Appearance
This report is written by MaltSci based on the latest literature and research findings
What are the neurobiological mechanisms of schizophrenia?
Abstract
Schizophrenia is a complex mental disorder characterized by a range of symptoms including delusions, hallucinations, and cognitive impairments, affecting approximately 1% of the global population. Understanding the neurobiological mechanisms underlying schizophrenia is essential for developing effective treatment strategies. This review synthesizes current knowledge on the neurotransmitter systems involved, particularly focusing on dopamine, glutamate, and serotonin, and their contributions to the disorder's pathology. The dopamine hypothesis has been a longstanding framework, suggesting that dysregulation of dopaminergic activity drives psychotic symptoms. However, recent research has expanded this view to include glutamatergic dysfunction, particularly involving NMDA receptor hypofunction, and serotonin receptor alterations, which play significant roles in cognitive and emotional disturbances. Neuroimaging studies reveal structural and functional brain abnormalities, particularly in the frontal and medial temporal lobes, highlighting connectivity issues and network dysfunction. Genetic and epigenetic factors further complicate the disorder's etiology, with heritable genetic variations interacting with environmental stressors to increase risk. Neuroinflammation has emerged as a critical component, with evidence of immune dysregulation contributing to the disorder's symptoms. This review also discusses current and emerging therapeutic approaches, emphasizing the need for innovative strategies that target the underlying biological processes rather than merely managing symptoms. By integrating findings from diverse research areas, this review aims to provide a comprehensive overview of the neurobiological mechanisms of schizophrenia, paving the way for future research and improved treatment outcomes.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Neurotransmitter Systems in Schizophrenia
- 2.1 Dopamine Hypothesis
- 2.2 Glutamate and NMDA Receptor Dysfunction
- 2.3 Role of Serotonin
- 3 Structural and Functional Brain Abnormalities
- 3.1 Neuroimaging Findings
- 3.2 Cortical and Subcortical Changes
- 3.3 Connectivity and Network Dysfunction
- 4 Genetic and Epigenetic Factors
- 4.1 Heritability and Genetic Risk Factors
- 4.2 Epigenetic Modifications and Environmental Interactions
- 5 Neuroinflammation and Immune System Involvement
- 5.1 Inflammatory Markers in Schizophrenia
- 5.2 Microglial Activation and Its Consequences
- 6 Current and Emerging Therapeutic Approaches
- 6.1 Pharmacological Treatments
- 6.2 Psychosocial Interventions
- 6.3 Future Directions in Therapy
- 7 Summary
1 Introduction
Schizophrenia is a complex and multifaceted mental disorder characterized by a range of symptoms, including delusions, hallucinations, disorganized thinking, and cognitive impairments. The prevalence of schizophrenia is approximately 1% globally, and it poses significant challenges not only to individuals affected but also to healthcare systems due to its chronic nature and associated disabilities. Understanding the neurobiological mechanisms underlying schizophrenia is crucial for developing effective treatment strategies and improving patient outcomes. Recent advancements in neuroscience have illuminated the intricate interplay between genetic, neurochemical, and environmental factors that contribute to the onset and progression of this disorder. As such, this review aims to synthesize current knowledge regarding the neurobiological mechanisms of schizophrenia, with a focus on neurotransmitter systems, structural brain abnormalities, and the role of neuroinflammation.
The significance of exploring the neurobiological underpinnings of schizophrenia lies in the potential for developing innovative therapeutic approaches that target the underlying biological processes of the disorder, moving beyond mere symptom management. The historical context of schizophrenia research has predominantly centered around the dopamine hypothesis, which posits that dysregulation of dopaminergic activity in the brain is a primary driver of psychotic symptoms [1]. However, this framework has evolved significantly, with increasing recognition of the roles played by other neurotransmitter systems, such as glutamate and serotonin, as well as neuroinflammatory processes that may exacerbate the disorder [2].
Current research highlights a multitude of neurobiological mechanisms implicated in schizophrenia. Neurotransmitter dysregulation is a well-established area of focus, particularly concerning the glutamatergic system and the N-methyl-D-aspartate (NMDA) receptor, which has been shown to be hypoactive in individuals with schizophrenia [3]. Furthermore, neuroimaging studies have revealed structural and functional brain abnormalities, including alterations in cortical and subcortical regions, as well as disrupted connectivity and network dysfunction [4]. The exploration of genetic and epigenetic factors has also gained momentum, revealing how gene-environment interactions can contribute to the risk of developing schizophrenia [5].
Neuroinflammation has emerged as a crucial factor in the pathophysiology of schizophrenia, with studies indicating that immune dysregulation and the activation of glial cells may play a significant role in the disorder's manifestation [6]. This highlights the importance of considering the immune system's involvement in the development and progression of schizophrenia, as well as its potential as a therapeutic target [7]. Recent evidence supports the "mild encephalitis" hypothesis, suggesting that neuroinflammatory processes may contribute to the cognitive and behavioral symptoms observed in schizophrenia [4].
The organization of this review will follow a structured approach, beginning with an examination of the neurotransmitter systems involved in schizophrenia, including the dopamine hypothesis, glutamate dysfunction, and the role of serotonin. Subsequently, we will delve into the structural and functional brain abnormalities associated with the disorder, exploring neuroimaging findings, cortical and subcortical changes, and connectivity issues. The discussion will then shift to genetic and epigenetic factors, emphasizing the heritability of schizophrenia and the impact of environmental interactions. Following this, we will explore the role of neuroinflammation and immune system involvement in schizophrenia, highlighting inflammatory markers and the consequences of microglial activation. Finally, we will discuss current and emerging therapeutic approaches, encompassing pharmacological treatments, psychosocial interventions, and future directions in therapy.
By synthesizing findings from recent research, this review aims to provide a comprehensive overview of how these neurobiological factors interact to influence the pathophysiology of schizophrenia. Ultimately, this understanding can pave the way for innovative therapeutic strategies that address the root causes of the disorder, potentially improving the quality of life for individuals affected by schizophrenia.
2 Neurotransmitter Systems in Schizophrenia
2.1 Dopamine Hypothesis
The neurobiological mechanisms underlying schizophrenia are complex and multifaceted, with a significant emphasis on neurotransmitter systems, particularly the dopamine hypothesis. This hypothesis posits that dysregulation of dopaminergic activity plays a central role in the pathophysiology of schizophrenia.
The dopamine hypothesis of schizophrenia has been supported by a substantial body of pharmacological evidence, suggesting that alterations in dopaminergic synaptic transmission are implicated in the disorder's pathogenesis. Despite ongoing debates regarding the completeness of this hypothesis, it remains a foundational concept in understanding schizophrenia. Research indicates that dopaminergic alterations are present not only in individuals with schizophrenia but also in those at genetic risk for the disorder who do not exhibit clinical symptoms, suggesting a "dopamine hypothesis of schizophrenia vulnerability" (Hirvonen & Hietala, 2011)[8].
However, the dopamine hypothesis alone does not account for all aspects of schizophrenia. Recent studies have identified the involvement of other neurotransmitter systems, such as glutamate and serotonin, in the disorder's etiology. For instance, the interaction between dopaminergic and glutamatergic systems has been proposed as a critical factor in the development of both positive and negative symptoms of schizophrenia. Specifically, NMDA receptor hypofunction has been implicated in the cognitive deficits associated with the disorder, indicating that glutamatergic dysfunction may also play a crucial role (Coyle, 2004)[9].
Additionally, neuroinflammatory processes have emerged as significant contributors to the pathophysiology of schizophrenia. Evidence suggests that aberrant immune mechanisms in both the peripheral and central nervous systems may influence the development and progression of the disorder, further complicating the neurobiological landscape (Watkins & Andrews, 2016)[7].
