Next Article in Journal
Difference between Keratinized- and Non-Keratinized-Originating Epithelium in the Process of Immune Escape of Oral Squamous Cell Carcinoma
Previous Article in Journal
Mechanotransduction Circuits in Human Pathobiology
Previous Article in Special Issue
Regional Microglial Response in Entorhino–Hippocampal Slice Cultures to Schaffer Collateral Lesion and Metalloproteinases Modulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Influence of Microglia on Neuroplasticity and Long-Term Cognitive Sequelae in Long COVID: Impacts on Brain Development and Beyond

by
Luana da Silva Chagas
1,2,3 and
Claudio Alberto Serfaty
1,2,3,*
1
Program of Neuroscience, Department of Neurobiology, Institute of Biology, Federal Fluminense University, Niterói 24210-201, Rio de Janeiro, Brazil
2
National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
3
Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(7), 3819; https://doi.org/10.3390/ijms25073819
Submission received: 1 March 2024 / Revised: 26 March 2024 / Accepted: 27 March 2024 / Published: 29 March 2024
(This article belongs to the Special Issue Recent Advances in Microglia Research)

Abstract

:
Microglial cells, the immune cells of the central nervous system, are key elements regulating brain development and brain health. These cells are fully responsive to stressors, microenvironmental alterations and are actively involved in the construction of neural circuits in children and the ability to undergo full experience-dependent plasticity in adults. Since neuroinflammation is a known key element in the pathogenesis of COVID-19, one might expect the dysregulation of microglial function to severely impact both functional and structural plasticity, leading to the cognitive sequelae that appear in the pathogenesis of Long COVID. Therefore, understanding this complex scenario is mandatory for establishing the possible molecular mechanisms related to these symptoms. In the present review, we will discuss Long COVID and its association with reduced levels of BDNF, altered crosstalk between circulating immune cells and microglia, increased levels of inflammasomes, cytokines and chemokines, as well as the alterations in signaling pathways that impact neural synaptic remodeling and plasticity, such as fractalkines, the complement system, the expression of SIRPα and CD47 molecules and altered matrix remodeling. Together, these complex mechanisms may help us understand consequences of Long COVID for brain development and its association with altered brain plasticity, impacting learning disabilities, neurodevelopmental disorders, as well as cognitive decline in adults.

1. Introduction

Neuroinflammation is mediated by reactive microglia and astrocytes in response to peripheral immune cell trafficking and plays a significant role in pathophysiological mechanisms of the acute phase of COVID-19 [1,2]. While astrocytes, but not microglia, are identified as the primary target and viral reservoir during SARS-CoV-2 infection [3], disruptions in cerebral homeostasis, including the potential of vascular damage, can alter microglial functional properties. Microglia dysregulation has implications for development and neuroplasticity across different life stages, including critical periods of neural circuitry development and the maintenance of healthy synapses in adults (reviewed in [4]. Understanding the neural damage induced by viral infections during the acute phase is crucial for unraveling the mechanisms behind ‘Long COVID’ that lead to abnormal conditions including cognitive and neuropsychiatric disorders.
Long COVID, also termed Post-Acute Sequelae of COVID-19 (PASC), is a multisystem syndrome that encompasses a variety of new, recurrent or continuous symptomatology that persist after an acute SARS-CoV-2 infection. Neurological symptoms may vary in duration, from 30 days up to weeks or several months without an alternative diagnosis [5,6,7,8]. Approximately 80% of hospitalized COVID-19 patients exhibit neurological symptoms, correlating with an increased risk of mortality. The extent of this association with late neurological manifestations remains unclear [9].
Long-term neurological complications have been seen in other respiratory infections, such as in cases of severe acute respiratory syndrome coronavirus (SARS) [10] and Middle East respiratory syndrome coronavirus (MERS) [11]. A 4-year follow-up study revealed that, despite physical recovery, certain psychiatric conditions, such as PTSD, depression, panic disorder and obsessive–compulsive disorder, along with chronic fatigue, persisted in SARS infection [12]. Furthermore, to this day, there is still no clear evidence regarding the cause of post-encephalitic parkinsonism symptoms emerging from the lethargic encephalitis (“the sleepy sickness”) that affected over one million people during the 1918 Spanish flu pandemic caused by influenza A (H1N1) [13,14]. More recently in history, a similar condition of parkinsonism was also reported in survivors of avian flu [15], and, more recently, of COVID-19 [16,17,18]. Despite studies being limited to raising evidence of a direct causal association, there is a clear need for more robust research on the matter.
A recent cohort assessed the increased risk of neurological, cognitive and mental health complications up to 1-year post-acute COVID-19, even in non-hospitalized patients. These included cerebrovascular issues, cognitive disorders like brain fog and Alzheimer’s disease, drowsiness, peripheral nervous system disorders, migraines, seizures, sensory alterations (persistent anosmia and/or ageusia, tinnitus, vision impairments), musculoskeletal disorders, Guillain–Barré syndrome, encephalitis, anxiety, depression, psychotic disorders and PTSD [19,20]. Such complications have been associated with signs of neurodegeneration [21], neuroinflammation [22], and demyelination [23], but little is discussed about the impairments associated with the abnormal neuroplasticity mechanisms that may be also involved.
In children, late manifestations of SARS-CoV-2 and its variants can induce symptoms such as headaches, fatigue, palpitations, anosmia, difficulty concentrating and insomnia. These symptoms may persist even in silent or mild cases of an acute COVID-19 infection [24], and may contribute to the neglect of potential risks of sequelae, particularly neurological/psychiatric sequelae, associated with Long COVID. Besides the nonspecific viral symptoms, the increased vulnerability to stressors induced by the pandemic (undernutrition, social isolation, sick family members) during the early stages of brain development may worsen cases of pediatric Long COVID, amplifying neuropsychiatric impacts [25]. Hence, establishing the prevalence of pediatric ‘Long COVID’ is essential for assessing risks related to delays in childhood neurodevelopment, academic performance and potential long-term medical and neuropsychiatric outcomes.
The present non-systematic review intends to discuss the central role of microglia in orchestrating the long-term outcomes resulting from the acute phase of COVID-19, focusing on mechanisms associated with neuroplasticity and abnormal synaptic pruning. These impairments may pose a greater risk and increase vulnerability to the development of neurological and neuropsychiatric symptoms of Long COVID, especially during childhood. We will also review the key mechanisms involved in microglial-dependent neuroplasticity that might be useful for establishing strategies for the rehabilitation of Long COVID symptoms.

