, may offer new hope for managing these debilitating conditions.

References

  • [1] Li Du;Ying Zhang;Yang Chen;Jie Zhu;Yi Yang;Hong-Liang Zhang. Role of Microglia in Neurological Disorders and Their Potentials as a Therapeutic Target.. Molecular neurobiology(IF=4.3). 2017. PMID:27830532. DOI: 10.1007/s12035-016-0245-0.
  • [2] Chao Gao;Jingwen Jiang;Yuyan Tan;Shengdi Chen. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets.. Signal transduction and targeted therapy(IF=52.7). 2023. PMID:37735487. DOI: 10.1038/s41392-023-01588-0.
  • [3] Shunqi Wang;Liangjing Pan;Chong Sun;Chaolin Ma;Haili Pan. Balancing Microglial Density and Activation in Central Nervous System Development and Disease.. Current issues in molecular biology(IF=3.0). 2025. PMID:40699743. DOI: 10.3390/cimb47050344.
  • [4] Kaitlyn K Thompson;Stella E Tsirka. The Diverse Roles of Microglia in the Neurodegenerative Aspects of Central Nervous System (CNS) Autoimmunity.. International journal of molecular sciences(IF=4.9). 2017. PMID:28245617. DOI: 10.3390/ijms18030504.
  • [5] Sarah A Kent;Veronique E Miron. Microglia regulation of central nervous system myelin health and regeneration.. Nature reviews. Immunology(IF=60.9). 2024. PMID:37452201. DOI: 10.1038/s41577-023-00907-4.
  • [6] Isabella Napoli;Harald Neumann. Protective effects of microglia in multiple sclerosis.. Experimental neurology(IF=4.2). 2010. PMID:19409897. DOI: 10.1016/j.expneurol.2009.04.024.
  • [7] Lijuan Zhang;Yafei Wang;Taohui Liu;Ying Mao;Bo Peng. Novel Microglia-based Therapeutic Approaches to Neurodegenerative Disorders.. Neuroscience bulletin(IF=5.8). 2023. PMID:36593381. DOI: 10.1007/s12264-022-01013-6.
  • [8] Shweta Pradip Jadhav. MicroRNAs in microglia: deciphering their role in neurodegenerative diseases.. Frontiers in cellular neuroscience(IF=4.0). 2024. PMID:38812793. DOI: 10.3389/fncel.2024.1391537.
  • [9] Luca Muzio;Alice Viotti;Gianvito Martino. Microglia in Neuroinflammation and Neurodegeneration: From Understanding to Therapy.. Frontiers in neuroscience(IF=3.2). 2021. PMID:34630027. DOI: 10.3389/fnins.2021.742065.
  • [10] Marco Colonna;Oleg Butovsky. Microglia Function in the Central Nervous System During Health and Neurodegeneration.. Annual review of immunology(IF=33.3). 2017. PMID:28226226. DOI: 10.1146/annurev-immunol-051116-052358.
  • [11] Juliane Obst;Emilie Simon;Renzo Mancuso;Diego Gomez-Nicola. The Role of Microglia in Prion Diseases: A Paradigm of Functional Diversity.. Frontiers in aging neuroscience(IF=4.5). 2017. PMID:28690540. DOI: 10.3389/fnagi.2017.00207.
  • [12] Bin Liu;Jau-Shyong Hong. Role of microglia in inflammation-mediated neurodegenerative diseases: mechanisms and strategies for therapeutic intervention.. The Journal of pharmacology and experimental therapeutics(IF=3.8). 2003. PMID:12490568. DOI: 10.1124/jpet.102.035048.
  • [13] Deepak Kumar Kaushik;Anirban Basu. A friend in need may not be a friend indeed: role of microglia in neurodegenerative diseases.. CNS & neurological disorders drug targets(IF=3.0). 2013. PMID:24070095. DOI: 10.2174/18715273113126660170.
  • [14] Qixue Wang;Mengna Lu;Xinyu Zhu;Xinyi Gu;Ting Zhang;Chenyi Xia;Li Yang;Ying Xu;Mingmei Zhou. The role of microglia immunometabolism in neurodegeneration: Focus on molecular determinants and metabolic intermediates of metabolic reprogramming.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2022. PMID:36076537. DOI: 10.1016/j.biopha.2022.113412.
  • [15] Min Guo;Yining Hao;Yiwei Feng;Haiqing Li;Yiting Mao;Qiang Dong;Mei Cui. Microglial Exosomes in Neurodegenerative Disease.. Frontiers in molecular neuroscience(IF=3.8). 2021. PMID:34045943. DOI: 10.3389/fnmol.2021.630808.
  • [16] Lawrence Rajendran;Rosa C Paolicelli. Microglia-Mediated Synapse Loss in Alzheimer's Disease.. The Journal of neuroscience : the official journal of the Society for Neuroscience(IF=4.0). 2018. PMID:29563239. DOI: 10.1523/JNEUROSCI.1136-17.2017.
  • [17] Yucong Xu;Wei Gao;Yingnan Sun;Minghua Wu. New insight on microglia activation in neurodegenerative diseases and therapeutics.. Frontiers in neuroscience(IF=3.2). 2023. PMID:38188026. DOI: 10.3389/fnins.2023.1308345.
  • [18] Na Li;Mingru Deng;Gonghui Hu;Nan Li;Haicheng Yuan;Yu Zhou. New Insights into Microglial Mechanisms of Memory Impairment in Alzheimer's Disease.. Biomolecules(IF=4.8). 2022. PMID:36421736. DOI: 10.3390/biom12111722.
  • [19] Yongle Cai;Jingliu Liu;Bin Wang;Miao Sun;Hao Yang. Microglia in the Neuroinflammatory Pathogenesis of Alzheimer's Disease and Related Therapeutic Targets.. Frontiers in immunology(IF=5.9). 2022. PMID:35558075. DOI: 10.3389/fimmu.2022.856376.