Furthermore, advancements in neuroimaging techniques have enhanced our understanding of the neurobiological underpinnings of schizophrenia, revealing neurochemical intermediate phenotypes that connect genetic predispositions to the clinical manifestations of the disorder. This integration of neuroimaging findings with neurotransmitter system dysfunction highlights the need for a comprehensive approach to studying schizophrenia, encompassing genetic, environmental, and neurobiological factors (Tost et al., 2010)[1].
In conclusion, while the dopamine hypothesis remains a cornerstone in the understanding of schizophrenia, it is increasingly recognized that the disorder's neurobiological mechanisms involve a complex interplay of multiple neurotransmitter systems, including glutamate and serotonin, as well as neuroinflammatory processes. This multifactorial perspective is essential for developing more effective therapeutic strategies and identifying reliable biomarkers for schizophrenia.
2.2 Glutamate and NMDA Receptor Dysfunction
Schizophrenia is a complex neuropsychiatric disorder characterized by a range of symptoms including hallucinations, delusions, cognitive deficits, and negative symptoms. A significant body of research suggests that dysregulation of the glutamatergic neurotransmission system, particularly involving the N-methyl-D-aspartate (NMDA) receptor, plays a crucial role in the pathophysiology of schizophrenia.
The glutamate hypothesis of schizophrenia posits that hypofunction of NMDA receptors is causally associated with both positive and negative symptoms of the disorder. According to the glutamate hypothesis, abnormalities in glutamate transmission may arise from a variety of mechanisms, including the modulation of glutamate synthesis, clearance, and the release of co-agonists like D-serine that are necessary for NMDA receptor activation (Mei et al., 2018; Labrie et al., 2012). Astrocytes, which are the primary regulatory glial cells in the brain, are integral to these processes, as they modulate glutamate metabolism and transmission. They are involved in the glutamate-glutamine cycle, where they help in synthesizing glutamate and clearing excess glutamate from the synaptic cleft via excitatory amino acid transporters (EAATs) (Mei et al., 2018).
Furthermore, D-serine, a co-agonist at the NMDA receptor, is crucial for its full activation. Evidence suggests that abnormalities in D-serine availability may underlie glutamatergic dysfunction in schizophrenia. Studies indicate that alterations in genes associated with the D-serine pathway could significantly impact the disorder's etiology and pathophysiology (Labrie et al., 2012). In addition, endogenous antagonists of the NMDA receptor, such as kynurenic acid, have been implicated in the disease, as elevated levels of kynurenic acid have been found in the cerebrospinal fluid of individuals with schizophrenia (Muller & Schwarz, 2006). This antagonist can block NMDA receptor function and contribute to the symptoms observed in schizophrenia.
Clinical and experimental findings support the notion that NMDA receptor dysfunction is linked to cognitive deficits and negative symptoms in schizophrenia. For instance, NMDA receptor antagonists, such as phencyclidine, can induce psychosis in healthy individuals, mimicking the symptoms of schizophrenia (Coyle, 1996). This has led to the hypothesis that the excitotoxic effects resulting from increased glutamate release, due to NMDA receptor dysfunction, may play a significant role in the development and progression of the disorder (Stone et al., 2007; Marsman et al., 2013).
Additionally, recent studies have shown that alterations in glutamatergic signaling may also involve changes in receptor trafficking and synaptic targeting, indicating a more complex dysregulation of glutamatergic pathways in schizophrenia (Kristiansen et al., 2007). The interaction between glutamatergic and dopaminergic systems further complicates the neurobiological landscape of schizophrenia, suggesting that therapies targeting these pathways may hold promise for improving outcomes in affected individuals (Jorratt et al., 2021; Inta et al., 2010).
In summary, the neurobiological mechanisms underlying schizophrenia, particularly concerning glutamate and NMDA receptor dysfunction, involve a multifaceted interplay of astrocytic regulation, D-serine availability, and the actions of endogenous antagonists. Understanding these mechanisms is essential for developing targeted therapies aimed at restoring glutamatergic balance and improving the clinical management of schizophrenia.
2.3 Role of Serotonin
Schizophrenia is a complex psychiatric disorder characterized by a heterogeneous etiology involving multiple neurotransmitter systems, including serotonin. The role of serotonin in schizophrenia has been extensively studied, and it is implicated in various aspects of the disorder's pathophysiology.
Serotonin, a neurotransmitter widely distributed throughout the central nervous system, plays a crucial role in mood regulation and cognitive functions. Dysregulation of serotonin transmission has been linked to several psychiatric disorders, including schizophrenia. Postmortem studies have revealed a reduction in specific serotonin receptor subtypes, particularly 5-HT2 receptors and serotonin reuptake sites, in the prefrontal cortex of patients with schizophrenia [10]. These findings suggest that alterations in serotonin receptor density and function may contribute to the cognitive and emotional disturbances observed in schizophrenia.
The serotonergic system's involvement in schizophrenia is further supported by the observation of genetic alterations within the serotonin pathway. Studies have identified several single nucleotide polymorphisms (SNPs) in serotonin-related genes that are more prevalent in individuals with schizophrenia compared to healthy controls. These genetic variations can influence the expression and function of serotonin receptors and transporters, thereby affecting neurotransmission and potentially contributing to the disorder's phenotypic diversity [11].
Moreover, the interaction between serotonin and other neurotransmitter systems, such as dopamine and glutamate, is critical in understanding schizophrenia's neurobiology. Schizophrenia is believed to arise from the pathological interplay of these systems, rather than from a single neurotransmitter's dysfunction [12]. For instance, neurotensin, a neuropeptide that modulates various neurotransmitter systems, has been shown to have altered concentrations in the cerebrospinal fluid of some schizophrenic patients, which normalizes with effective antipsychotic treatment [12]. This highlights the potential for targeting multiple neurotransmitter systems in developing more effective treatments for schizophrenia.
Research has also focused on the presynaptic mechanisms of neurotransmitter release, revealing that dysregulations in synaptic vesicle trafficking may impact serotonin signaling in key brain regions implicated in schizophrenia [13]. Such dysregulations could disrupt the delicate balance of neurotransmitter interactions necessary for normal cognitive and emotional functioning.
Additionally, the serotonergic system's involvement in the mechanism of action of antipsychotic drugs is noteworthy. Antipsychotics, particularly atypical ones, have been shown to increase serotonin neurotransmission, which may be crucial for their therapeutic effects [12]. This relationship suggests that enhancing serotonin signaling could alleviate some of the symptoms of schizophrenia.
In conclusion, the neurobiological mechanisms underlying schizophrenia involve complex interactions between serotonin and other neurotransmitter systems. Dysregulation of serotonin receptors, genetic alterations in the serotonin pathway, and interactions with dopaminergic and glutamatergic systems all contribute to the disorder's pathophysiology. Understanding these mechanisms is essential for developing targeted therapies that address the multifaceted nature of schizophrenia.
3 Structural and Functional Brain Abnormalities
3.1 Neuroimaging Findings
Schizophrenia is characterized by a range of neurobiological mechanisms that manifest through various structural and functional brain abnormalities, as evidenced by neuroimaging findings. Research has consistently highlighted the involvement of key brain regions, particularly the frontal and medial temporal lobes, which are crucial for cognitive functions. These areas exhibit significant deficits, correlating with the symptoms of schizophrenia, such as hallucinations and cognitive dysfunctions. For instance, Cannon (1996) emphasizes that individuals with lesions in these regions display symptoms akin to those seen in schizophrenia, indicating the importance of these structures in the disorder's pathophysiology [14].