2. Neuroinflammation Mechanism during the Pathogenesis of COVID-19

During COVID-19 pathogenesis, much of the mechanisms involved in neuroinflammation result from systemic inflammation that blunts monoamine neurotransmission, decreases trophic factors and activates both astrocytes and microglia. Beyond the phagocytosis of damaged cells, activated glia release inflammatory mediators result in an excess of glutamate, the activation of the quinolinic acid pathway, pro-inflammatory cytokines expression, such as TNF-α and interleukins, and abnormal complement cascade activation [26,27]. Elevated levels of glutamate and NMDA receptors impact neuronal activity and excitotoxicity, potentially causing alterations in learning, memory and neuroplasticity, along with hallucinations and nightmares that could exacerbate or establish a new onset of Long COVID neurological symptoms (reviewed in [28,29]).
In the case of COVID-19, the pathophysiological process is initiated by three main factors inherent to this disease: peripheral cytokine storm, vascular hyperpermeability and the dysregulation of the renin–angiotensin system in the CNS (discussed in [30]. In summary, the pathobiological process begins with the virus reaching the ACE-2 enzyme receptors that are widely distributed in all organs, including neural tissue. The downregulation of ACE2 and recognition of the viral Spike glycoprotein by cells of the innate immune system establish local inflammation through the cytokine storm, and this varies according to the viral entry route (respiratory, gastrointestinal, etc.). This, in turn, is amplified by other immune cells (neutrophils, effector T cells, monocytes and macrophages), adding cytokines such as interleukin-6 (IL-6), interleukin-1b (IL-1β) and tumor necrosis factor-alpha (TNF-α) to the inflammatory process [31,32].
Among the two debated routes of SARS-CoV-2 entry into the brain, we have the hematogenous route [33], including a transcellular pathway by which SARS-CoV-2 can cross the BBB [34], and the axonal transport from olfactory sensory neurons [35]. In an attempt to investigate the cause of anosmia, researchers failed to find evidence of viral infection in these neurons. Clinical studies revealed that support cells, not neurons, are the primary targets of SARS-CoV-2 in the upper airways, based on post-mortem tissue analyses of individuals who died from COVID-19 complications [36].
Nevertheless, anatomically, the olfactory bulb could be a convenient route for the virus to reach the hippocampus, a structure associated with cognitive processing, learning and short-term memory, which potentially justifies the possibility of an accelerated cognitive decline in individuals who are more susceptible to respiratory infections [37]. For instance, studies in hamsters that analyzed the impact of SARS-CoV-2 infection on the olfactory pathway observed an incomplete recovery of sensory neurons, prolonged glial activation in the olfactory bulb and a reduced dendritic spine density in the hippocampus after the acute phase of infection, supporting the long-lasting olfactory and cognitive effects observed in Long COVID [38].
Despite the existence of a selective blood–brain barrier (BBB), the central nervous system (CNS) is in constant interaction with the peripheral immune system through the choroid plexus and circumventricular organs/regions, which are also possible entry routes for the virus into the CNS, along with the olfactory epithelium and the BBB itself, damaged by systemic inflammation (reviewed in [30]. BBB damage is correlated with the neurological sequelae of COVID-19 due to the downregulation of endothelial ACE2 receptors. BBB disruption, in association with hyperinflammation, results in endothelins that promote the extravasation of pro-inflammatory cells and activate cytokine cascades and complement activation in the brain parenchyma, contributing to vascular fragility and a hypercoagulable state [39,40,41].
The association between neuroinflammation and the neurological sequelae of Long COVID has been supported by the work of Yang and colleagues, demonstrating that subpopulations of microglia and astrocytes associated with COVID-19 share characteristics with pathological states previously observed in human neurodegenerative diseases [2,42,43]. The molecular disruption related to COVID-19 overlaps with those found in chronic neurological diseases, where genetic variants associated with cognitive decline, schizophrenia and depression reside [2]. A longitudinal study detected that certain cytokines, such as IL-1β, IL-4 and IL-6, and markers of neuronal dysfunction, such as beta-amyloid protein, total tau, p-T181-tau, light chain neurofilament and neurogranin, a marker of neuronal extracellular vesicles, remain elevated in the plasma of patients recovering from COVID-19 who self-reported neurological issues around 1 to 3 months after the initial infection [44]. TNF-α has also been shown with increased plasma levels in Long COVID patients [45]. This suggests that the neuroinflammatory processes during initial SARS-CoV-2 virus infection induce sustained reprogramming in the phenotype of CNS cells contributing to peripheral inflammation, neuroinflammation, neurodegeneration and persistent systemic effects after COVID-19 and determine the neurological symptoms observed in Long COVID.
Besides this, much evidence indicates that human neurons are less susceptible to SARS-CoV-2 infection compared to astrocytes, recognized as a primary target for secondary CNS infection and a viral reservoir [3,46]. While microglia are key players in inflammatory processes in neurological diseases [26], there is limited evidence of direct SARS-CoV-2 infection in these cells.
Nevertheless, microglia remain crucial in the broader context of neuroinflammatory responses in Long COVID, alongside neuroimmune and neuroplastic reactions. For instance, the NLRP3 inflammasome, implicated in neuroinflammation, modulates neuroplasticity. Studies in mice show that the pharmacological inhibition of the NLRP3 inflammasome prevents synaptic failure and promotes long-term potentiation (LTP) [47]. Moreover, its inactivation also promotes antidepressant effects by neuroplasticity enhancement [48]. Preclinical evidence suggests that SARS-CoV-2 activates the NLRP3 inflammasome, potentially contributing to the emergence or persistence of neurological and psychiatric symptoms through altered neuroplastic mechanisms [49]. Additionally, the elevated ATP levels induced by SARS-CoV-2 may also stimulate the NLRP3 inflammasome through P2X7 receptor hyperactivation, primarily expressed in microglia and astrocytes and associated with neuroinvasive and neuroinflammatory processes in psychiatric and neurodegenerative diseases [50]. Therefore, P2X7 and NLRP3 present promising therapeutic avenues to be explored for the treatment of neurological complications in COVID-19 patients.

3. Microglial and Neuroplasticity: Impact on Neurological Outcomes of Long COVID

Microglia play a fundamental role in CNS plasticity. They are actively enrolled in synaptic selection and the activity-dependent reorganization of neural circuits throughout development [4]. Among various physiological functions, microglia promote the formation [51], maturation [52] and selective elimination of immature synapses [53], a basic requirement for proper brain development. Microglia are actively involved in neuroplasticity; they have been shown to regulate extracellular matrix remodeling [54] and to be necessary for use-dependent cortical plasticity [55,56].
As the CNS matures, microglia monitor any threats to brain homeostasis through their “sensome” and processes and are capable of sensing molecular changes in the microenvironment and neuronal activity [57,58]. When the environment challenges the nervous system (e.g., brain injury, infection, alcohol exposure or dietary restrictions), microglia transit between dynamic reactive phenotypes and modify their density, morphology and molecular signature, which results in the proper tuning of their function [59]. For instance, early malnutrition [60], early life stress or maternal immune activation alter the microglial role in synaptic pruning and, thus, plasticity, impacting social behavior [61] and sensory neural processing [62].
Thus, as a primary strategy for homeostasis maintenance, microglia permanently assume the role of making any necessary adjustments for the construction or restoration of neural function. In environmental CNS imbalance, microglia respond through an extensive system of molecular recognition (chemokines, cytokines, trophic factors) and interact bidirectionally with neural and immune cells to influence their response. Therefore, microglial dysfunction, marked by a loss of physiological functions or an excessive inflammatory response, implies a higher risk of neurodevelopmental disorders such as autism and schizophrenia [4].
In humans, a comprehensive longitudinal study using the UK Biobank dataset detected significant gray matter loss within approximately 6 months between imaging sessions, with a reduction in the thickness and cortical volume of the left hemisphere [63]. This loss occurred in areas such as the anterior parahippocampal gyrus (episodic memory), lateral orbitofrontal cortex (secondary olfactory cortex) and superior insula (anxiety behavior). Additionally, when hospitalization was considered as a variable, alterations were detected in areas associated with smell, emotion, memory and learning, such as the left cingulate cortex, right hippocampus and right amygdala, related to anxiety behavior. Interestingly, most of the participants experienced only mild symptoms of COVID-19, even though the authors detected an accelerated reduction in whole-brain volume and more pronounced cognitive declines associated with heightened atrophy of a cognitive lobule in the cerebellum (crus II), among other long-lasting deleterious effects on brain structure and function, when compared to the control group [63]. This study provides a distinctive perspective on COVID-19-related changes in the brain structure, shedding light on the interplay between neuroplasticity and the potential for neurocognitive outcomes.
In the adult brain, microglia play a crucial role in long-term plasticity and neurogenesis and are essential for learning and memory formation. Traditional neurogenic sites, like the olfactory bulb and the hippocampal dentate gyrus, continuously generate new neurons throughout life [64]. The microglia function on this matter was demonstrated when the microglial depletion led to less functional neurons in the olfactory bulb, affecting olfactory responsiveness [65]. The proper integration of new neurons into hippocampal circuits, both structurally and functionally, also relies on microglial function [66]. In homeostatic conditions, microglia interact with synapses, synchronizing local neuronal activity and influencing behavioral and cognitive functions [67]. These processes are regulated by the physiological production of inflammatory cytokines, such as interleukins and TNF-α, which contribute to adult neurogenesis and efficient axonal plasticity in response to peripheral inflammation [68,69].
Another crucial player under investigation, due its role in neuronal regulation, survival and neural plasticity, is the brain-derived neurotrophic factor (BDNF). Reductions in plasma and brain BDNF levels are common in patients with psychiatric and neurodegenerative diseases, possibly secondary to a state of chronic inflammation affecting the brain [70]. Considering that BDNF is downstream in the ACE2-Mas axis [71], the current literature discusses the potential of the SARS-CoV-2-ACE2 interaction to reduce its levels, with implications for the pathogenesis of Neuro-COVID [72]. Indeed, microglial BDNF is essential for motor learning improvement in structural, behavioral and electrophysiological impairments in animals with the conditioned depletion of this molecule [73]. Besides this, Gonzalez and colleagues have previously shown that the intrahippocampal stimulation of the immune system with pro-inflammatory cytokines decreases BDNF expression and potentially compromises the brain’s neuroplastic functions, such as neurogenesis, LTP and dendritic sprouting [74]. Moreover, longitudinal studies to delve into the investigation of the association between trophic support that imbalances and structural changes leading to neurological or neuropsychiatric manifestations in Long COVID patients are scarce. Recently, a meta-analysis assessed relevant studies that identified lower BDNF levels in patients with COVID-19 when compared to healthy controls; this dysregulation was also related to the severity of the disease and reported in a unique study that assessed Long COVID patients, possibly linking this mechanism to neurological outcomes [75].
Despite the importance of microglia in CNS homeostasis, other immune cells also seem to contribute to its physiological role [76]. In addition to the immune response and neural activity, memory and learning tasks and social behavior are modulated by the secretion of cytokines by CD4+ T lymphocytes in the meningeal spaces, the latter being associated with IFN-γ [77]. The release of IFN-γ by T lymphocytes is also implicated in synaptic elimination by microglia and cognitive sequelae, as illustrated by spatial learning deficits in an animal model of flavivirus infection [78]. Also, a subset of innate lymphocytes is involved in cortical inhibitory synapse formation with impacts on social behavior development [79]. This may have important implications in the recent context of COVID-19, where the mechanisms associated with the long-term cognitive sequelae of Long COVID are still under investigation.