  • [20] Robert E Mrak. Neuropathology and the neuroinflammation idea.. Journal of Alzheimer's disease : JAD(IF=3.1). 2009. PMID:19584454. DOI: 10.3233/JAD-2009-1158.
  • [21] Waleed Hassan Almalki;Salem Salman Almujri. Therapeutic approaches to microglial dysfunction in Alzheimer's disease: Enhancing phagocytosis and metabolic regulation.. Pathology, research and practice(IF=3.2). 2024. PMID:39342887. DOI: 10.1016/j.prp.2024.155614.
  • [22] Jorge Correale. The role of microglial activation in disease progression.. Multiple sclerosis (Houndmills, Basingstoke, England)(IF=5.0). 2014. PMID:24812046. DOI: 10.1177/1352458514533230.
  • [23] Félix Distéfano-Gagné;Sara Bitarafan;Steve Lacroix;David Gosselin. Roles and regulation of microglia activity in multiple sclerosis: insights from animal models.. Nature reviews. Neuroscience(IF=26.7). 2023. PMID:37268822. DOI: 10.1038/s41583-023-00709-6.
  • [24] Vincent Pons;Serge Rivest. Beneficial Roles of Microglia and Growth Factors in MS, a Brief Review.. Frontiers in cellular neuroscience(IF=4.0). 2020. PMID:33173466. DOI: 10.3389/fncel.2020.00284.
  • [25] Yunrong Nan;Shuting Ni;Mei Liu;Kaili Hu. The emerging role of microglia in the development and therapy of multiple sclerosis.. International immunopharmacology(IF=4.7). 2024. PMID:39476566. DOI: 10.1016/j.intimp.2024.113476.
  • [26] Hubert Mado;Monika Adamczyk-Sowa;Paweł Sowa. Role of Microglial Cells in the Pathophysiology of MS: Synergistic or Antagonistic?. International journal of molecular sciences(IF=4.9). 2023. PMID:36768183. DOI: 10.3390/ijms24031861.
  • [27] Joshua A Smith;Arabinda Das;Swapan K Ray;Naren L Banik. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases.. Brain research bulletin(IF=3.7). 2012. PMID:22024597. DOI: 10.1016/j.brainresbull.2011.10.004.
  • [28] Ling Xu;Dan He;Ying Bai. Microglia-Mediated Inflammation and Neurodegenerative Disease.. Molecular neurobiology(IF=4.3). 2016. PMID:26659872. DOI: 10.1007/s12035-015-9593-4.
  • [29] Charannya Sozheesvari Subhramanyam;Cheng Wang;Qidong Hu;S Thameem Dheen. Microglia-mediated neuroinflammation in neurodegenerative diseases.. Seminars in cell & developmental biology(IF=6.0). 2019. PMID:31077796. DOI: 10.1016/j.semcdb.2019.05.004.
  • [30] R Bryan Rock;Phillip K Peterson. Microglia as a pharmacological target in infectious and inflammatory diseases of the brain.. Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology(IF=3.5). 2006. PMID:18040778. DOI: 10.1007/s11481-006-9012-8.
  • [31] Shofiul Azam;Md Ezazul Haque;In-Su Kim;Dong-Kug Choi. Microglial Turnover in Ageing-Related Neurodegeneration: Therapeutic Avenue to Intervene in Disease Progression.. Cells(IF=5.2). 2021. PMID:33466587. DOI: 10.3390/cells10010150.
  • [32] Andrei Turkin;Oksana Tuchina;Friederike Klempin. Microglia Function on Precursor Cells in the Adult Hippocampus and Their Responsiveness to Serotonin Signaling.. Frontiers in cell and developmental biology(IF=4.3). 2021. PMID:34109176. DOI: 10.3389/fcell.2021.665739.
  • [33] Silke Miller;Maria-Jesus Blanco. Small molecule therapeutics for neuroinflammation-mediated neurodegenerative disorders.. RSC medicinal chemistry(IF=3.6). 2021. PMID:34223157. DOI: 10.1039/d1md00036e.
  • [34] André F Batista;Khyrul A Khan;Maria-Tzousi Papavergi;Cynthia A Lemere. The Importance of Complement-Mediated Immune Signaling in Alzheimer's Disease Pathogenesis.. International journal of molecular sciences(IF=4.9). 2024. PMID:38255891. DOI: 10.3390/ijms25020817.
  • [35] Emiliano Peña-Altamira;Federica Prati;Francesca Massenzio;Marco Virgili;Antonio Contestabile;Maria Laura Bolognesi;Barbara Monti. Changing paradigm to target microglia in neurodegenerative diseases: from anti-inflammatory strategy to active immunomodulation.. Expert opinion on therapeutic targets(IF=4.4). 2016. PMID:26568363. DOI: 10.1517/14728222.2016.1121237.
  • [36] Yan Wang;Limei Cui;He Zhao;Huhuifen He;Liang Chen;Xicheng Song;Dawei Liu;Jingjing Qiu;Yan Sun. Exploring the Connectivity of Neurodegenerative Diseases: Microglia as the Center.. Journal of inflammation research(IF=4.1). 2023. PMID:38107384. DOI: 10.2147/JIR.S440377.
  • [37] Alessandra Biffi. Hematopoietic stem cell gene therapy to halt neurodegeneration.. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics(IF=6.9). 2024. PMID:39276677. DOI: 10.1016/j.neurot.2024.e00440.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

Microglia · Neurodegenerative Diseases · Neuroinflammation · Cytokines · Therapeutic Strategies


© 2025 MaltSci