Neuroimaging studies have revealed that both structural and functional abnormalities are prevalent not only in patients with schizophrenia but also in their close relatives, suggesting a genetic component to these changes. Ahmed et al. (2013) note that neuroimaging phenotypes may serve as potential endophenotypes for schizophrenia, as they can predict the emergence of psychosis in high-risk individuals [15]. This predictive capability underscores the significance of identifying neurobiological markers associated with the disorder.
Functional dysconnectivity is another critical aspect of schizophrenia's neurobiology. Kraguljac and Lahti (2021) discuss how neuroimaging can elucidate brain circuitry alterations, revealing that specific brain circuits are preferentially affected. These alterations are already detectable in first-episode psychosis patients, suggesting that early intervention may be beneficial [16]. Furthermore, the studies indicate that dopaminergic dysregulation plays a pivotal role in the disorder, with antipsychotic treatments showing some efficacy in alleviating neural abnormalities [16].
The concept of neurodevelopmental origins is also supported by neuroimaging evidence. Research indicates that abnormalities in brain structure and function may arise from both genetic predispositions and environmental factors, such as obstetric complications, which can exacerbate the risk of developing schizophrenia [14]. Watanabe et al. (2010) highlight the role of inflammatory processes, particularly cytokines, in the pathogenesis of schizophrenia, suggesting that immune responses may interact with neurodevelopmental pathways [17].
Moreover, the connectomics approach has shed light on the structural connectivity within the brain, revealing that schizophrenia may result from disruptions in the brain's integrative thought processes. Van den Heuvel and Fornito (2014) discuss how advancements in neuroimaging techniques have allowed for the reconstruction of comprehensive neural connectivity maps, providing insights into how abnormalities in these networks contribute to the disorder [18].
In summary, the neurobiological mechanisms of schizophrenia are multifaceted, involving structural and functional brain abnormalities, dysconnectivity in neural circuits, genetic and environmental interactions, and inflammatory processes. Neuroimaging studies play a crucial role in elucidating these mechanisms, offering valuable insights into the disorder's pathophysiology and potential therapeutic targets.
3.2 Cortical and Subcortical Changes
Schizophrenia is characterized by a range of neurobiological mechanisms, particularly involving structural and functional abnormalities in both cortical and subcortical regions of the brain. Evidence from various studies indicates that these abnormalities play a significant role in the pathophysiology of the disorder.
Cortical dysfunction in schizophrenia is often associated with altered expression of RNA and proteins that are crucial for neurotransmission, metabolism, and myelination. The molecular mechanisms underlying these alterations remain largely elusive, yet they are believed to involve transcriptional dysregulation, which includes changes in DNA and histone modifications as well as microRNA-mediated post-transcriptional mechanisms [19]. This suggests that epigenetic factors may contribute significantly to the neurobiology of schizophrenia.
Moreover, schizophrenia has been implicated as a neurodevelopmental disorder that affects the prefrontal and temporal cortical neural systems. There is considerable evidence indicating presynaptic changes in subcortical dopamine neurotransmission, along with alterations in cortical glutamatergic and GABAergic systems [20]. These findings are supported by functional neuroimaging studies that reveal cortical abnormalities, which are believed to underlie the cognitive deficits commonly observed in schizophrenia [20].
The entorhinal cortex, a critical area involved in cognitive processes and olfactory function, has been identified as exhibiting both structural and functional abnormalities in individuals with schizophrenia. These changes are thought to be central to the cognitive impairments associated with the disorder [21]. Additionally, the medial temporal lobe has been identified as a region where abnormalities are likely present in all individuals with schizophrenia, suggesting a disturbance in the normal pattern of brain development that could be linked to the clinical symptoms of the disorder [22].
Postmortem studies and in vivo imaging have revealed a variety of abnormalities in brain structure and function, particularly in the frontal and medial temporal lobe regions. These abnormalities may stem from genetic predispositions, obstetric complications, and developmental anomalies that affect neuronal organization [14]. The evidence indicates that the interaction of genetic factors with environmental influences may play a crucial role in the development of these structural changes [22].
Furthermore, there is growing recognition of the neurovascular unit dysfunction and blood-brain barrier hyperpermeability in schizophrenia. These factors may contribute to the cognitive and behavioral symptoms of the disorder by affecting cerebral perfusion and the homeostatic processes of the cerebral microenvironment [4]. The disruption of the blood-brain barrier can also facilitate interactions between brain innate and peripheral adaptive immunity, which may perpetuate neuroinflammatory responses and exacerbate symptoms [4].
In summary, the neurobiological mechanisms of schizophrenia are multifaceted, involving significant structural and functional changes in cortical and subcortical areas. These changes are influenced by a combination of genetic, epigenetic, and environmental factors, leading to the complex clinical manifestations of the disorder. Understanding these mechanisms is crucial for developing effective therapeutic strategies for individuals affected by schizophrenia.
3.3 Connectivity and Network Dysfunction
Schizophrenia is widely regarded as a disorder characterized by significant abnormalities in brain connectivity and network dysfunction. The neurobiological mechanisms underlying these phenomena are multifaceted, involving both structural and functional brain abnormalities.
The dysconnectivity hypothesis posits that schizophrenia arises from disrupted brain connectivity, which manifests as both anatomical and functional dysconnectivity. For instance, a comprehensive review highlights that schizophrenia is hypothesized to result from abnormal structural connectivity, particularly within the prefrontal cortex (PFC) and its connections to other brain regions. Neuroimaging studies utilizing diffusion tensor imaging (DTI) and functional magnetic resonance imaging (fMRI) have provided in vivo evidence for such disruptions, linking them to impaired cognitive functions and aberrant behaviors observed in patients with schizophrenia (Zhou et al., 2015)[23].
Furthermore, the evidence indicates that schizophrenia is associated with a reduction in structural connectivity, as observed through magnetic resonance imaging (MRI). However, the relationship between structural and functional connectivity is complex; for example, increased functional connectivity has been reported in some studies, suggesting a dysregulation of brain activity that may arise from abnormal neurodevelopmental wiring (Fornito & Bullmore, 2015)[24]. This paradox of decreased structural connectivity coexisting with increased functional connectivity reflects a potential compensatory mechanism or a maladaptive response within the brain's network architecture.
Moreover, developmental disruptions in neural connectivity are believed to play a critical role in the pathophysiology of schizophrenia. Longitudinal studies indicate that connectivity disruptions may emerge during critical periods of brain development, suggesting that genetic and environmental factors contribute to the establishment of intra- and interregional connectivity in individuals at risk for the disorder (Karlsgodt et al., 2008)[25].
Additionally, the involvement of neurovascular mechanisms has garnered attention in recent years. Abnormalities in neurovascular interactions and blood-brain barrier (BBB) hyperpermeability have been proposed as contributing factors to the neurobiology of schizophrenia. These vascular changes may influence cerebral perfusion and contribute to cognitive and behavioral symptoms, potentially linking peripheral vascular dysfunction with central nervous system pathology (Najjar et al., 2017)[4].
Overall, the integration of findings from neuroimaging, genetic studies, and investigations into neurovascular mechanisms paints a complex picture of schizophrenia as a disorder fundamentally linked to disrupted brain connectivity and network dysfunction. Understanding these neurobiological mechanisms is essential for developing targeted therapeutic strategies aimed at addressing the cognitive and emotional deficits associated with the disorder.
4 Genetic and Epigenetic Factors
4.1 Heritability and Genetic Risk Factors
Schizophrenia is a complex psychiatric disorder with a multifaceted etiology that includes significant genetic and epigenetic components. The heritability of schizophrenia is estimated to be around 80%, indicating a strong genetic influence on its development. However, the genetic basis of schizophrenia is not straightforward, as numerous studies have shown that no single genetic variant can account for the disorder. Instead, schizophrenia is considered a polygenic condition, where multiple genes contribute to susceptibility, with the cumulative effect of these genetic variations influencing the risk of developing the disorder [26].