4. Molecular Signals That Regulate Microglia-Dependent Neuroplasticity

Molecular cues, identified as “find-me”, eat-me” and “do not eat me” signals, regulate the proper guidance of microglia/macrophages towards phagocytosis, which is relevant for sculpting neural connections during development and, more recently, has been implicated in many neurocognitive and neuropsychiatric conditions (reviewed in [80].
In different CNS areas, it has been well established that synaptic pruning mediated by microglia is complement-dependent during postnatal development [81,82,83], with microglia being responsive to the fractalkine primarily released by neurons and endothelium [84]. Evidence in the adult brain indicates how fractalkine-mediated neuroplasticity can modulate cognitive function. The absence of Cx3CR1, the fractalkine receptor in microglia, impacts neurogenesis, weakening learning tasks [83,85]. Abnormal levels of TGF-β or CX3CR1 have also been shown to result in aberrant neuroplasticity in adulthood [85,86,87]. The immune role in cognitive function has been demonstrated in CX3CR1 KO animals, where microglia’s function in hippocampal synaptic maturation was analyzed, showing significant delays associated with the number of microglia. Subsequent studies revealed alterations in social interaction and neural connectivity in adulthood [83,88,89].
In 2020, Scott-Hewitt and colleagues showed in vivo that the synaptic exposure of phosphatidylserine (PS) in the hippocampus and retinogeniculate areas assumes a temporal dynamics correspondence with synaptic pruning, while C1q-deficient mice failed to properly refine retinogeniculate connections, suggesting a functional interaction between the complement system and PS-mediated synaptic pruning [90]. There are increasing studies in the literature assessing the implications of C1q as an early driver of synaptic loss in neurodegenerative and neuroinflammatory conditions [91,92] (reviewed in [93]). Microglial depletion or the inhibition of its phagocytic function prevents forgetting, dissociation of engram cells and complement-mediated synaptic loss, allowing mice to forget non-essential environmental cues [94]. Deficits in hippocampal neurotransmission, as well as cognitive and behavioral impairments, are also associated with a lack of C1q in adulthood [95]. Like SARS-CoV-2, residual cognitive sequelae are also present in other viruses. The West Nile virus (WNV), a neurotropic RNA virus like SARS-CoV-2, can promote the elimination of presynaptic terminals after the viral infection of adult hippocampal neurons in a complement-dependent manner, and it is associated with memory dysfunction in human studies and animal models [27].
On the other hand, CD47 signaling and its SIRPα receptor constitute a well-known example of a “don’t eat-me” signal that prevents aberrant microglia-mediated phagocytosis. CD47 is localized in more active synapses, and disruptions to either CD47 or SIRPα in knockout studies increased synaptic over-pruning in the retinogeniculate system during early development [96]. Intriguingly, it appears that the proteolytic cleavage of SIRPα by metalloproteinases (MMP) in response to neuronal activity releases an extracellular SIRPα domain, which binds to presynaptic CD47 and promotes the maturation of presynaptic terminals [97]. The regulatory role of SIRPα in synaptic pruning was evident in a model of Alzheimer’s Disease, where the authors observed that the loss of microglial SIRPα increased synaptic loss mediated by microglia engulfment and enhanced cognitive impairment, while, in human tissue, microglial SIRPα expression declines alongside the progression of Alzheimer’s disease [98]. Indeed, synaptic elimination mechanisms in adults, as evidenced by microglial dysfunction in synaptic pruning, have been suggested in studies using post-mortem tissue from COVID-19 patients [99] and neuroimaging studies showing signs of synaptic loss [63].
Although the role of neuronal activity and microglia in synaptic refinement is well established [82,100], we still do not know exactly how these two distinct mechanisms are linked and how neural activity can be translated into local cues mediating the microglial engulfment of synaptic elements. Most of the complement-based studies on Neuro COVID or Long COVID studies have, so far, focused on these molecular signals as mechanisms of virus resistance [101] and on the worsening of coagulopathies that enhances the risk of Long COVID [102]. Up to this point, these findings underscore the need for a deeper understanding of how viral infections can impact molecular aspects of microglial function on synaptic selection and influence neural plasticity as an underlying cause in the neurological symptoms found in Long COVID.

5. Critical Periods of Brain Development and the Risk of Long COVID in Children

The infant brain is under rapid development and is highly susceptible to environmental stressors. Understanding the pathophysiological mechanisms behind the risks associated with the acute effects of COVID-19 on the developing brain during critical periods has been debated elsewhere [59], but little is known about the long-term cognitive, behavioral and psychiatric implications related to Long COVID in children and adolescents. Besides this, COVID-19 frequently presents mild acute clinical manifestations in children, which could possibly mask the potential risks of sequelae and raise appropriate concerns for public health.
Considering that microglia actively participate in brain plasticity by sculpting neural circuits and selecting and eliminating synapses, inappropriate networks may arise as a consequence of abnormal microglial performance during critical periods of development. If not detected or reversed within this sensitive developmental window, this can lead to long-term neurodevelopmental and psychiatric disorders [103]. A preliminary case–control study in young patients detected a significant increase in microgliosis in younger COVID-19 patients compared to older ones [104]. This suggests a potentially primed, more reactive microglia response in younger patients who, despite exhibiting mild acute symptoms, might be prone to the onset of Long COVID sequelae.
Recently, a study using human organoids provided evidence that SARS-CoV-2 infection may lead to excessive, disordered and premature synaptic elimination during the course of the disease, similar to what is observed in other neurological disorders such as schizophrenia, Alzheimer’s disease and Parkinson’s disease [105]. The disruption of neural circuitry integrity through an abnormal microglia-mediated synaptic over-pruning emerges as a potential mechanism in the onset of neurological, cognitive and psychiatric symptoms in COVID-19 recovery patients.
To investigate the impact of immune activation and microglial priming on vulnerabilities to neurodevelopmental disorders, microglial-like cells with a pro-inflammatory phenotype were successfully generated from mononuclear cells derived from the umbilical cord of both exposed and unexposed mothers [106]. This approach validates the search for strategies that could help to identify neonates and children with vulnerabilities caused by SARS-CoV-2, among other viral infections. Besides this, this highlights the importance of microglial function and the risks posed by its priming in neural development and subsequent disease emergence later in life.
Regarding synaptic maturation, at the end of critical periods, the perineuronal networks (PNNs), specialized reticular formations of the compact extracellular matrix that envelop neuronal subsets and stabilize proximal synapses, can be modified by microglia sculpting [107]. Furthermore, it has been shown that microglial depletion induces increased cortical perineuronal nets and heightened neural activity in excitatory cortical neurons [108]. Also, it has been demonstrated that MMP-9 contributes to the stabilization of synapses during critical periods of development and the inhibition of MMP-9 activity in vivo resulted in an altered topographical refinement of retinocollicular connections [109]. Therefore, the dysregulation of extracellular matrix dynamics due to neuroinflammation and microglial dysfunction may disrupt critical periods and neural circuit formation, posing a risk for cognitive and psychiatric sequelae following viral infection.
Also, it has been shown in the rodent visual system that synaptic plasticity and circuitry responses to environmental modifications depend on microglial functional integrity [56]. Therefore, microglial homeostasis is a crucial issue during brain development, when synaptic adjustments are necessary for the correct processing of neural information. Since microglial homeostasis can be disrupted not only by viral infections but also by exposure to nutritional stress, alcohol consumption during pregnancy, environmental stress and lack of appropriate care during infancy, ensuring plain conditions of neural development during the first years of life seems to be necessary for the full development of cognitive functions [4].
In conclusion, neuroplasticity alterations mediated by microglial dysfunction and abnormal synaptic pruning appear as key elements in Long COVID syndrome, affecting both the developing and the adult brain (Figure 1). Therefore, future research should address strategies to reestablish appropriate synaptic plasticity, including the comprehension of molecular mechanisms involved in abnormal synaptic pruning, the signaling involved in the interaction between the complement system and the expression of molecular signals in synaptic selection as important issues for the resolution of cognitive deficits following Long COVID syndrome found in infants and adults. While Long COVID causes important but frequently time-limited alterations in adults, it is noteworthy that SARS-CoV-2 infections in children may result in possible life alterations in neural circuitry development and plasticity, impacting learning abilities and socialization throughout life.

Author Contributions

Conceptualization, L.d.S.C.; Writing, L.d.S.C. and C.A.S.; Original draft preparation, L.d.S.C.; Supervision, C.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

Claudio Serfaty FAPERJ/CNE E-26/201.004/2021.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Spudich, S.; Nath, A. Nervous system consequences of COVID-19. Science 2022, 375, 267–269. [Google Scholar] [CrossRef]
  2. Yang, A.C.; Kern, F.; Losada, P.M.; Agam, M.R.; Maat, C.A.; Schmartz, G.P.; Fehlmann, T.; Stein, J.A.; Schaum, N.; Lee, D.P.; et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature 2021, 595, 565–571. [Google Scholar] [CrossRef] [PubMed]
  3. Crunfli, F.; Carregari, V.C.; Veras, F.P.; Silva, L.S.; Nogueira, M.H.; Antunes, A.; Vendramini, P.H.; Valenca, A.G.F.; Brandao-Teles, C.; Zuccoli, G.D.S.; et al. Morphological, cellular, and molecular basis of brain infection in COVID-19 patients. Proc. Natl. Acad. Sci. USA 2022, 119, e2200960119. [Google Scholar] [CrossRef] [PubMed]
  4. Chagas, L.D.S.; Sandre, P.C.; Ribeiro, E.R.N.C.A.; Marcondes, H.; Oliveira Silva, P.; Savino, W.; Serfaty, C.A. Environmental Signals on Microglial Function during Brain Development, Neuroplasticity, and Disease. Int. J. Mol. Sci. 2020, 21, 2111. [Google Scholar] [CrossRef] [PubMed]
  5. Nalbandian, A.; Sehgal, K.; Gupta, A.; Madhavan, M.V.; McGroder, C.; Stevens, J.S.; Cook, J.R.; Nordvig, A.S.; Shalev, D.; Sehrawat, T.S.; et al. Post-acute COVID-19 syndrome. Nat. Med. 2021, 27, 601–615. [Google Scholar] [CrossRef] [PubMed]
  6. Huang, C.; Huang, L.; Wang, Y.; Li, X.; Ren, L.; Gu, X.; Kang, L.; Guo, L.; Liu, M.; Zhou, X.; et al. 6-month consequences of COVID-19 in patients discharged from hospital: A cohort study. Lancet 2021, 397, 220–232. [Google Scholar] [CrossRef] [PubMed]
  7. Reese, J.T.; Blau, H.; Casiraghi, E.; Bergquist, T.; Loomba, J.J.; Callahan, T.J.; Laraway, B.; Antonescu, C.; Coleman, B.; Gargano, M.; et al. Generalisable long COVID subtypes: Findings from the NIH N3C and RECOVER programmes. EBioMedicine 2023, 87, 104413. [Google Scholar] [CrossRef] [PubMed]
  8. Danesh, V.; Arroliga, A.C.; Bourgeois, J.A.; Boehm, L.M.; McNeal, M.J.; Widmer, A.J.; McNeal, T.M.; Kesler, S.R. Symptom Clusters Seen in Adult COVID-19 Recovery Clinic Care Seekers. J. Gen. Intern. Med. 2023, 38, 442–449. [Google Scholar] [CrossRef] [PubMed]
  9. Chou, S.H.; Beghi, E.; Helbok, R.; Moro, E.; Sampson, J.; Altamirano, V.; Mainali, S.; Bassetti, C.; Suarez, J.I.; McNett, M. Global Incidence of Neurological Manifestations Among Patients Hospitalized With COVID-19-A Report for the GCS-NeuroCOVID Consortium and the ENERGY Consortium. JAMA Netw Open 2021, 4, e2112131. [Google Scholar] [CrossRef]
  10. Hong, K.S.; Lee, K.H.; Chung, J.H.; Shin, K.C.; Choi, E.Y.; Jin, H.J.; Jang, J.G.; Lee, W.; Ahn, J.H. Clinical Features and Outcomes of 98 Patients Hospitalized with SARS-CoV-2 Infection in Daegu, South Korea: A Brief Descriptive Study. Yonsei Med. J. 2020, 61, 431–437. [Google Scholar] [CrossRef]
  11. Lee, S.M.; Kang, W.S.; Cho, A.R.; Kim, T.; Park, J.K. Psychological impact of the 2015 MERS outbreak on hospital workers and quarantined hemodialysis patients. Compr. Psychiatry 2018, 87, 123–127. [Google Scholar] [CrossRef]
  12. Lam, M.H.; Wing, Y.K.; Yu, M.W.; Leung, C.M.; Ma, R.C.; Kong, A.P.; So, W.Y.; Fong, S.Y.; Lam, S.P. Mental morbidities and chronic fatigue in severe acute respiratory syndrome survivors: Long-term follow-up. Arch. Intern. Med. 2009, 169, 2142–2147. [Google Scholar] [CrossRef] [PubMed]
  13. McCall, S.; Vilensky, J.A.; Gilman, S.; Taubenberger, J.K. The relationship between encephalitis lethargica and influenza: A critical analysis. J. Neurovirol. 2008, 14, 177–185. [Google Scholar] [CrossRef]
  14. Hoffman, L.A.; Vilensky, J.A. Encephalitis lethargica: 100 years after the epidemic. Brain 2017, 140, 2246–2251. [Google Scholar] [CrossRef] [PubMed]
  15. Henry, J.; Smeyne, R.J.; Jang, H.; Miller, B.; Okun, M.S. Parkinsonism and neurological manifestations of influenza throughout the 20th and 21st centuries. Parkinsonism Relat. Disord. 2010, 16, 566–571. [Google Scholar] [CrossRef]
  16. Merello, M.; Bhatia, K.P.; Obeso, J.A. SARS-CoV-2 and the risk of Parkinson’s disease: Facts and fantasy. Lancet Neurol. 2021, 20, 94–95. [Google Scholar] [CrossRef] [PubMed]
  17. Cohen, M.E.; Eichel, R.; Steiner-Birmanns, B.; Janah, A.; Ioshpa, M.; Bar-Shalom, R.; Paul, J.J.; Gaber, H.; Skrahina, V.; Bornstein, N.M.; et al. A case of probable Parkinson’s disease after SARS-CoV-2 infection. Lancet Neurol. 2020, 19, 804–805. [Google Scholar] [CrossRef]
  18. Mendez-Guerrero, A.; Laespada-Garcia, M.I.; Gomez-Grande, A.; Ruiz-Ortiz, M.; Blanco-Palmero, V.A.; Azcarate-Diaz, F.J.; Rabano-Suarez, P.; Alvarez-Torres, E.; de Fuenmayor-Fernandez de la Hoz, C.P.; Vega Perez, D.; et al. Acute hypokinetic-rigid syndrome following SARS-CoV-2 infection. Neurology 2020, 95, e2109–e2118. [Google Scholar] [CrossRef]
  19. Xu, E.; Xie, Y.; Al-Aly, Z. Long-term neurologic outcomes of COVID-19. Nat. Med. 2022, 28, 2406–2415. [Google Scholar] [CrossRef]
  20. Ferrando, S.J.; Lynch, S.; Ferrando, N.; Dornbush, R.; Shahar, S.; Klepacz, L. Anxiety and posttraumatic stress in post-acute sequelae of COVID-19: Prevalence, characteristics, comorbidity, and clinical correlates. Front. Psychiatry 2023, 14, 1160852. [Google Scholar] [CrossRef]
  21. Zarifkar, P.; Peinkhofer, C.; Benros, M.E.; Kondziella, D. Frequency of Neurological Diseases After COVID-19, Influenza A/B and Bacterial Pneumonia. Front. Neurol. 2022, 13, 904796. [Google Scholar] [CrossRef] [PubMed]
  22. Braga, J.; Lepra, M.; Kish, S.J.; Rusjan, P.M.; Nasser, Z.; Verhoeff, N.; Vasdev, N.; Bagby, M.; Boileau, I.; Husain, M.I.; et al. Neuroinflammation After COVID-19 With Persistent Depressive and Cognitive Symptoms. JAMA Psychiatry 2023, 80, 787–795. [Google Scholar] [CrossRef] [PubMed]
  23. Zanin, L.; Saraceno, G.; Panciani, P.P.; Renisi, G.; Signorini, L.; Migliorati, K.; Fontanella, M.M. SARS-CoV-2 can induce brain and spine demyelinating lesions. Acta Neurochir. 2020, 162, 1491–1494. [Google Scholar] [CrossRef] [PubMed]
  24. Buonsenso, D.; Munblit, D.; De Rose, C.; Sinatti, D.; Ricchiuto, A.; Carfi, A.; Valentini, P. Preliminary evidence on long COVID in children. Acta Paediatr. 2021, 110, 2208–2211. [Google Scholar] [CrossRef] [PubMed]