Recent research has highlighted the role of epigenetic mechanisms in schizophrenia, particularly how these processes mediate the interaction between genetic predispositions and environmental factors. Epigenetic modifications, such as DNA methylation and histone modifications, can lead to changes in gene expression without altering the underlying DNA sequence. These modifications can be influenced by various environmental exposures, including stress, infections, and nutritional factors, which may contribute to the onset of schizophrenia [5][27].
The concept of epigenetic dysregulation is crucial for understanding schizophrenia's pathophysiology. For instance, studies have indicated that environmental stressors, especially during critical developmental periods, can result in lasting epigenetic changes that affect neurobiological processes. Such alterations may disrupt normal brain development and function, thereby increasing vulnerability to psychotic disorders [28][29].
In addition to genetic and epigenetic factors, neuroinflammation has emerged as a significant contributor to the neurobiological mechanisms underlying schizophrenia. Evidence suggests that inflammatory processes mediated by cytokines may play a crucial role in the disorder's etiology. Maternal infections and other environmental factors can activate the immune response, leading to elevated levels of pro-inflammatory cytokines, which in turn may influence brain development and function [6][17].
Furthermore, the interplay between genetic predispositions and environmental factors is increasingly understood through the lens of the immune-brain axis. This axis encompasses how immune responses can affect neuronal development and synaptic plasticity, potentially leading to the behavioral and cognitive deficits characteristic of schizophrenia [6].
In summary, the neurobiological mechanisms of schizophrenia involve a complex interplay of genetic and epigenetic factors, where heritable genetic variations interact with environmental influences to shape the risk of developing the disorder. This interaction is mediated through epigenetic modifications and inflammatory processes, underscoring the multifactorial nature of schizophrenia's etiology and the need for a comprehensive understanding of its underlying biological mechanisms [30][31].
4.2 Epigenetic Modifications and Environmental Interactions
Schizophrenia is characterized by a complex interplay of genetic and environmental factors that contribute to its neurobiological mechanisms. Recent research emphasizes the significance of epigenetic modifications, which serve as a crucial link between environmental influences and gene expression alterations associated with the disorder.
Epigenetic mechanisms, including DNA methylation, histone modifications, and non-coding RNAs, have been implicated in the pathophysiology of schizophrenia. These modifications can lead to lasting changes in gene expression, particularly during critical developmental periods. For instance, environmental exposures such as maternal immune infection, obstetric complications, childhood trauma, and substance abuse can induce epigenetic alterations that influence neurobiological processes relevant to schizophrenia [29]. The concept of missing heritability in schizophrenia, where genetic factors alone do not account for the phenotypic variation observed, suggests that epigenetic mechanisms play a pivotal role in modulating the expression of genes involved in the disorder [5].
Research has demonstrated that psychosis-associated environmental exposures may result in long-lasting epigenetic changes that affect neurobiological pathways, including dopamine, NMDA, and GABA signaling [32]. These pathways are crucial for normal cognitive function and their dysregulation is linked to the symptomatology of schizophrenia. Moreover, the dynamic nature of the epigenome allows for potential reversibility of these modifications, which opens avenues for therapeutic interventions targeting epigenetic changes [28].
The interplay between genetic predisposition and environmental stressors underscores the multifactorial etiology of schizophrenia. Studies have shown that while heritability estimates for schizophrenia are around 80%, various environmental factors significantly contribute to the disorder's development [29]. This includes social determinants, prenatal stress, and exposure to toxins, all of which can lead to epigenetic modifications that exacerbate or mitigate the risk of developing schizophrenia [33].
The investigation into epigenetic alterations in schizophrenia has been facilitated by advancements in epigenomic profiling technologies, enabling researchers to explore how these modifications affect gene expression and contribute to disease pathology [28]. Notably, the identification of epigenetic biomarkers in peripheral tissues such as blood or saliva presents a promising approach for early detection and intervention strategies [29].
In summary, the neurobiological mechanisms underlying schizophrenia are significantly influenced by epigenetic modifications resulting from environmental interactions. These findings highlight the importance of understanding both genetic and non-genetic factors in the etiology of schizophrenia, paving the way for innovative therapeutic strategies that target the epigenetic landscape associated with the disorder.
5 Neuroinflammation and Immune System Involvement
5.1 Inflammatory Markers in Schizophrenia
Schizophrenia is a complex neurodevelopmental disorder characterized by a combination of cognitive, behavioral, and emotional dysregulations. Recent studies have increasingly focused on the role of neuroinflammation and immune system abnormalities in its pathogenesis, suggesting that these factors significantly contribute to the disorder's clinical manifestations.
A growing body of evidence indicates that immune dysregulation and neuroinflammation are integral to the etiology of schizophrenia. For instance, it has been shown that schizophrenia is associated with abnormalities across all components of the immune system, including innate and adaptive immunity, as well as humoral and cellular immunity [34]. Notably, there is evidence of increased levels of pro-inflammatory cytokines, such as interleukins (IL-6, IL-8), tumor necrosis factor-alpha (TNF-α), and C-reactive protein in patients with schizophrenia [35][36]. These inflammatory markers are often elevated in both the blood and cerebrospinal fluid of affected individuals, suggesting a systemic inflammatory response [37].
The involvement of neuroinflammation in schizophrenia is further supported by findings of activated microglia, the resident immune cells of the central nervous system (CNS), which respond to pathological changes by producing pro-inflammatory cytokines and free radicals. This activation can lead to neuronal degeneration and has been implicated in the cognitive deficits observed in schizophrenia [35][38]. Moreover, the chronic low-grade inflammation associated with the disorder may disrupt neurotransmitter systems, particularly dopaminergic, serotonergic, and glutamatergic pathways, which are crucial for mood and cognition [39].
Recent research has highlighted the role of specific immune cell types, such as T helper (Th) 17 cells, which produce pro-inflammatory cytokines and may contribute to the permeability of the blood-brain barrier [40]. The dysregulation of these immune responses not only exacerbates neuroinflammation but may also create a feedback loop that perpetuates the disorder's symptoms [41].
Furthermore, maternal infections during pregnancy and early-life stress have been linked to increased risk for developing schizophrenia, emphasizing the significance of immune responses during critical developmental periods [17]. These environmental factors may interact with genetic predispositions to influence the immune system's functionality, leading to a heightened inflammatory state that contributes to the pathophysiology of schizophrenia [6].
Overall, the interplay between genetic, environmental, and immune factors culminates in a neuroinflammatory response that is central to the pathogenesis of schizophrenia. Understanding these mechanisms may open avenues for new therapeutic strategies aimed at mitigating inflammation and improving clinical outcomes for individuals suffering from this debilitating disorder [40][42].
5.2 Microglial Activation and Its Consequences
Schizophrenia is increasingly recognized as a complex psychiatric disorder where neuroinflammation and immune system dysregulation play critical roles in its pathogenesis. Central to this understanding is the activation of microglia, the resident immune cells of the central nervous system (CNS), which have been implicated in various neurobiological mechanisms associated with schizophrenia.
Microglial activation is characterized by morphological changes and increased expression of pro-inflammatory cytokines, which can lead to neuronal damage and contribute to the pathophysiology of schizophrenia. Studies have shown that activated microglia may engage in excessive synaptic pruning, a process that is crucial during brain development but becomes detrimental when dysregulated in adults. This excessive pruning is believed to primarily affect glutamatergic neurons, leading to synaptic loss, which is a hallmark of schizophrenia (Hartmann et al., 2024) [43].