  25. de Figueiredo, C.S.; Sandre, P.C.; Portugal, L.C.L.; Mazala-de-Oliveira, T.; da Silva Chagas, L.; Raony, I.; Ferreira, E.S.; Giestal-de-Araujo, E.; Dos Santos, A.A.; Bomfim, P.O. COVID-19 pandemic impact on children and adolescents’ mental health: Biological, environmental, and social factors. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 106, 110171. [Google Scholar] [CrossRef] [PubMed]
  26. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Munch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
  27. Vasek, M.J.; Garber, C.; Dorsey, D.; Durrant, D.M.; Bollman, B.; Soung, A.; Yu, J.; Perez-Torres, C.; Frouin, A.; Wilton, D.K.; et al. A complement-microglial axis drives synapse loss during virus-induced memory impairment. Nature 2016, 534, 538–543. [Google Scholar] [CrossRef] [PubMed]
  28. Boldrini, M.; Canoll, P.D.; Klein, R.S. How COVID-19 Affects the Brain. JAMA Psychiatry 2021, 78, 682–683. [Google Scholar] [CrossRef] [PubMed]
  29. Roman, M.; Irwin, M.R. Novel neuroimmunologic therapeutics in depression: A clinical perspective on what we know so far. Brain Behav. Immun. 2020, 83, 7–21. [Google Scholar] [CrossRef]
  30. Murta, V.; Villarreal, A.; Ramos, A.J. Severe Acute Respiratory Syndrome Coronavirus 2 Impact on the Central Nervous System: Are Astrocytes and Microglia Main Players or Merely Bystanders? ASN Neuro 2020, 12, 1759091420954960. [Google Scholar] [CrossRef]
  31. Arunachalam, P.S.; Wimmers, F.; Mok, C.K.P.; Perera, R.; Scott, M.; Hagan, T.; Sigal, N.; Feng, Y.; Bristow, L.; Tak-Yin Tsang, O.; et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science 2020, 369, 1210–1220. [Google Scholar] [CrossRef]
  32. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e1039. [Google Scholar] [CrossRef] [PubMed]
  33. Pellegrini, L.; Albecka, A.; Mallery, D.L.; Kellner, M.J.; Paul, D.; Carter, A.P.; James, L.C.; Lancaster, M.A. SARS-CoV-2 Infects the Brain Choroid Plexus and Disrupts the Blood-CSF Barrier in Human Brain Organoids. Cell Stem Cell 2020, 27, 951–961.e955. [Google Scholar] [CrossRef]
  34. Zhang, L.; Zhou, L.; Bao, L.; Liu, J.; Zhu, H.; Lv, Q.; Liu, R.; Chen, W.; Tong, W.; Wei, Q.; et al. SARS-CoV-2 crosses the blood-brain barrier accompanied with basement membrane disruption without tight junctions alteration. Signal Transduct. Target. Ther. 2021, 6, 337. [Google Scholar] [CrossRef]
  35. Meinhardt, J.; Radke, J.; Dittmayer, C.; Franz, J.; Thomas, C.; Mothes, R.; Laue, M.; Schneider, J.; Brunink, S.; Greuel, S.; et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat. Neurosci. 2021, 24, 168–175. [Google Scholar] [CrossRef]
  36. Khan, M.; Yoo, S.J.; Clijsters, M.; Backaert, W.; Vanstapel, A.; Speleman, K.; Lietaer, C.; Choi, S.; Hether, T.D.; Marcelis, L.; et al. Visualizing in deceased COVID-19 patients how SARS-CoV-2 attacks the respiratory and olfactory mucosae but spares the olfactory bulb. Cell 2021, 184, 5932–5949.e5915. [Google Scholar] [CrossRef] [PubMed]
  37. Dolgin, E. The science behind COVID’s assault on smell. Nature 2022, 606, S5–S6. [Google Scholar] [CrossRef] [PubMed]
  38. Kishimoto-Urata, M.; Urata, S.; Kagoya, R.; Imamura, F.; Nagayama, S.; Reyna, R.A.; Maruyama, J.; Yamasoba, T.; Kondo, K.; Hasegawa-Ishii, S.; et al. Prolonged and extended impacts of SARS-CoV-2 on the olfactory neurocircuit. Sci. Rep. 2022, 12, 5728. [Google Scholar] [CrossRef]
  39. Najjar, S.; Najjar, A.; Chong, D.J.; Pramanik, B.K.; Kirsch, C.; Kuzniecky, R.I.; Pacia, S.V.; Azhar, S. Central nervous system complications associated with SARS-CoV-2 infection: Integrative concepts of pathophysiology and case reports. J. Neuroinflamm. 2020, 17, 231. [Google Scholar] [CrossRef]
  40. Wang, T.; Chen, R.; Liu, C.; Liang, W.; Guan, W.; Tang, R.; Tang, C.; Zhang, N.; Zhong, N.; Li, S. Attention should be paid to venous thromboembolism prophylaxis in the management of COVID-19. Lancet Haematol. 2020, 7, e362–e363. [Google Scholar] [CrossRef]
  41. Ranucci, M.; Ballotta, A.; Di Dedda, U.; Baryshnikova, E.; Dei Poli, M.; Resta, M.; Falco, M.; Albano, G.; Menicanti, L. The procoagulant pattern of patients with COVID-19 acute respiratory distress syndrome. J. Thromb. Haemost. 2020, 18, 1747–1751. [Google Scholar] [CrossRef] [PubMed]
  42. Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B.; et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell 2017, 169, 1276–1290.e1217. [Google Scholar] [CrossRef] [PubMed]
  43. Mathys, H.; Davila-Velderrain, J.; Peng, Z.; Gao, F.; Mohammadi, S.; Young, J.Z.; Menon, M.; He, L.; Abdurrob, F.; Jiang, X.; et al. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 2019, 570, 332–337. [Google Scholar] [CrossRef]
  44. Sun, B.; Tang, N.; Peluso, M.J.; Iyer, N.S.; Torres, L.; Donatelli, J.L.; Munter, S.E.; Nixon, C.C.; Rutishauser, R.L.; Rodriguez-Barraquer, I.; et al. Characterization and Biomarker Analyses of Post-COVID-19 Complications and Neurological Manifestations. Cells 2021, 10, 386. [Google Scholar] [CrossRef] [PubMed]
  45. Saito, S.; Shahbaz, S.; Luo, X.; Osman, M.; Redmond, D.; Cohen Tervaert, J.W.; Li, L.; Elahi, S. Metabolomic and immune alterations in long COVID patients with chronic fatigue syndrome. Front. Immunol. 2024, 15, 1341843. [Google Scholar] [CrossRef] [PubMed]
  46. Andrews, M.G.; Mukhtar, T.; Eze, U.C.; Simoneau, C.R.; Ross, J.; Parikshak, N.; Wang, S.; Zhou, L.; Koontz, M.; Velmeshev, D.; et al. Tropism of SARS-CoV-2 for human cortical astrocytes. Proc. Natl. Acad. Sci. USA 2022, 119, e2122236119. [Google Scholar] [CrossRef] [PubMed]
  47. Qi, Y.; Klyubin, I.; Cuello, A.C.; Rowan, M.J. NLRP3-dependent synaptic plasticity deficit in an Alzheimer’s disease amyloidosis model in vivo. Neurobiol. Dis. 2018, 114, 24–30. [Google Scholar] [CrossRef] [PubMed]
  48. Xia, B.; Tong, Y.; Xia, C.; Chen, C.; Shan, X. alpha-Cyperone Confers Antidepressant-Like Effects in Mice via Neuroplasticity Enhancement by SIRT3/ROS Mediated NLRP3 Inflammasome Deactivation. Front. Pharmacol. 2020, 11, 577062. [Google Scholar] [CrossRef] [PubMed]
  49. Albornoz, E.A.; Amarilla, A.A.; Modhiran, N.; Parker, S.; Li, X.X.; Wijesundara, D.K.; Aguado, J.; Zamora, A.P.; McMillan, C.L.D.; Liang, B.; et al. SARS-CoV-2 drives NLRP3 inflammasome activation in human microglia through spike protein. Mol. Psychiatry 2023, 28, 2878–2893. [Google Scholar] [CrossRef]