The role of microglia in schizophrenia is further complicated by their interaction with other glial cells, such as astrocytes. Dysregulated astrocytic activity can exacerbate microglial activation, creating a feedback loop that amplifies neuroinflammation. This neuroinflammatory state has been linked to cognitive deficits and the manifestation of both positive and negative symptoms in schizophrenia (Faustmann et al., 2025) [44].
Research has highlighted that chronic low-grade inflammation, indicated by elevated levels of pro-inflammatory cytokines like IL-6 and TNF-α, is present in individuals with schizophrenia. This inflammatory profile is thought to disrupt the blood-brain barrier and alter neurotransmitter systems, particularly dopaminergic and glutamatergic pathways, which are critical for mood regulation and cognitive function (Huang et al., 2025) [42].
Moreover, microglial dysfunction is not merely a consequence of the disorder but may also serve as a potential biomarker for the disease. Studies utilizing neuroimaging and postmortem brain analyses have demonstrated that microglial activation correlates with structural brain abnormalities, including reductions in gray and white matter volumes (Laskaris et al., 2016) [45]. The implications of these findings suggest that neuroinflammation could contribute to the neuroprogressive nature of schizophrenia, wherein structural changes in the brain evolve over the course of the illness, often exacerbated by episodes of acute relapse (Howes & McCutcheon, 2017) [46].
In conclusion, the activation of microglia and the resultant neuroinflammatory processes are central to the neurobiological mechanisms underlying schizophrenia. These processes not only contribute to the cognitive and emotional disturbances characteristic of the disorder but also highlight potential therapeutic targets for intervention, such as anti-inflammatory strategies that could mitigate the adverse effects of microglial activation and improve clinical outcomes in patients with schizophrenia.
6 Current and Emerging Therapeutic Approaches
6.1 Pharmacological Treatments
Schizophrenia is a complex neuropsychiatric disorder characterized by a multifaceted etiology involving genetic, environmental, neurodevelopmental, and neurochemical factors. The neurobiological mechanisms underlying schizophrenia are intricate, encompassing dysregulation of neurotransmitter systems, neuroinflammatory processes, and alterations in neurogenesis and synaptic plasticity.
The pathophysiology of schizophrenia is often attributed to an imbalance in neurotransmitter systems, particularly involving dopamine, glutamate, and serotonin. The dopamine hypothesis remains a cornerstone of understanding schizophrenia, suggesting that hyperactivity in dopaminergic pathways contributes to positive symptoms such as hallucinations and delusions. However, the effectiveness of current antipsychotic treatments, which primarily target dopamine D2 receptors, is limited, especially concerning negative and cognitive symptoms[47][48].
Neuroinflammation has emerged as a critical factor in the pathogenesis of schizophrenia. Studies indicate that pro-inflammatory cytokines play a significant role in neuroinflammatory responses that can lead to synaptic dysfunction and neuronal injury. Mechanisms involving the NLRP3 inflammasome, NF-κB signaling, and MAPK/ERK pathways have been implicated in exacerbating cognitive and negative symptoms through oxidative stress and neuronal impairment[2][49]. The interaction between neuroinflammation and neurogenesis is also crucial, as inflammatory processes can disrupt the normal development of neural circuits, contributing to the disorder's progression[48].
Furthermore, epigenetic factors are increasingly recognized for their role in schizophrenia. Environmental influences, such as maternal infection and stress, can lead to epigenetic modifications that affect gene expression and contribute to the disorder's etiology. These modifications can alter synaptic and metabolic gene expression, impacting neuronal function and behavior[5][50]. The integration of genetic, epigenetic, and environmental data is essential for a comprehensive understanding of schizophrenia and its treatment[51].
Current pharmacological treatments primarily focus on antipsychotic medications that target dopaminergic and serotonergic systems. While these medications are effective in managing positive symptoms, their impact on negative and cognitive symptoms is often inadequate, highlighting the need for novel therapeutic approaches[52]. Emerging strategies include targeting neuroinflammatory pathways, utilizing agents that affect glutamatergic and GABAergic systems, and exploring the role of the gut microbiome in schizophrenia[53][54].
Recent advances have led to the development of novel pharmacological agents, such as lumateperone and xanomeline, which act through mechanisms beyond dopamine antagonism[55]. Additionally, the potential of anti-inflammatory agents as adjunct therapies is being explored, aiming to enhance the overall efficacy of treatment regimens[48].
In conclusion, the neurobiological mechanisms of schizophrenia are multifaceted, involving a complex interplay of neurotransmitter dysregulation, neuroinflammation, and epigenetic modifications. The limitations of current pharmacological treatments underscore the necessity for ongoing research to identify and develop more effective therapeutic strategies that address the full spectrum of symptoms associated with this debilitating disorder.
6.2 Psychosocial Interventions
Schizophrenia is characterized by a complex interplay of neurobiological mechanisms that contribute to its diverse symptoms, which can be categorized into positive, negative, and cognitive deficits. The underlying pathophysiology is multifaceted, involving neurotransmitter dysregulation, neuroinflammatory responses, and neurodevelopmental abnormalities.
One prominent mechanism is the dysregulation of neurotransmitter systems, particularly involving dopamine and glutamate. The dopamine hypothesis has long been a cornerstone of schizophrenia research, suggesting that excessive dopaminergic activity in certain brain regions contributes to positive symptoms such as hallucinations and delusions. However, current pharmacological treatments primarily targeting dopamine receptors have shown limited efficacy in addressing negative symptoms and cognitive deficits, highlighting the need for a broader understanding of the disorder's etiology (McCutcheon et al., 2020; Muszyński et al., 2025).
Recent studies have expanded the focus beyond traditional neurotransmitter models to include neuroinflammation and oxidative stress as critical factors in schizophrenia. Elevated levels of pro-inflammatory cytokines, such as IL-6 and TNF-α, and aberrant microglial activation have been implicated in neuronal dysfunction and cognitive impairments associated with the disorder (Huang et al., 2025). Additionally, the involvement of neuroinflammatory pathways, such as the activation of the NLRP3 inflammasome and the kynurenine pathway, suggests that inflammation may exacerbate synaptic dysfunction and cognitive deficits in schizophrenia (Datta et al., 2025).
The role of neurogenesis and neurodevelopmental processes has also gained attention. Impaired neurogenesis and aberrant developmental trajectories may contribute to the onset and progression of schizophrenia. Advances in imaging and proteomics have facilitated the exploration of these mechanisms, revealing how neuroinflammation and hormonal signaling interact with neurodevelopmental factors (So et al., 2025).
In light of these insights, emerging therapeutic approaches are being explored to target these neurobiological mechanisms. For instance, therapies that modulate neuroinflammatory responses or target glutamatergic systems are under investigation as potential adjuncts to traditional antipsychotics. The use of anti-inflammatory agents, such as minocycline, and antioxidants like N-acetylcysteine, has shown promise in mitigating cognitive deficits and enhancing overall treatment efficacy (Huang et al., 2025; Muszyński et al., 2025).
Furthermore, the gut-brain axis is being recognized as a significant area of interest, with dysbiosis of the gut microbiome potentially influencing the pathophysiology of schizophrenia. Research suggests that gut microbiota can affect brain function through immune modulation and neurotransmitter production, opening avenues for psychobiotic therapies that may complement existing pharmacological treatments (Munawar et al., 2021).
Despite these advancements, the translation of experimental findings into effective clinical applications remains challenging. Future research should prioritize biomarker-driven approaches and precision medicine to optimize individualized treatment outcomes, integrating both pharmacological and psychosocial interventions to enhance the quality of life for individuals with schizophrenia (Begni et al., 2025; Wu et al., 2021).
Overall, the evolving understanding of the neurobiological mechanisms underlying schizophrenia necessitates a comprehensive approach that combines novel pharmacological strategies with psychosocial interventions to address the full spectrum of symptoms and improve patient outcomes.
6.3 Future Directions in Therapy
Schizophrenia is a complex neuropsychiatric disorder characterized by a diverse array of symptoms, including cognitive deficits, emotional disturbances, and behavioral abnormalities. The neurobiological mechanisms underlying schizophrenia are multifaceted, involving dysregulation of neurotransmitter systems, neuroinflammatory processes, and neurodevelopmental disturbances.
The dopamine hypothesis has historically dominated the understanding of schizophrenia, positing that dysregulation of dopaminergic pathways contributes to the manifestation of psychotic symptoms. However, emerging research highlights the significance of other neurotransmitter systems, including glutamate and GABA, as well as the cholinergic system, which has shown promise in therapeutic applications. For instance, therapies targeting muscarinic receptors, such as the xanomeline-trospium combination, have gained attention for their potential to alleviate symptoms while minimizing side effects (Begni et al., 2025; Datta et al., 2025).
Neuroinflammation has also been identified as a critical factor in the pathogenesis of schizophrenia. Elevated levels of pro-inflammatory cytokines and aberrant microglial activation contribute to neuronal dysfunction and synaptic impairment, which are associated with cognitive and negative symptoms of the disorder. This inflammatory response is believed to be exacerbated by environmental factors, such as infections during critical periods of neurodevelopment (Huang et al., 2025; Watanabe et al., 2010). Current research is exploring the potential of anti-inflammatory agents as adjunctive therapies to traditional antipsychotics, aiming to address the underlying neuroinflammatory processes (Huang et al., 2025).
Additionally, neurodevelopmental aspects play a crucial role in the etiology of schizophrenia. Genetic predispositions combined with environmental stressors can lead to altered neurodevelopmental trajectories, resulting in cognitive and behavioral impairments (McCutcheon et al., 2020). Recent studies have indicated that disruptions in neurogenesis and alterations in the gut microbiome may also contribute to the disorder's pathology, suggesting that gut-brain interactions could be a novel therapeutic target (Munawar et al., 2021; So et al., 2025).
Future therapeutic directions in schizophrenia treatment should focus on a more integrative approach that encompasses biomarker-driven strategies and precision medicine. This includes identifying specific biomarkers that can guide treatment decisions and the development of combination therapies that target multiple pathways simultaneously. For instance, integrating immunomodulatory therapies with traditional antipsychotics may enhance treatment efficacy and improve patient outcomes (Huang et al., 2025).
Furthermore, the exploration of novel drug candidates that act on non-dopaminergic systems is gaining momentum. These include compounds that modulate glutamatergic and GABAergic neurotransmission, as well as those that target neuroinflammatory pathways. Advancements in gene therapy and the application of nanotechnology may also provide new avenues for addressing the genetic and epigenetic underpinnings of schizophrenia (Muszyński et al., 2025; Wu et al., 2021).
In summary, the neurobiological mechanisms of schizophrenia are complex and involve a confluence of neurotransmitter dysregulation, neuroinflammation, and neurodevelopmental disturbances. Emerging therapeutic approaches are increasingly focused on targeting these multifactorial aspects, paving the way for more effective and personalized treatment strategies. Continued research into the molecular and cellular pathways involved in schizophrenia will be essential for developing innovative therapies that address the disorder's full spectrum of symptoms and improve patient quality of life.
7 Conclusion
The neurobiological mechanisms underlying schizophrenia reveal a complex interplay of genetic, neurochemical, and environmental factors that contribute to its multifaceted symptoms. Key findings from current research highlight the involvement of neurotransmitter systems, particularly dopamine, glutamate, and serotonin, in the disorder's pathophysiology. While the dopamine hypothesis remains foundational, the roles of glutamate and serotonin dysregulation, as well as neuroinflammatory processes, are increasingly recognized as critical components in understanding schizophrenia. Neuroimaging studies have elucidated structural and functional brain abnormalities, emphasizing the importance of connectivity and network dysfunction in the disorder. Furthermore, genetic and epigenetic factors play a significant role in shaping individual susceptibility to schizophrenia, with environmental influences contributing to the expression of these genetic predispositions. Future research should prioritize integrative approaches that target these diverse mechanisms, exploring novel pharmacological strategies and psychosocial interventions that address the full spectrum of symptoms. The ongoing investigation into the neurobiological underpinnings of schizophrenia is essential for developing effective and personalized treatment options that improve patient outcomes and quality of life.
References
- [1] Heike Tost;Tajvar Alam;Andreas Meyer-Lindenberg. Dopamine and psychosis: theory, pathomechanisms and intermediate phenotypes.. Neuroscience and biobehavioral reviews(IF=7.9). 2010. PMID:19559045. DOI: 10.1016/j.neubiorev.2009.06.005.
- [2] Aastha Datta;Himani Rana;Shareen Singh;Thakur Gurjeet Singh. Targeting neurochemical and immune dysregulation in schizophrenia: From molecular mechanisms to emerging therapeutic strategies.. Progress in neuro-psychopharmacology & biological psychiatry(IF=3.9). 2025. PMID:41138899. DOI: 10.1016/j.pnpbp.2025.111535.
- [3] Tomás R Guilarte;Mark Opler;Mikhail Pletnikov. Is lead exposure in early life an environmental risk factor for Schizophrenia? Neurobiological connections and testable hypotheses.. Neurotoxicology(IF=3.9). 2012. PMID:22178136. DOI: 10.1016/j.neuro.2011.11.008.
- [4] Souhel Najjar;Silky Pahlajani;Virginia De Sanctis;Joel N H Stern;Amanda Najjar;Derek Chong. Neurovascular Unit Dysfunction and Blood-Brain Barrier Hyperpermeability Contribute to Schizophrenia Neurobiology: A Theoretical Integration of Clinical and Experimental Evidence.. Frontiers in psychiatry(IF=3.2). 2017. PMID:28588507. DOI: 10.3389/fpsyt.2017.00083.
- [5] Ariel Cariaga-Martinez;Jerónimo Saiz-Ruiz;Raúl Alelú-Paz. From Linkage Studies to Epigenetics: What We Know and What We Need to Know in the Neurobiology of Schizophrenia.. Frontiers in neuroscience(IF=3.2). 2016. PMID:27242407. DOI: 10.3389/fnins.2016.00202.
- [6] Ashley L Comer;Micaël Carrier;Marie-Ève Tremblay;Alberto Cruz-Martín. The Inflamed Brain in Schizophrenia: The Convergence of Genetic and Environmental Risk Factors That Lead to Uncontrolled Neuroinflammation.. Frontiers in cellular neuroscience(IF=4.0). 2020. PMID:33061891. DOI: 10.3389/fncel.2020.00274.
- [7] Crystal C Watkins;Sarah Ramsay Andrews. Clinical studies of neuroinflammatory mechanisms in schizophrenia.. Schizophrenia research(IF=3.5). 2016. PMID:26235751. DOI: 10.1016/j.schres.2015.07.018.
- [8] Jussi Hirvonen;Jarmo Hietala. Dysfunctional brain networks and genetic risk for schizophrenia: specific neurotransmitter systems.. CNS neuroscience & therapeutics(IF=5.0). 2011. PMID:21199447. DOI: 10.1111/j.1755-5949.2010.00223.x.
- [9] Joseph T Coyle. The GABA-glutamate connection in schizophrenia: which is the proximate cause?. Biochemical pharmacology(IF=5.6). 2004. PMID:15451393. DOI: 10.1016/j.bcp.2004.07.034.