  50. Ribeiro, D.E.; Oliveira-Giacomelli, A.; Glaser, T.; Arnaud-Sampaio, V.F.; Andrejew, R.; Dieckmann, L.; Baranova, J.; Lameu, C.; Ratajczak, M.Z.; Ulrich, H. Hyperactivation of P2X7 receptors as a culprit of COVID-19 neuropathology. Mol. Psychiatry 2021, 26, 1044–1059. [Google Scholar] [CrossRef]
  51. Kim, H.J.; Cho, M.H.; Shim, W.H.; Kim, J.K.; Jeon, E.Y.; Kim, D.H.; Yoon, S.Y. Deficient autophagy in microglia impairs synaptic pruning and causes social behavioral defects. Mol. Psychiatry 2017, 22, 1576–1584. [Google Scholar] [CrossRef] [PubMed]
  52. Miyamoto, A.; Wake, H.; Ishikawa, A.W.; Eto, K.; Shibata, K.; Murakoshi, H.; Koizumi, S.; Moorhouse, A.J.; Yoshimura, Y.; Nabekura, J. Microglia contact induces synapse formation in developing somatosensory cortex. Nat. Commun. 2016, 7, 12540. [Google Scholar] [CrossRef] [PubMed]
  53. Weinhard, L.; di Bartolomei, G.; Bolasco, G.; Machado, P.; Schieber, N.L.; Neniskyte, U.; Exiga, M.; Vadisiute, A.; Raggioli, A.; Schertel, A.; et al. Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction. Nat. Commun. 2018, 9, 1228. [Google Scholar] [CrossRef]
  54. Nguyen, P.T.; Dorman, L.C.; Pan, S.; Vainchtein, I.D.; Han, R.T.; Nakao-Inoue, H.; Taloma, S.E.; Barron, J.J.; Molofsky, A.B.; Kheirbek, M.A.; et al. Microglial Remodeling of the Extracellular Matrix Promotes Synapse Plasticity. Cell 2020, 182, 388–403.e315. [Google Scholar] [CrossRef]
  55. Sipe, G.O.; Lowery, R.L.; Tremblay, M.E.; Kelly, E.A.; Lamantia, C.E.; Majewska, A.K. Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex. Nat. Commun. 2016, 7, 10905. [Google Scholar] [CrossRef]
  56. Ma, X.; Chen, K.; Cui, Y.; Huang, G.; Nehme, A.; Zhang, L.; Li, H.; Wei, J.; Liong, K.; Liu, Q.; et al. Depletion of microglia in developing cortical circuits reveals its critical role in glutamatergic synapse development, functional connectivity, and critical period plasticity. J. Neurosci. Res. 2020, 98, 1968–1986. [Google Scholar] [CrossRef]
  57. Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [PubMed]
  58. Hickman, S.E.; Kingery, N.D.; Ohsumi, T.K.; Borowsky, M.L.; Wang, L.C.; Means, T.K.; El Khoury, J. The microglial sensome revealed by direct RNA sequencing. Nat. Neurosci. 2013, 16, 1896–1905. [Google Scholar] [CrossRef]
  59. da Silva Chagas, L.; Sandre, P.C.; de Velasco, P.C.; Marcondes, H.; Ribeiro, E.R.N.C.A.; Barreto, A.L.; Alves Mauro, L.B.; Ferreira, J.H.; Serfaty, C.A. Neuroinflammation and Brain Development: Possible Risk Factors in COVID-19-Infected Children. Neuroimmunomodulation 2021, 28, 22–28. [Google Scholar] [CrossRef]
  60. Sandre, P.C.; da Silva Chagas, L.; de Velasco, P.C.; Galvani, R.G.; Dias Fraga, K.Y.; Tavares do Carmo, M.D.G.; Vianna, P.H.O.; Bonomo, A.C.; Serfaty, C.A. Chronic nutritional restriction of omega-3 fatty acids induces a pro-inflammatory profile during the development of the rat visual system. Brain Res. Bull. 2021, 174, 366–378. [Google Scholar] [CrossRef]
  61. Dayananda, K.K.; Ahmed, S.; Wang, D.; Polis, B.; Islam, R.; Kaffman, A. Early life stress impairs synaptic pruning in the developing hippocampus. Brain Behav. Immun. 2023, 107, 16–31. [Google Scholar] [CrossRef] [PubMed]
  62. Ren, J.; Yan, Y.; Cheng, S.; Long, J.; Zhang, H.; Wang, J.; Shen, Y.; Zhou, Y.D.; Anderson, M.P. Maternal immune activation alters visual acuity and retinogeniculate axon pruning in offspring mice. Brain Behav. Immun. 2020, 89, 518–523. [Google Scholar] [CrossRef]
  63. Douaud, G.; Lee, S.; Alfaro-Almagro, F.; Arthofer, C.; Wang, C.; McCarthy, P.; Lange, F.; Andersson, J.L.R.; Griffanti, L.; Duff, E.; et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature 2022, 604, 697–707. [Google Scholar] [CrossRef] [PubMed]
  64. Noguchi, H.; Castillo, J.G.; Nakashima, K.; Pleasure, S.J. Suppressor of fused controls perinatal expansion and quiescence of future dentate adult neural stem cells. Elife 2019, 8, e42918. [Google Scholar] [CrossRef] [PubMed]
  65. Wallace, J.; Lord, J.; Dissing-Olesen, L.; Stevens, B.; Murthy, V.N. Microglial depletion disrupts normal functional development of adult-born neurons in the olfactory bulb. Elife 2020, 9, e50531. [Google Scholar] [CrossRef]
  66. Bolos, M.; Perea, J.R.; Terreros-Roncal, J.; Pallas-Bazarra, N.; Jurado-Arjona, J.; Avila, J.; Llorens-Martin, M. Absence of microglial CX3CR1 impairs the synaptic integration of adult-born hippocampal granule neurons. Brain Behav. Immun. 2018, 68, 76–89. [Google Scholar] [CrossRef] [PubMed]
  67. Akiyoshi, R.; Wake, H.; Kato, D.; Horiuchi, H.; Ono, R.; Ikegami, A.; Haruwaka, K.; Omori, T.; Tachibana, Y.; Moorhouse, A.J.; et al. Microglia Enhance Synapse Activity to Promote Local Network Synchronization. eNeuro 2018, 5. [Google Scholar] [CrossRef] [PubMed]
  68. Belenguer, G.; Duart-Abadia, P.; Jordan-Pla, A.; Domingo-Muelas, A.; Blasco-Chamarro, L.; Ferron, S.R.; Morante-Redolat, J.M.; Farinas, I. Adult Neural Stem Cells Are Alerted by Systemic Inflammation through TNF-alpha Receptor Signaling. Cell Stem Cell 2021, 28, 285–299.e289. [Google Scholar] [CrossRef] [PubMed]
  69. Chagas, L.D.S.; Trindade, P.; Gomes, A.L.T.; Mendonca, H.R.; Campello-Costa, P.; Faria Melibeu, A.D.C.; Linden, R.; Serfaty, C.A. Rapid plasticity of intact axons following a lesion to the visual pathways during early brain development is triggered by microglial activation. Exp. Neurol. 2019, 311, 148–161. [Google Scholar] [CrossRef]
  70. Lima Giacobbo, B.; Doorduin, J.; Klein, H.C.; Dierckx, R.; Bromberg, E.; de Vries, E.F.J. Brain-Derived Neurotrophic Factor in Brain Disorders: Focus on Neuroinflammation. Mol. Neurobiol. 2019, 56, 3295–3312. [Google Scholar] [CrossRef]
  71. Kermanshahi, S.; Gholami, M.; Motaghinejad, M. Can Infection of COVID-19 Virus Exacerbate Alzheimer’s Symptoms? Hypothetic Possible Role of Angiotensin-Converting Enzyme-2/Mas/ Brain-Derived Neurotrophic Factor Axis and Tau Hyper-phosphorylation. Adv. Biomed. Res. 2020, 9, 36. [Google Scholar]
  72. Azoulay, D.; Shehadeh, M.; Chepa, S.; Shaoul, E.; Baroum, M.; Horowitz, N.A.; Kaykov, E. Recovery from SARS-CoV-2 infection is associated with serum BDNF restoration. J. Infect. 2020, 81, e79–e81. [Google Scholar] [CrossRef] [PubMed]
  73. Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., 3rd; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609. [Google Scholar] [CrossRef]
  74. Gonzalez, P.; Machado, I.; Vilcaes, A.; Caruso, C.; Roth, G.A.; Schioth, H.; Lasaga, M.; Scimonelli, T. Molecular mechanisms involved in interleukin 1-beta (IL-1beta)-induced memory impairment. Modulation by alpha-melanocyte-stimulating hormone (alpha-MSH). Brain Behav. Immun. 2013, 34, 141–150. [Google Scholar] [CrossRef]
  75. Shafiee, A.; Seighali, N.; Teymouri Athar, M.; Abdollahi, A.K.; Jafarabady, K.; Bakhtiyari, M. Levels of brain-derived neurotrophic factor (BDNF) among patients with COVID-19: A systematic review and meta-analysis. Eur. Arch. Psychiatry Clin. Neurosci. 2023. [Google Scholar] [CrossRef] [PubMed]
  76. Elmore, M.R.; Najafi, A.R.; Koike, M.A.; Dagher, N.N.; Spangenberg, E.E.; Rice, R.A.; Kitazawa, M.; Matusow, B.; Nguyen, H.; West, B.L.; et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 2014, 82, 380–397. [Google Scholar] [CrossRef]
  77. Filiano, A.J.; Xu, Y.; Tustison, N.J.; Marsh, R.L.; Baker, W.; Smirnov, I.; Overall, C.C.; Gadani, S.P.; Turner, S.D.; Weng, Z.; et al. Unexpected role of interferon-gamma in regulating neuronal connectivity and social behaviour. Nature 2016, 535, 425–429. [Google Scholar] [CrossRef] [PubMed]
  78. Garber, C.; Soung, A.; Vollmer, L.L.; Kanmogne, M.; Last, A.; Brown, J.; Klein, R.S. T cells promote microglia-mediated synaptic elimination and cognitive dysfunction during recovery from neuropathogenic flaviviruses. Nat. Neurosci. 2019, 22, 1276–1288. [Google Scholar] [CrossRef] [PubMed]
  79. Barron, J.J.; Mroz, N.M.; Taloma, S.E.; Dahlgren, M.W.; Ortiz-Carpena, J.; Dorman, L.C.; Vainchtein, I.D.; Escoubas, C.C.; Molofsky, A.B.; Molofsky, A.V. Group 2 innate lymphoid cells promote inhibitory synapse development and social behavior. bioRxiv 2023. [CrossRef]
  80. Zhu, H.; Guan, A.; Liu, J.; Peng, L.; Zhang, Z.; Wang, S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J. Neuroinflamm. 2023, 20, 223. [Google Scholar] [CrossRef]
  81. Tremblay, M.E.; Lowery, R.L.; Majewska, A.K. Microglial interactions with synapses are modulated by visual experience. PLoS Biol. 2010, 8, e1000527. [Google Scholar] [CrossRef]
  82. Schafer, D.P.; Lehrman, E.K.; Kautzman, A.G.; Koyama, R.; Mardinly, A.R.; Yamasaki, R.; Ransohoff, R.M.; Greenberg, M.E.; Barres, B.A.; Stevens, B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 2012, 74, 691–705. [Google Scholar] [CrossRef] [PubMed]
  83. Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [PubMed]
  84. Sunnemark, D.; Eltayeb, S.; Nilsson, M.; Wallstrom, E.; Lassmann, H.; Olsson, T.; Berg, A.L.; Ericsson-Dahlstrand, A. CX3CL1 (fractalkine) and CX3CR1 expression in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis: Kinetics and cellular origin. J. Neuroinflamm. 2005, 2, 17. [Google Scholar] [CrossRef]
  85. Rogers, J.T.; Morganti, J.M.; Bachstetter, A.D.; Hudson, C.E.; Peters, M.M.; Grimmig, B.A.; Weeber, E.J.; Bickford, P.C.; Gemma, C. CX3CR1 deficiency leads to impairment of hippocampal cognitive function and synaptic plasticity. J. Neurosci. 2011, 31, 16241–16250. [Google Scholar] [CrossRef]
  86. Koeglsperger, T.; Li, S.; Brenneis, C.; Saulnier, J.L.; Mayo, L.; Carrier, Y.; Selkoe, D.J.; Weiner, H.L. Impaired glutamate recycling and GluN2B-mediated neuronal calcium overload in mice lacking TGF-beta1 in the CNS. Glia 2013, 61, 985–1002. [Google Scholar] [CrossRef] [PubMed]
  87. Butovsky, O.; Jedrychowski, M.P.; Moore, C.S.; Cialic, R.; Lanser, A.J.; Gabriely, G.; Koeglsperger, T.; Dake, B.; Wu, P.M.; Doykan, C.E.; et al. Identification of a unique TGF-beta-dependent molecular and functional signature in microglia. Nat. Neurosci. 2014, 17, 131–143. [Google Scholar] [CrossRef]
  88. Zhan, Y.; Paolicelli, R.C.; Sforazzini, F.; Weinhard, L.; Bolasco, G.; Pagani, F.; Vyssotski, A.L.; Bifone, A.; Gozzi, A.; Ragozzino, D.; et al. Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior. Nat. Neurosci. 2014, 17, 400–406. [Google Scholar] [CrossRef]
  89. Paolicelli, R.C.; Gross, C.T. Microglia in development: Linking brain wiring to brain environment. Neuron Glia Biol. 2011, 7, 77–83. [Google Scholar] [CrossRef]
  90. Scott-Hewitt, N.; Perrucci, F.; Morini, R.; Erreni, M.; Mahoney, M.; Witkowska, A.; Carey, A.; Faggiani, E.; Schuetz, L.T.; Mason, S.; et al. Local externalization of phosphatidylserine mediates developmental synaptic pruning by microglia. EMBO J. 2020, 39, e105380. [Google Scholar] [CrossRef]
  91. Shi, Q.; Colodner, K.J.; Matousek, S.B.; Merry, K.; Hong, S.; Kenison, J.E.; Frost, J.L.; Le, K.X.; Li, S.; Dodart, J.C.; et al. Complement C3-Deficient Mice Fail to Display Age-Related Hippocampal Decline. J. Neurosci. 2015, 35, 13029–13042. [Google Scholar] [CrossRef]
  92. Hong, S.; Beja-Glasser, V.F.; Nfonoyim, B.M.; Frouin, A.; Li, S.; Ramakrishnan, S.; Merry, K.M.; Shi, Q.; Rosenthal, A.; Barres, B.A.; et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 2016, 352, 712–716. [Google Scholar] [CrossRef] [PubMed]
  93. Lee, J.D.; Coulthard, L.G.; Woodruff, T.M. Complement dysregulation in the central nervous system during development and disease. Semin. Immunol. 2019, 45, 101340. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, C.; Yue, H.; Hu, Z.; Shen, Y.; Ma, J.; Li, J.; Wang, X.D.; Wang, L.; Sun, B.; Shi, P.; et al. Microglia mediate forgetting via complement-dependent synaptic elimination. Science 2020, 367, 688–694. [Google Scholar] [CrossRef] [PubMed]
  95. Perez-Alcazar, M.; Daborg, J.; Stokowska, A.; Wasling, P.; Bjorefeldt, A.; Kalm, M.; Zetterberg, H.; Carlstrom, K.E.; Blomgren, K.; Ekdahl, C.T.; et al. Altered cognitive performance and synaptic function in the hippocampus of mice lacking C3. Exp. Neurol. 2014, 253, 154–164. [Google Scholar] [CrossRef] [PubMed]
  96. Lehrman, E.K.; Wilton, D.K.; Litvina, E.Y.; Welsh, C.A.; Chang, S.T.; Frouin, A.; Walker, A.J.; Heller, M.D.; Umemori, H.; Chen, C.; et al. CD47 Protects Synapses from Excess Microglia-Mediated Pruning during Development. Neuron 2018, 100, 120–134.e126. [Google Scholar] [CrossRef] [PubMed]
  97. Toth, A.B.; Terauchi, A.; Zhang, L.Y.; Johnson-Venkatesh, E.M.; Larsen, D.J.; Sutton, M.A.; Umemori, H. Synapse maturation by activity-dependent ectodomain shedding of SIRPalpha. Nat. Neurosci. 2013, 16, 1417–1425. [Google Scholar] [CrossRef]