- [10] D C Ohuoha;T M Hyde;J E Kleinman. The role of serotonin in schizophrenia: an overview of the nomenclature, distribution and alterations of serotonin receptors in the central nervous system.. Psychopharmacology(IF=3.3). 1993. PMID:7831440. DOI: 10.1007/BF02245003.
- [11] Maria Baou;Vassiliki A Boumba;Petros Petrikis;Georgios Rallis;Theodore Vougiouklakis;Venetsanos Mavreas. A review of genetic alterations in the serotonin pathway and their correlation with psychotic diseases and response to atypical antipsychotics.. Schizophrenia research(IF=3.5). 2016. PMID:26644303. DOI: .
- [12] E B Binder;B Kinkead;M J Owens;C B Nemeroff. The role of neurotensin in the pathophysiology of schizophrenia and the mechanism of action of antipsychotic drugs.. Biological psychiatry(IF=9.0). 2001. PMID:11743941. DOI: 10.1016/s0006-3223(01)01211-2.
- [13] Chijioke N Egbujo;Duncan Sinclair;Chang-Gyu Hahn. Dysregulations of Synaptic Vesicle Trafficking in Schizophrenia.. Current psychiatry reports(IF=6.7). 2016. PMID:27371030. DOI: 10.1007/s11920-016-0710-5.
- [14] T D Cannon. Abnormalities of brain structure and function in schizophrenia: implications for aetiology and pathophysiology.. Annals of medicine(IF=4.3). 1996. PMID:9017112. DOI: 10.3109/07853899608999117.
- [15] Anthony O Ahmed;Peter F Buckley;Mona Hanna. Neuroimaging schizophrenia: a picture is worth a thousand words, but is it saying anything important?. Current psychiatry reports(IF=6.7). 2013. PMID:23397252. DOI: 10.1007/s11920-012-0345-0.
- [16] Nina Vanessa Kraguljac;Adrienne Carol Lahti. Neuroimaging as a Window Into the Pathophysiological Mechanisms of Schizophrenia.. Frontiers in psychiatry(IF=3.2). 2021. PMID:33776813. DOI: 10.3389/fpsyt.2021.613764.
- [17] Yuichiro Watanabe;Toshiyuki Someya;Hiroyuki Nawa. Cytokine hypothesis of schizophrenia pathogenesis: evidence from human studies and animal models.. Psychiatry and clinical neurosciences(IF=6.2). 2010. PMID:20602722. DOI: 10.1111/j.1440-1819.2010.02094.x.
- [18] Martijn P van den Heuvel;Alex Fornito. Brain networks in schizophrenia.. Neuropsychology review(IF=5.0). 2014. PMID:24500505. DOI: 10.1007/s11065-014-9248-7.
- [19] David P Gavin;Schahram Akbarian. Epigenetic and post-transcriptional dysregulation of gene expression in schizophrenia and related disease.. Neurobiology of disease(IF=5.6). 2012. PMID:22182689. DOI: 10.1016/j.nbd.2011.12.008.
- [20] M F Egan;D R Weinberger. Neurobiology of schizophrenia.. Current opinion in neurobiology(IF=5.2). 1997. PMID:9384552. DOI: 10.1016/s0959-4388(97)80092-x.
- [21] Kun Li;Liju Qian;Chenchen Zhang;Jiajia Zhang;Chuang Xue;Yuebing Zhang;Wei Deng. The entorhinal cortex and cognitive impairment in schizophrenia: A comprehensive review.. Progress in neuro-psychopharmacology & biological psychiatry(IF=3.9). 2025. PMID:39672531. DOI: 10.1016/j.pnpbp.2024.111218.
- [22] G W Roberts. Schizophrenia: a neuropathological perspective.. The British journal of psychiatry : the journal of mental science(IF=7.6). 1991. PMID:2015455. DOI: 10.1192/bjp.158.1.8.
- [23] Yuan Zhou;Lingzhong Fan;Chenxiang Qiu;Tianzi Jiang. Prefrontal cortex and the dysconnectivity hypothesis of schizophrenia.. Neuroscience bulletin(IF=5.8). 2015. PMID:25761914. DOI: 10.1007/s12264-014-1502-8.
- [24] Alex Fornito;Edward T Bullmore. Reconciling abnormalities of brain network structure and function in schizophrenia.. Current opinion in neurobiology(IF=5.2). 2015. PMID:25238608. DOI: .
- [25] Katherine H Karlsgodt;Daqiang Sun;Amy M Jimenez;Evan S Lutkenhoff;Rachael Willhite;Theo G M van Erp;Tyrone D Cannon. Developmental disruptions in neural connectivity in the pathophysiology of schizophrenia.. Development and psychopathology(IF=3.7). 2008. PMID:18838043. DOI: 10.1017/S095457940800062X.
- [26] Ye Lv;Lin Wen;Wen-Juan Hu;Chong Deng;Hui-Wen Ren;Ya-Nan Bao;Bo-Wei Su;Ping Gao;Zi-Yue Man;Yi-Yang Luo;Cheng-Jie Li;Zhi-Xin Xiang;Bing Wang;Zhi-Lin Luan. Schizophrenia in the genetic era: a review from development history, clinical features and genomic research approaches to insights of susceptibility genes.. Metabolic brain disease(IF=3.5). 2024. PMID:37542622. DOI: 10.1007/s11011-023-01271-x.
- [27] Masaki Nishioka;Miki Bundo;Kiyoto Kasai;Kazuya Iwamoto. DNA methylation in schizophrenia: progress and challenges of epigenetic studies.. Genome medicine(IF=11.2). 2012. PMID:23234572. DOI: 10.1186/gm397.
- [28] Bart P F Rutten;Jonathan Mill. Epigenetic mediation of environmental influences in major psychotic disorders.. Schizophrenia bulletin(IF=4.8). 2009. PMID:19783603. DOI: 10.1093/schbul/sbp104.
- [29] Behnaz Khavari;Murray J Cairns. Epigenomic Dysregulation in Schizophrenia: In Search of Disease Etiology and Biomarkers.. Cells(IF=5.2). 2020. PMID:32764320. DOI: 10.3390/cells9081837.
- [30] Anil Srivastava;Oluwagbenga Dada;Jessica Qian;Nzaar Al-Chalabi;Ali Bani Fatemi;Philip Gerretsen;Ariel Graff;Vincenzo De Luca. Epigenetics of Schizophrenia.. Psychiatry research(IF=3.9). 2021. PMID:34638051. DOI: 10.1016/j.psychres.2021.114218.
- [31] Thabo Magwai;Khanyiso Bright Shangase;Fredrick Otieno Oginga;Bonginkosi Chiliza;Thabisile Mpofana;Khethelo Richman Xulu. DNA Methylation and Schizophrenia: Current Literature and Future Perspective.. Cells(IF=5.2). 2021. PMID:34831111. DOI: 10.3390/cells10112890.
- [32] Kimberly R Shorter;Brooke H Miller. Epigenetic mechanisms in schizophrenia.. Progress in biophysics and molecular biology(IF=4.5). 2015. PMID:25958205. DOI: .
- [33] Jacob Peedicayil. Genome-Environment Interactions and Psychiatric Disorders.. Biomedicines(IF=3.9). 2023. PMID:37189827. DOI: 10.3390/biomedicines11041209.
- [34] Evgeny A Ermakov;Mark M Melamud;Valentina N Buneva;Svetlana A Ivanova. Immune System Abnormalities in Schizophrenia: An Integrative View and Translational Perspectives.. Frontiers in psychiatry(IF=3.2). 2022. PMID:35546942. DOI: 10.3389/fpsyt.2022.880568.
- [35] Alexandre Vallée. Neuroinflammation in Schizophrenia: The Key Role of the WNT/β-Catenin Pathway.. International journal of molecular sciences(IF=4.9). 2022. PMID:35269952. DOI: 10.3390/ijms23052810.
- [36] Juan C Leza;Borja García-Bueno;Miquel Bioque;Celso Arango;Mara Parellada;Kim Do;Patricio O'Donnell;Miguel Bernardo. Inflammation in schizophrenia: A question of balance.. Neuroscience and biobehavioral reviews(IF=7.9). 2015. PMID:26092265. DOI: .
- [37] Norbert Müller. Inflammation in Schizophrenia: Pathogenetic Aspects and Therapeutic Considerations.. Schizophrenia bulletin(IF=4.8). 2018. PMID:29648618. DOI: 10.1093/schbul/sby024.
- [38] Akira Monji;Takahiro A Kato;Yoshito Mizoguchi;Hideki Horikawa;Yoshihiro Seki;Mina Kasai;Yusuke Yamauchi;Shigeto Yamada;Shigenobu Kanba. Neuroinflammation in schizophrenia especially focused on the role of microglia.. Progress in neuro-psychopharmacology & biological psychiatry(IF=3.9). 2013. PMID:22192886. DOI: .
- [39] Norbert Müller;Elif Weidinger;Bianka Leitner;Markus J Schwarz. The role of inflammation in schizophrenia.. Frontiers in neuroscience(IF=3.2). 2015. PMID:26539073. DOI: 10.3389/fnins.2015.00372.
- [40] Mikhail Melnikov;Anna Lopatina;Anastasiya Sviridova;Dmitriy Chugunov;Theofanis Vavilis;Vasileios-Periklis Stamatellos;Eleni Stamoula;Marco Cosentino. The role of neuroinflammation in schizophrenia: Focus on Th17 cells functions.. Asian journal of psychiatry(IF=4.5). 2025. PMID:40683090. DOI: 10.1016/j.ajp.2025.104629.
- [41] Monojit Debnath;Michael Berk. Th17 pathway-mediated immunopathogenesis of schizophrenia: mechanisms and implications.. Schizophrenia bulletin(IF=4.8). 2014. PMID:24711545. DOI: 10.1093/schbul/sbu049.
- [42] Hao Huang;Zijie Luo;Jieshu Min;Wenjie Luo;Xujia Zhou;Changxu Wang. Targeting Neuroinflammation in Schizophrenia: A comprehensive review of mechanisms and pharmacological interventions.. International immunopharmacology(IF=4.7). 2025. PMID:40424655. DOI: 10.1016/j.intimp.2025.114910.
- [43] Sophia-Marie Hartmann;Johanna Heider;Richard Wüst;Andreas J Fallgatter;Hansjürgen Volkmer. Microglia-neuron interactions in schizophrenia.. Frontiers in cellular neuroscience(IF=4.0). 2024. PMID:38510107. DOI: 10.3389/fncel.2024.1345349.
- [44] Timo Jendrik Faustmann;Franco Corvace;Pedro M Faustmann;Fatme Seval Ismail. Influence of antipsychotic drugs on microglia-mediated neuroinflammation in schizophrenia: perspectives in an astrocyte-microglia co-culture model.. Frontiers in psychiatry(IF=3.2). 2025. PMID:40171306. DOI: 10.3389/fpsyt.2025.1522128.
- [45] L E Laskaris;M A Di Biase;I Everall;G Chana;A Christopoulos;E Skafidas;V L Cropley;C Pantelis. Microglial activation and progressive brain changes in schizophrenia.. British journal of pharmacology(IF=7.7). 2016. PMID:26455353. DOI: 10.1111/bph.13364.
- [46] O D Howes;R McCutcheon. Inflammation and the neural diathesis-stress hypothesis of schizophrenia: a reconceptualization.. Translational psychiatry(IF=6.2). 2017. PMID:28170004. DOI: 10.1038/tp.2016.278.
- [47] Robert A McCutcheon;Tiago Reis Marques;Oliver D Howes. Schizophrenia-An Overview.. JAMA psychiatry(IF=17.1). 2020. PMID:31664453. DOI: 10.1001/jamapsychiatry.2019.3360.
- [48] Miu Tsz-Wai So;Ata Ullah;Abdul Waris;Fahad A Alhumaydhi. The Etiological Role of Impaired Neurogenesis in Schizophrenia: Interactions with Inflammatory, Microbiome and Hormonal Signaling.. International journal of molecular sciences(IF=4.9). 2025. PMID:41097077. DOI: 10.3390/ijms26199814.
- [49] Kyoung-Sae Na;Han-Yong Jung;Yong-Ku Kim. The role of pro-inflammatory cytokines in the neuroinflammation and neurogenesis of schizophrenia.. Progress in neuro-psychopharmacology & biological psychiatry(IF=3.9). 2014. PMID:23123365. DOI: .
- [50] Schahram Akbarian. Epigenetic mechanisms in schizophrenia.. Dialogues in clinical neuroscience(IF=8.9). 2014. PMID:25364289. DOI: .
- [51] Rachana R;Harshit Devtalla;Karishma Rana;Siva Prasad Panda;Arushi Agrawal;Shreya Kadyan;Divya Jindal;Pranav Pancham;Deepshikha Yadav;Niraj Kumar Jha;Saurabh Kumar Jha;Vivek Gupta;Manisha Singh. A comprehensive update on genetic inheritance, epigenetic factors, associated pathology, and recent therapeutic intervention by gene therapy in schizophrenia.. Chemical biology & drug design(IF=3.3). 2024. PMID:37994213. DOI: 10.1111/cbdd.14374.
- [52] Józef Muszyński;Agnieszka Bienert;Rasha Wafaie Elsorady;Filip Rybakowski. New pharmacological approaches in the treatment of schizophrenia.. Pharmacological reports : PR(IF=3.8). 2025. PMID:40198498. DOI: 10.1007/s43440-025-00722-9.
- [53] Daniela Rodrigues-Amorim;Tania Rivera-Baltanás;Benito Regueiro;Carlos Spuch;María Elena de Las Heras;Raul Vázquez-Noguerol Méndez;Maria Nieto-Araujo;Carolina Barreiro-Villar;Jose Manuel Olivares;Roberto Carlos Agís-Balboa. The role of the gut microbiota in schizophrenia: Current and future perspectives.. The world journal of biological psychiatry : the official journal of the World Federation of Societies of Biological Psychiatry(IF=3.8). 2018. PMID:29383983. DOI: 10.1080/15622975.2018.1433878.
- [54] Nayla Munawar;Khansa Ahsan;Khalid Muhammad;Aftab Ahmad;Munir A Anwar;Iltaf Shah;Ahlam Khalifa Al Ameri;Fadwa Al Mughairbi. Hidden Role of Gut Microbiome Dysbiosis in Schizophrenia: Antipsychotics or Psychobiotics as Therapeutics?. International journal of molecular sciences(IF=4.9). 2021. PMID:34299291. DOI: 10.3390/ijms22147671.
- [55] Matej Ľupták;Danica Michaličková;Zdeněk Fišar;Eva Kitzlerová;Jana Hroudová. Novel approaches in schizophrenia-from risk factors and hypotheses to novel drug targets.. World journal of psychiatry(IF=3.4). 2021. PMID:34327122. DOI: 10.5498/wjp.v11.i7.277.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Schizophrenia · Neurobiological Mechanisms · Neurotransmitter Systems · Neuroinflammation · Therapeutic Strategies
© 2025 MaltSci