  98. Ding, X.; Wang, J.; Huang, M.; Chen, Z.; Liu, J.; Zhang, Q.; Zhang, C.; Xiang, Y.; Zen, K.; Li, L. Loss of microglial SIRPalpha promotes synaptic pruning in preclinical models of neurodegeneration. Nat. Commun. 2021, 12, 2030. [Google Scholar] [CrossRef] [PubMed]
  99. Mukerji, S.S.; Solomon, I.H. What can we learn from brain autopsies in COVID-19? Neurosci. Lett. 2021, 742, 135528. [Google Scholar] [CrossRef]
  100. Katz, L.C.; Shatz, C.J. Synaptic activity and the construction of cortical circuits. Science 1996, 274, 1133–1138. [Google Scholar] [CrossRef]
  101. Arganaraz, G.A.; Palmeira, J.D.F.; Arganaraz, E.R. Phosphatidylserine inside out: A possible underlying mechanism in the inflammation and coagulation abnormalities of COVID-19. Cell Commun. Signal. 2020, 18, 190. [Google Scholar] [CrossRef] [PubMed]
  102. Bohan, D.; Van Ert, H.; Ruggio, N.; Rogers, K.J.; Badreddine, M.; Aguilar Briseno, J.A.; Elliff, J.M.; Rojas Chavez, R.A.; Gao, B.; Stokowy, T.; et al. Phosphatidylserine receptors enhance SARS-CoV-2 infection. PLoS Pathog. 2021, 17, e1009743. [Google Scholar] [CrossRef] [PubMed]
  103. Paolicelli, R.C.; Ferretti, M.T. Function and Dysfunction of Microglia during Brain Development: Consequences for Synapses and Neural Circuits. Front. Synaptic. Neurosci. 2017, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  104. Matschke, J.; Lahann, H.; Krasemann, S.; Altmeppen, H.; Pfefferle, S.; Galliciotti, G.; Fitzek, A.; Sperhake, J.P.; Ondruschka, B.; Busch, M.; et al. Young COVID-19 Patients Show a Higher Degree of Microglial Activation When Compared to Controls. Front. Neurol. 2022, 13, 908081. [Google Scholar] [CrossRef] [PubMed]
  105. Samudyata; Oliveira, A.O.; Malwade, S.; Rufino de Sousa, N.; Goparaju, S.K.; Gracias, J.; Orhan, F.; Steponaviciute, L.; Schalling, M.; Sheridan, S.D.; et al. SARS-CoV-2 promotes microglial synapse elimination in human brain organoids. Mol. Psychiatry 2022, 27, 3939–3950. [Google Scholar] [CrossRef] [PubMed]
  106. Sheridan, S.D.; Thanos, J.M.; De Guzman, R.M.; McCrea, L.T.; Horng, J.E.; Fu, T.; Sellgren, C.M.; Perlis, R.H.; Edlow, A.G. Umbilical cord blood-derived microglia-like cells to model COVID-19 exposure. Transl. Psychiatry 2021, 11, 179. [Google Scholar] [CrossRef] [PubMed]
  107. Crapser, J.D.; Arreola, M.A.; Tsourmas, K.I.; Green, K.N. Microglia as hackers of the matrix: Sculpting synapses and the extracellular space. Cell. Mol. Immunol. 2021, 18, 2472–2488. [Google Scholar] [CrossRef] [PubMed]
  108. Liu, Y.J.; Spangenberg, E.E.; Tang, B.; Holmes, T.C.; Green, K.N.; Xu, X. Microglia Elimination Increases Neural Circuit Connectivity and Activity in Adult Mouse Cortex. J. Neurosci. 2021, 41, 1274–1287. [Google Scholar] [CrossRef]
  109. Oliveira-Silva, P.; Jurgilas, P.B.; Trindade, P.; Campello-Costa, P.; Perales, J.; Savino, W.; Serfaty, C.A. Matrix metalloproteinase-9 is involved in the development and plasticity of retinotectal projections in rats. Neuroimmunomodulation 2007, 14, 144–149. [Google Scholar] [CrossRef]
Figure 1. Neuroplasticity impairments due to microglial dysfunction as a central mechanism for “Long COVID” neurological outcomes. Various mechanisms, including neuroimmune interactions, lead to microglial dysfunction, which directly impairs neural functions, reproducing “Long COVID” neurological and cognitive symptoms. Some mechanisms related to microglial regulation are being explored, such as the decrease in microglial BDNF levels and involvement of NLRP3 inflammasome, linked to the decrease in LTP; the dysregulated expression of cytokines, chemokines and complement system molecules that interfere with proper synaptic plasticity and induce insufficient or excessive synaptic stripping; the lack of microglia influence on matrix remodeling that promotes E/I imbalance and the impairment of neural circuit maturation; and the cross-talk between microglia and other circulating immune cells, like CD4+ T lymphocytes, which further exacerbates these effects. These mechanisms illustrate how abnormal plasticity contributes to Long COVID neurological complications, underscoring the importance of targeting them for therapeutic strategies to improve neuroplasticity.
Figure 1. Neuroplasticity impairments due to microglial dysfunction as a central mechanism for “Long COVID” neurological outcomes. Various mechanisms, including neuroimmune interactions, lead to microglial dysfunction, which directly impairs neural functions, reproducing “Long COVID” neurological and cognitive symptoms. Some mechanisms related to microglial regulation are being explored, such as the decrease in microglial BDNF levels and involvement of NLRP3 inflammasome, linked to the decrease in LTP; the dysregulated expression of cytokines, chemokines and complement system molecules that interfere with proper synaptic plasticity and induce insufficient or excessive synaptic stripping; the lack of microglia influence on matrix remodeling that promotes E/I imbalance and the impairment of neural circuit maturation; and the cross-talk between microglia and other circulating immune cells, like CD4+ T lymphocytes, which further exacerbates these effects. These mechanisms illustrate how abnormal plasticity contributes to Long COVID neurological complications, underscoring the importance of targeting them for therapeutic strategies to improve neuroplasticity.
Ijms 25 03819 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chagas, L.d.S.; Serfaty, C.A. The Influence of Microglia on Neuroplasticity and Long-Term Cognitive Sequelae in Long COVID: Impacts on Brain Development and Beyond. Int. J. Mol. Sci. 2024, 25, 3819. https://doi.org/10.3390/ijms25073819

AMA Style

Chagas LdS, Serfaty CA. The Influence of Microglia on Neuroplasticity and Long-Term Cognitive Sequelae in Long COVID: Impacts on Brain Development and Beyond. International Journal of Molecular Sciences. 2024; 25(7):3819. https://doi.org/10.3390/ijms25073819

Chicago/Turabian Style

Chagas, Luana da Silva, and Claudio Alberto Serfaty. 2024. "The Influence of Microglia on Neuroplasticity and Long-Term Cognitive Sequelae in Long COVID: Impacts on Brain Development and Beyond" International Journal of Molecular Sciences 25, no. 7: 3819. https://doi.org/10.3390/ijms25073819

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop