Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Cell. Neurosci., 27 November 2025

Sec. Non-Neuronal Cells

Volume 19 - 2025 | https://doi.org/10.3389/fncel.2025.1690400

This article is part of the Research TopicReviews in non-neuronal cells 2024 & 2025View all 7 articles

Glial interactions in the formation and plasticity of the corpus callosum

  • 1Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
  • 2Laboratory of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland

The corpus callosum (CC) is the largest interhemispheric commissure in the eutherian brain, enabling inter-hemispheric sensory integration and higher-order cognitive functions. Historically viewed through a neuron- and axon-centric lens, extensive research has established that glial cells (astrocytes, oligodendrocytes, and microglia) are essential regulators of CC ontogenesis. Astrocytic guidepost cells sculpt midline architecture and secrete axonal guidance cues; oligodendrocytes drive callosal axonal maturation and myelination; and microglia regulate their fasciculation and pruning, myelination patterns, and synaptic refinement. In addition to these cell-specific roles, coordinated bidirectional signaling between neurons and glia ensures that axon targeting, maturation, and interhemispheric integration proceed in a precisely orchestrated manner. Disruptions to these glial functions are implicated in congenital and developmental brain pathologies, including malformations and CC agenesis. This review integrates molecular, developmental, and translational insights to provide a comprehensive, mechanistic understanding of glial contributions to CC development and how their dysfunction shapes pathology.

Introduction

The corpus callosum (CC) constitutes the principal interhemispheric commissure in the brain of placental mammals and is fundamental for coordinating bilateral sensory, motor, and cognitive functions. Although the ontogeny of the CC has been historically attributed to the pathfinding behaviors of cortical projection neurons, early studies have identified non-neuronal glial structures as major players in the formation of the CC (Silver et al., 1982; Wahlsten, 1987). While intrinsic neuronal programs remain critical to this process, growing evidence from the last decades has revealed a more complex and multifactorial mechanism that highlights the vital roles of non-neuronal cell types, particularly glial cells, in orchestrating the sequence of developmental events that establish the correct formation of the CC (Katz et al., 1983; Shu and Richards, 2001; Liu et al., 2023; Gavrish et al., 2024; Rodríguez-Pérez et al., 2024).

Current evidence indicates that astrocytes, oligodendrocytes, and microglia participate actively in callosal formation and remodeling. Astrocytes located at the midline generate structural substrates such as the glial wedge and secrete guidance cues, including Slits and Netrins, which regulate axonal attraction and repulsion during midline crossing (Shu et al., 2003; Gobius et al., 2016). Glial cells also contribute to later stages of callosal development through synaptogenic functions and metabolic support. Oligodendrocyte precursor cells (OPCs) populate the callosal region during embryogenesis and initiate myelination in a tightly regulated, regionally specific, and activity-dependent manner (Simons et al., 2024). This process is governed by paracrine signaling pathways, including Sonic Hedgehog (Shh), PDGF, Wnt/β-catenin, Notch, and thyroid hormone signaling, as well as by neuron-derived ligands such as Neuregulin-1 (Calver et al., 1998; Choudhry et al., 2014; Meyer et al., 2018). Microglia, the resident immune cells of the central nervous system, contribute to axon fasciculation, the elimination of redundant or mistargeted axons, the refinement of synaptic circuits, and the regulation of oligodendrocyte dynamics (Cunningham et al., 2013; Filipello et al., 2018; Irfan et al., 2022; Lawrence et al., 2024).

By comparing the ontogenesis of the CC in humans and rodents, this review highlights both conserved and species-specific mechanisms, contextualizes developmental timelines, anatomical organization, and the spatial dynamics of axonal crossing, with a focus on the emergence of key glial structures such as the glial wedge, midline zipper glia (MZG), indusium griseum (IGG), and subcallosal sling (SCS), and their molecular signaling. Next, we elaborate on the roles of astrocytes, with emphasis on their morphogenetic contributions to neuroanatomic remodeling of the midline during fetal stages, as well as axon-guidance cues molecules (e.g., Slit/Robo, Netrin, Draxin pathways), and regulatory functions in synaptic refinement and metabolic homeostasis. Then, we focus on oligodendrocytes, detailing the sequential phases of OPCs proliferation, migration, and differentiation, and highlighting the influence of signaling networks in regulating callosal myelination in the late phase of callosal maturation. Next, we examine the multifaceted functions of microglia in callosal development, ranging from the regulation of axonal fasciculation and neuronal pruning to the phagocytosis of excess OPCs and modulation of local inflammatory environments. Thereafter, we explore how cellular actions integrate into coherent developmental signaling, highlighting neuron-glia communication pathways as the main actors regulating CC prenatal ontogenesis and postnatal function. Finally, we discuss genetic knockout models and human case studies, which connect disrupted glial signaling to specific callosal pathologies, and outline translational tools and emerging methodologies, such as single-cell transcriptomics, iPSC-derived organoids, and enhancer activity-dependent genetic tools, that enable mechanistic investigations of specific glial functions. By assessing the most recent evidence on the cellular and molecular architecture of glial contributions to CC development, this review serves as a resource for understanding the role of glial cells in the ontogenesis of interhemispheric networks in mammals, as well as the formation and integration of the signaling between glial and neuronal cells in the development and agenesis of the corpus callosum.

Corpus callosum ontogenesis in human and rodents

The development of the CC follows a conserved sequence of events across mammalian species, although the timing and spatial organization differ between rodents and humans. In humans, the corpus callosum starts forming around gestational week 8 (GW8), and the first axons cross the midline between GW12 and GW20. After birth, the CC continues to mature, primarily through the gradual accumulation of myelin around axons, a process that lasts into adolescence (Paul, 2011; Birnbaum et al., 2020). In mice, callosal axons start crossing the midline during late pregnancy, around embryonic days 17–18 (E17–E18), and this process continues actively during the first 2–3 weeks after birth (Table 1). Despite these temporal differences, the main stages of callosal formation, axon emergence, guidance, and crossing through the midline, and then targeting the opposite hemisphere, remain conserved among placental (e.g., eutherian) mammals (De León Reyes et al., 2020).

TABLE 1
www.frontiersin.org

Table 1. Timing of callosal development in humans and mice.

Glial structures at the midline are critical in guiding axons by secreting signaling molecules and drive neuronal axons through their pathfinding. Astrocytes located in the glial wedge and indusium griseum glia, along with the transient subcallosal sling, create a signaling environment that shapes axon paths. The glial wedge, in particular, secretes Slit proteins that direct axons toward the appropriate crossing zone (Shu et al., 2003; Gobius et al., 2016). At the same time, glial cells from ventral midline regions release Netrin-1, which attracts axons that express the DCC receptor (Kang et al., 2018; Sagi-Dain et al., 2020; Morcom et al., 2021b). The balance between repulsion and attraction guides callosal axons across the midline, while molecules like L1CAM and extracellular matrix proteins promote axon bundling and growth (Gómez et al., 2008; Peotter et al., 2022). Other signaling molecules further fine-tune this process. Ephrin-A and EphA receptors, Semaphorin3C and its receptor Nrp1, and Reelin signaling help organize cortical structure and axon sorting (Hu et al., 2003; Cargnin et al., 2018; Mire et al., 2018; Zhi et al., 2025). Genetic disruption of these systems in animal models, such as knockouts of Netrin-1, DCC, or Slit/Robo, leads to misrouted axons, absent callosal connections, or the formation of Probst bundles, where axons fail to cross the midline and instead run along the ipsilateral hemisphere (Unni et al., 2012; Fothergill et al., 2014; Galichet et al., 2021). During this period, both neurons and glial cells migrate to reshape the midline tissue architecture. Astrocyte-mediated remodeling of the interhemispheric fissure (IHF) is essential to allow axons to bridge the hemispheres (Gobius et al., 2016; Liu et al., 2023). This coordinated sequence of growth, signaling, and structural change appears conserved across placental mammals, reflecting a shared molecular foundation for interhemispheric connectivity mediated by the fetal glia.

Astrocytes in CC development

Astrocytes are star-shaped glial cells traditionally known for maintaining homeostasis and supporting neurons. Far from passive support cells, astrocytes play active roles in CC development (Verkhratsky and Nedergaard, 2018; Meyer et al., 2018; Stogsdill et al., 2023). During embryogenesis, specialized astroglial cells at the midline act as navigational guideposts for callosal axons. They form key structures such as the glial wedge and midline “zipper” glia, which secrete guidance factors that drive axon crossing the midline (Shu et al., 2003; Gobius et al., 2016; Verkhratsky and Nedergaard, 2018; Liu et al., 2023). For example, midline astrocytes release Slit proteins that bind Robo receptors on cortical callosal axons, driving them to appropriate trajectories and conveying axonal bundles toward crossing the interhemispheric fissure (Gobius et al., 2016; Liu et al., 2023). This ensures that pioneer axons approach the midline correctly and do not erroneously enter other brain regions. Concurrently, astrocytes produce several trophic and attractants like growth factors (e.g., FGF8, IGF-1) to encourage axon extension toward the midline (Fothergill et al., 2014; Suarez et al., 2014; Figure 1). Disruption of astroglial guideposts has severe consequences: ablating or mispositioning the glial wedge in animal models leads to callosal agenesis, highlighting that the astrocytic guide function is indispensable (Bedeschi et al., 2006; Clegg et al., 2019; Rapti, 2023).

FIGURE 1
Diagram showing the development of the corpus callosum from 15 to 17 days post coitum (dpc). Panel A shows the presence of cells in the indusium griseum and glial wedge cells. Panel B at 16 dpc introduces FGF8 and IGF-1 markers. Panel C at 17 dpc illustrates the alignment of midline zipper glia and corpus callosum formation. Each panel highlights interhemispheric fissure, cortical colossal neurons, and relevant cell types with a legend on the right.

Figure 1. Multi-stage inter-hemispheric fissure remodeling mediated by the glial structures localized at the midline (indusium griseum, midline zipper glia, and glial wedge). (A) Midline glial structures change conformation throughout multiple developmental stages and (B) secrete growth factors, such as FGF8 and IGF1, which in turn stimulate axon extension toward the midline, (C) leading to normal development of the CC. Created in BioRender (https://app.biorender.com/citation/6904bbd3cf3d6e0fe32c2298).

Beyond guidance, astrocytes act as a physical scaffold for migrating neuronal axons. They extend long processes that interdigitate with the axonal growth cones, offering a substrate for inter-neurons connectivity (Meyer-Franke et al., 1999). Astrocytes can also modulate the extracellular environment by secreting extracellular matrix molecules that promote axon adhesion and bundling (Song and Dityatev, 2018; Lagos-Cabré et al., 2020). In these ways, astrocytes ensure that the nascent callosal axons maintain the correct trajectory as the corpus callosum forms. Recent studies have shown that the transformation of radial glia into the so-called midline “zipper” astroglia and their anatomical remodeling in the interhemispheric fissure are crucial steps for callosal axons to cross the midline (Gobius et al., 2016, 2017; Morcom et al., 2021b).

Astrocytes also shuttle nutrients (glucose and lactate, in particular) from blood to neurons and clean up excess neurotransmitters or ions, thereby optimizing the microenvironment for growing axons (Brown et al., 2004; Goursaud et al., 2009; Morrison et al., 2013). Such metabolic support is crucial, as axonal growth and subsequent myelination are energy-demanding processes. This support is mediated by glucose uptake through astrocytic GLUT1 transporters and conversion to lactate, which is then delivered to neurons via monocarboxylate transporters (MCT1 in astrocytes and MCT2 in neurons) to fuel oxidative phosphorylation. Additionally, astrocytes regulate extracellular glutamate and potassium through EAATs and Kir4.1 channels, maintaining ionic balance critical for axonal signaling and growth cone motility. Recent studies suggest that lactate shuttling not only sustains neuronal metabolism but also acts as a signaling molecule promoting axon elongation through NADH-mediated redox signaling and BDNF expression (Mason, 2017; Debernardi et al., 2003; Eid et al., 2019; Yang et al., 2014).

Notably, astrocytes express high levels of transporters for lactate and ions, positioning them as key regulators of energy supply to developing white matter and callosal projections (Fayol et al., 2004; Rinholm et al., 2011; Meyer et al., 2018; Matthiesen et al., 2022).

In later development, astrocytes influence synaptogenesis and circuit refinement in the forming corpus callosum (Levitt, 2003; Clarke and Barres, 2013). They secrete factors such as thrombospondins (TSP-1, -2) and Chordin-like 1 that promote synapse formation among callosal projection neurons (Christopherson et al., 2005; Kucukdereli et al., 2011). Astrocytes also release trophic factors (e.g., brain-derived neurotrophic factor, BDNF) that fortify emerging synaptic connections (Bathina and Das, 2015). Moreover, astrocytes help eliminate weaker synapses by engulfing synaptic debris and releasing signals that activate microglial pruning (discussed below). For instance, astrocyte-derived TGF-β induces neurons to express the complement protein C1q, which tags weak synapses for removal by microglia (Stevens et al., 2007). Simultaneously, astrocyte-secreted IL-33 acts on microglia to enhance their engulfment of synapses during developmental remodeling (Vainchtein et al., 2018). This bidirectional role of astrocytes toward neurons and non-neuronal cells ensures that robust interhemispheric synapses persist while redundant ones are pruned, which is a crucial process for efficient inter-hemispheric connectivity.

In summary, astrocytes are indispensable architects of corpus callosum development. State-of-the-art research emphasizes their multifaceted contributions from anatomical remodeling, to guiding axons at the earliest stages and maintaining homeostasis and regulating synaptogenesis in the developing and maturing CC. In disease conditions, astrocytes can also contribute to pathology—reactive astroglia may form scars in the CC after injury or demyelination, and astrocytic dysfunction is implicated in disorders that feature callosal myelination defects (Bonetto et al., 2021; Gzielo and Nikiforuk, 2021; Morcom et al., 2021b). Conversely, enhancing astrocytic support functions is being explored therapeutically (e.g., transplanting astrocyte progenitors to promote regeneration) (Sullivan et al., 2020). Overall, astrocytes in the CC are not mere supporters of neuronal development and functions, but they are active participants, ensuring that callosal axons find their paths, interacting with neurons through metabolic support and signaling molecules.

Oligodendrocytes in CC development

Oligodendrocytes are the myelin-forming glia of the central nervous system, crucial for the maturation and plasticity of axonal networks and the CC. Oligodendrocyte precursor cells (OPCs) arise from the ventricular and subventricular zones during mid-gestation and migrate later into the developing corpus callosum (Mitew et al., 2014). In rodents, OPCs start colonizing the CC around late embryonic stages; in humans, OPCs are present in the fetal CC and expand postnatally as myelination ramps up. The migration, proliferation, and differentiation of OPCs in the callosal region are tightly regulated by multiple signaling pathways in the developing brain (Galichet et al., 2021; Buchanan et al., 2023; Akinlaja and Nishiyama, 2024). The pathways and molecular actors affecting OPC development are listed in Table 2.

TABLE 2
www.frontiersin.org

Table 2. Crucial signaling pathways for CC development in the fetal brain.

Once OPCs have migrated and proliferated in the corpus callosum, they differentiate into mature oligodendrocytes. This differentiation is marked by the expression of myelin proteins (e.g., MBP, PLP, MOG) and by the wrapping of oligodendrocyte processes around axons to form myelin internodes. Myelination in the CC starts perinatally in many mammals and accelerates during infancy. In humans, myelination proceeds roughly from posterior to anterior (the splenium myelinates before the genu), continuing into the second and third decades of life (Mount and Monje, 2017; Kuhn et al., 2019; Hughes, 2021).

Oligodendrocytes provide metabolic and trophic support to neurons by delivering lactate to axons and buffering ions, thus helping maintain axonal homeostasis (Kuhn et al., 2019; Kennedy et al., 2025). If oligodendrocyte development is perturbed, axonal caliber and survival can be affected. For example, in mouse models where oligodendrocyte generation is blocked or significantly delayed, callosal axons show stunted growth and some degenerate over time, demonstrating that oligodendrocytes are essential partners in axon integrity (Simons and Nave, 2016; Simons et al., 2024).

Recent research highlights the role of oligodendrocytes in activity-dependent plasticity. Rather than being static, myelination by oligodendrocytes can adapt in response to neuronal activity, a phenomenon termed adaptive myelination (Gibson et al., 2014; Knowles et al., 2022). Studies show that learning new skills or exposing animals to enriched environments induces additional myelin formation in relevant callosal pathways (Tripathi et al., 2017; Nicholson et al., 2022). This suggests that oligodendrocytes contribute to refining neural circuits even beyond early development. Myelin plasticity may help fine-tune neural signal transmission between hemispheres, thereby affecting functions like language, motor coordination, and cognition that rely on the CC (Xin and Chan, 2020). For instance, increased motor learning in mice leads to heightened oligodendrogenesis and myelination in motor callosal fibers (Simons and Nave, 2016; Simons et al., 2024). A recent review noted that myelin changes can strengthen circuit function underlying learning and memory (Bonetto et al., 2021). Mechanistically, neuronal firing can promote the proliferation and differentiation of nearby OPCs (through glutamatergic signaling and activity-induced astrocytic ATP/K+ release), leading to new myelin sheaths on active axons. This plastic myelination is thought to support circuit plasticity by optimizing conduction speed, and its dysregulation could contribute to developmental disorders (Chen et al., 2018; Yang et al., 2020).

Proper oligodendrocyte development in the CC is critical for its formation and adequate inter-hemispheric functions. Hypomyelination of the corpus callosum in infancy can manifest as developmental delay, poor motor coordination, or cognitive deficits. For example, preterm infants with periventricular leukomalacia (white matter injury) often show reduced callosal volume and later motor and cognitive impairments; this is due in part to loss or injury of OPCs during a vulnerable perinatal period (Back, 2017). Genetic disorders affecting oligodendrocytes, such as leukodystrophies, typically involve the corpus callosum prominently (diagnosed as a thinned CC or abnormal T2 signal on MRI) and lead to severe neurological symptoms (van der Knaap and Bugiani, 2017). Diffusion tensor imaging studies in schizophrenia and autism have found subtle callosal white matter microstructural abnormalities, suggesting that oligodendrocyte dysregulation may contribute to the connectivity changes in these conditions (Haigh et al., 2019; Faraji et al., 2023). In addition, research in multiple sclerosis has raised interest in enhancing oligodendrocyte regeneration; several experimental therapies that promote OPC differentiation (e.g., the antihistamine clemastine and other remyelinating agents) are being explored and associated with developmental myelination disorders (Yamazaki and Ohno, 2025).

In summary, oligodendrocytes are central to the formation, maturation, and functions of the CC. Through tightly regulated developmental programs, oligodendrocyte lineage cells populate the CC, wrap axons with myelin, and support axonal function. Advances in developmental neuroscience and glial biology continue to shed light on the molecular mechanisms regulating oligodendrogenesis and myelination in the CC, offering insights into how disruptions in these processes lead to neurodevelopmental disorders and how activity or experience can influence callosal connectivity via adaptive myelination.

Microglia in CC development

Microglia are the resident immune cells of the central nervous system and emerge early during embryonic development. Unlike astrocytes and oligodendrocytes, that originate from the neuroectoderm, the microglia derives from the yolk sac cells progenitors colonizing the embryonic brain before the formation of the blood-brain barrier (Ginhoux et al., 2010). In mice, microglia begin colonizing the brain around embryonic day 9.5, and by mid-gestation, they are widely distributed in developing white matter tracts, including the developing corpus callosum (Alliot et al., 1999; Thion et al., 2018).

Microglia play essential roles in regulating axon guidance, structural organization, and synaptic remodeling. In early stages, microglia help organize callosal axons into compact bundles by promoting fasciculation (Fujita and Yamashita, 2021). During early brain development, microglia facilitate axon fasciculation through a limited set of well-defined molecular pathways. The adaptor protein DAP12 (TYROBP) mediates microglial signaling required for callosal axon bundling, as its loss causes marked defasciculation (Pont-Lezica et al., 2014). In parallel, neuronal Netrin-G1 and microglial NGL1 (LRRC4C) interactions recruit microglia to developing axon tracts, while microglia-derived IGF-1 provides trophic support that stabilizes and promotes axonal growth (Fujita et al., 2020). Additionally, microglia secrete thrombospondins and remodel the extracellular matrix via MMP-2/9, creating a permissive environment for axon bundling. Together, these pathways establish the main molecular framework through which microglia orchestrate axon fasciculation during central nervous system (CNS) development (Squarzoni et al., 2014; Fujita et al., 2020). Experiments using microglia-deficient models, such as Pu.1 knockout mice, have shown that the absence of microglia leads to defasciculated and misrouted callosal axons (Pont-Lezica et al., 2014). This suggests that microglia not only interact physically with growing axons but also secrete trophic factors that help maintain axonal alignment, such as IGF-1 and BDNF to promote axon growth, and adhesion molecules like ICAM-1, VCAM-1, and integrins to stabilize and fasciculate developing axon bundles (Carlstrom et al., 2011; Guo et al., 2018; Constantin et al., 1999).

Microglia play a critical role in refining neural circuits through synaptic remodeling, selectively eliminating weak or inactive synapses to optimize connectivity. This process involves molecular mechanisms such as the complement cascade (e.g., C1q, C3) and receptors including CR3 and TREM2, which help microglia identify and engulf unnecessary synapses (Neniskyte and Gross, 2017; Ball et al., 2022; Guedes et al., 2022). In the context of corpus callosum development, synaptic pruning by microglia ensures that functionally relevant interhemispheric connections are maintained, contributing to efficient neural communication between hemispheres. Disruptions in microglial synaptic remodeling can lead to abnormal callosal connectivity, such as overconnectivity or hypoconnectivity, which have been linked to neurodevelopmental disorders including autism spectrum disorder and schizophrenia (Luo and Wang, 2024). In addition, microglia have been implicated in axonal pruning in other brain regions or developmental stages, evidence for direct microglial removal of callosal axons during corpus callosum formation is limited and remains an area of ongoing investigation.

In the postnatal period, microglia refine callosal circuitry through activity-dependent synaptic remodeling. Astrocyte-derived molecules such as IL-33 and TGF-β modulate microglial behavior by enhancing their capacity to prune inactive synapses, thus supporting efficient neural signaling between hemispheres (Vainchtein et al., 2018; Zhang et al., 2025).

Microglia also influence oligodendrocyte progenitor development. In early postnatal life, they organize surplus OPCs, which help regulate the density and spacing of myelinating cells; consistent with this, Nemes-Baran et al. (2020) demonstrated that fractalkine receptor-deficient mice exhibit reduced microglial engulfment leading to an increased number of oligodendrocytes but paradoxically reduced myelin thickness, suggesting that microglial regulation is critical not just for OPC number but also for their maturation and myelination efficiency (Nemes-Baran et al., 2020).

Microglia also release factors such as IGF-1 that promote oligodendrocyte maturation and myelin production. Under pathological conditions, however, activated microglia can secrete inflammatory cytokines that impair OPC differentiation and reduce myelination, contributing to white matter injury (Smith et al., 2012; Rusin et al., 2024).

Microglia surveillance throughout the CNS, by clearing apoptotic cells, responding to environmental insults, and maintaining tissue homeostasis, play a major role in CC development and maturation. In the corpus callosum, microglia contribute specifically by shaping axonal architecture, refining synapses, and regulating myelination. Disruption of microglial function, whether through genetic or environmental factors, impaired callosal developmental processes and lead to callosal malformations or other connectivity defects (Santos and Fields, 2021; McNamara et al., 2023).

Neuron-glia crosstalk in corpus callosum development

Astrocytes, oligodendrocytes, and microglia collaborate to orchestrate corpus callosum formation through tightly regulated molecular and cellular mechanisms. Midline astrocytes act as guideposts, forming the glial wedge and zipper glial structures that secrete guidance cues such as Slit, Netrin-1, and growth factors (FGF8, IGF-1) to direct pioneer callosal axons, while providing scaffolding and metabolic support via GLUT1/MCT1-mediated lactate shuttling and EAAT/Kir4.1-mediated ionic homeostasis. Oligodendrocyte precursor cells (OPCs) migrate into the developing CC under the influence of growth factors, chemokines, and neuronal activity, differentiating into myelinating oligodendrocytes that express MBP, PLP, and MOG; these cells not only enable efficient action potential conduction but also support axonal metabolism and activity-dependent plasticity through adaptive myelination. Microglia further refine CC circuitry by promoting axon fasciculation via DAP12, NGL1/Netrin-G1, and IGF-1, pruning excess axons and synapses through complement signaling (C1q/C3–CR3/TREM2) and modulating oligodendrocyte maturation, with astrocyte-derived IL-33 and TGF-β shaping microglial activity. Together, these glial populations integrate structural, metabolic, and signaling functions to ensure precise axon guidance, myelination, and synaptic connectivity, and disruptions in any of these pathways can lead to callosal malformations or connectivity deficits implicated in neurodevelopmental disorders (Gobius et al., 2016; Fujita et al., 2020; Pont-Lezica et al., 2014).

As discussed above, the development of the CC relies on precisely coordinated communication between neurons and glial cells, which together regulate axon guidance, myelination, and synaptic remodeling. This intercellular dialogue occurs through the dynamic exchange of molecular signals (ligands, receptors, and intercellular junctions) that direct the spatial and temporal patterning of CC development (Table 3). Understanding the functional consequences of these pathways requires examining not only the molecules involved but also their sources, targets, and downstream effects.

TABLE 3
www.frontiersin.org

Table 3. Key integrated pathways.

A well-characterized pathway is the Slit-Robo signaling axis, essential for midline navigation. Midline astrocytes in the glial wedge and indusium griseum glia secrete Slit2, which binds to Robo1/2 receptors expressed on cortical callosal axons. This interaction exerts a driving force that channels axons toward and across the interhemispheric fissure, preventing them from straying into inappropriate regions (Unni et al., 2012; Gonda et al., 2020). In parallel, radial glia and ventral midline cells produce Netrin-1, which binds to DCC receptors on the same neurons, acting as an attractive signal (Unni et al., 2012; Fothergill et al., 2014). Together, these cues form a molecular corridor guiding axons to and through the midline. Robo expression increases after crossing, sensitizing axons to Slit-mediated repulsion and preventing re-crossing (Fothergill et al., 2014).

Additional astrocyte-secreted factors, including fibroblast growth factor 8 (FGF8) and insulin-like growth factor 1 (IGF-1), further contribute to axonal extension and navigation by modulating the extracellular matrix and enhancing axonal responsiveness to guidance cues (Dyer et al., 2016; Zhang W. et al., 2023). Neuron-to-glia signaling also plays a crucial role in the development of oligodendrocytes and myelination. Neurons express membrane-bound Neuregulin-1 (NRG1), which interacts with ErbB2/3 receptors on OPCs. This interaction promotes OPC survival, migration, and terminal differentiation (Kim et al., 2003; Hu et al., 2024). The magnitude of NRG1 signaling is proportional to neuronal activity, allowing active axons to be preferentially myelinated. Astrocytes influence this pathway indirectly by releasing extracellular modulators that affect ErbB receptor expression and NRG1 availability.

Microglia support corpus callosum formation by interacting with growing axons, shaping axonal architecture, and maintaining a permissive environment for midline crossing during fetal development. After birth, microglia contribute to the refinement of callosal projections by participating in synaptic remodeling and final inter-hemispheric connectivity. Neurons release CX3CL1 (fractalkine), which binds CX3CR1 receptors on microglia, regulating their surveillance and phagocytic activity, including synaptic pruning (Paolicelli et al., 2014; Pawelec et al., 2020; Wang et al., 2021). Astrocytes also signal via interleukin-33 (IL-33), promoting microglial phagocytosis of underactive synapses in an activity-dependent manner, thus reinforcing active synapses and eliminating superfluous ones (Vainchtein et al., 2018; Han R. T. et al., 2023).

Crosstalk involving activity-dependent feedback loops further regulates glial function. Neuronal firing elevates extracellular ATP and potassium, which are detected by astrocytes and OPCs. These glial cells respond by releasing lactate, growth factors, or pro-differentiation signals that support axon migration and promote myelination (Chen et al., 2024).

Recently, DRAXIN, a secreted chemorepulsive guidance protein, has been shown to play a critical role in CC formation by regulating the astroglial-dependent remodeling of the interhemispheric fissure that precedes axon crossing (Morcom et al., 2021a). In mice, DRAXIN is expressed by dorsal radial glia and emerging midline zipper glia (MZG progenitors) and interacts genetically and biochemically with DCC to modulate callosal axon trajectories (Islam et al., 2009). Loss of Draxin disrupts early steps of IHF remodeling, namely MZG specification, radial glial process extension, and somal translocation, which result in an enlarged, leptomeningeal-filled fissure and failure of pioneer axons to cross (Morcom et al., 2021a). Thus, DRAXIN functions upstream of Slit–Robo and Netrin-DCC signals by establishing the astrocytic scaffold necessary for these cues to guide axons across the midline.

Bidirectional signaling among neurons, astrocytes, oligodendrocytes, and microglia thus forms a coordinated network guiding the successive ontogenesis of the CC, from initial pathfinding and axonal elongation to glial anatomical remodeling, myelin wrapping, and synaptic culling. Disruptions in these communication pathways have been implicated in multiple neurodevelopmental disorders, including autism spectrum disorder, schizophrenia, and congenital callosal agenesis (Szepesi et al., 2018). A mechanistic understanding of these interactions not only clarifies the cellular basis of CC development but also points to potential therapeutic targets for correcting or preventing interhemispheric connectivity deficits.

Callosal developmental malformations in human disorders and animal models

Experimental models, particularly genetically modified mice, have been pivotal in elucidating the molecular and cellular mechanisms underlying CC development. By targeting specific genes involved in axon guidance, midline patterning, glial development, and synaptic remodeling, these models replicate key features of human callosal malformations and provide insights into conserved developmental pathways. Many gene deletions in mice result in partial or complete callosal agenesis, and corresponding human mutations frequently lead to similar neurodevelopmental anomalies (Gavrish et al., 2024).

One of the best-characterized models involves the deletion of Dcc (Deleted in Colorectal Carcinoma), a receptor for the guidance molecule Netrin-1. Mice lacking Dcc exhibit complete agenesis of the CC, with axons failing to cross the midline and instead forming Probst bundles (Fothergill et al., 2014; Morcom et al., 2021b). In humans, heterozygous mutations in DCC cause congenital syndrome and partial callosal agenesis, emphasizing the translational value of knock-out mouse models (Marsh et al., 2017, 2018).

Similarly, knockout of Ntn1, encoding Netrin-1 protein, also leads to severe callosal pathfinding defects in mice, reinforcing the essential nature of the Netrin-DCC axis in midline crossing (Sagi-Dain et al., 2020). Double knockout of Robo1/2, the receptors for Slit proteins, results in aberrant axonal trajectories and misrouting of callosal fibers into subcortical regions (Andrews et al., 2006; Fouquet et al., 2007; Tamada et al., 2008). Mice with disrupted Slit2 also show axons accumulating near the midline without successful crossing, recapitulating features observed in humans with agenesis due to midline glial dysfunction and similar mutations (Long et al., 2004; Figure 2).

FIGURE 2
Cross-sectional diagram of the brain with three panels highlighting corpus callosum development. Panel A shows “Wild Type” with typical nerve pathways. Panel B labeled “Robo1 -/-” and Panel C labeled “Slit -/-” depict altered pathways with stop signals, indicating genetic modifications affecting neuronal development.

Figure 2. Schematic representations of CC developmental malformations in two genetic mouse models. (A) The successful development of the CC in wild type brain is mediated by signaling interactions between glial structures and neurons (i.e., Slit1-Robo1 pathway). (B) Knock-out mice carrying mutations in the Robo1 receptor gene display callosal malformation due to impaired neuronal responsivity to the Slit1 guidance factor. (C) Knock-out mice lacking the glia-derived Slit1 guidance factor exhibit similar callosal malformations as in the Robo1 mutant, although resulting from disrupted ligand secretion by the glial structures. Created in BioRender (https://app.biorender.com/citation/6904bbd3cf3d6e0fe32c2298).

The X-linked L1CAM gene, encoding an axonal cell adhesion molecule, is another major player in callosal development. L1cam-deficient mice show the absence of the CC and abnormal axon fasciculation. In humans, mutations in L1CAM cause CRASH syndrome (Corpus callosum agenesis, Retardation, Adducted thumbs, Spastic paraplegia, and Hydrocephalus), a well-documented X-linked disorder linked to glial tissue and its surrounding extracellular environment (Fransen et al., 1995; Yamasaki et al., 1997).

Knockouts affecting midline glia-specific morphogens also result in callosal abnormalities. For example, mice lacking Fgfr1 or Gli3, involved in the glial wedge formation and dorsal telencephalon patterning, develop agenesis or hypoplasia of the CC (Magnani et al., 2014; Gobius et al., 2016). These models underscore the importance of astrocyte-mediated midline remodeling in creating a permissive path for axons.

Mutations in genes affecting oligodendrocyte function and myelination also lead to structural anomalies in the CC. Mice deficient in Plp1 or Mbp show delayed or incomplete callosal myelination (Arinrad et al., 2023). In humans, leukodystrophies such as Pelizaeus-Merzbacher disease (linked to PLP1 mutations) and metachromatic leukodystrophy (linked to ARSA) feature callosal thinning and signal changes visible on MRI (Groeschel et al., 2011; Laukka et al., 2014). Hypomyelination and disrupted oligodendrocyte development during key developmental windows can thus significantly affect callosal integrity (Anderson et al., 2018).

Disruptions in microglial signaling also result in callosal defects. Cx3cr1-knockout mice exhibit impaired synaptic pruning and excess dendritic spines, alongside transient delays in callosal refinement (Paolicelli et al., 2011). Mice with maternal immune activation (MIA) or deletions in complement components (e.g., C1q, C3, Trem2) show altered synaptic density and impaired microglial pruning, leading to aberrant interhemispheric connectivity (Filipello et al., 2018; Yan et al., 2024). These immune-related findings have been linked to risk factors for neurodevelopmental disorders such as autism and schizophrenia.

Furthermore, knockout or knockdown of IL33, an astrocyte-derived cytokine essential for microglia-mediated synapse elimination, impairs developmental pruning and results in circuit hyperconnectivity. These phenotypes parallel features observed in human ASD brains, which often show increased spine density and abnormal CC morphology (Vainchtein et al., 2018).

In addition to targeted knockouts, naturally occurring inbred strains have demonstrated genetic backgrounds predisposing to callosal agenesis. For example, the BTBR T+ Itpr3/ mouse strain exhibits a complete absence of the CC, and attendant forebrain connectivity deficits, making it a widely used model for studying idiopathic agenesis and its behavioral and metabolic correlates (Vega-Pons et al., 2017; Martin et al., 2021; Morcom et al., 2021a; Viscomi et al., 2025). Similarly, BALB/c mice display variable thinning or partial absence of the rostral CC, correlating with altered interhemispheric connectivity although less penetrant (Wahlsten, 1989; Wahlsten and Schalomon, 1994; Bohlen et al., 2012). These inbred models underscore that polygenic and strain-specific factors, beyond single-gene disruptions, can profoundly impact midline glial patterning and axon guidance in the developing CC.

Collectively, mouse models have provided crucial evidence that both neuronal and non-neuronal genes regulate CC development. The parallel occurrence of similar phenotypes in humans with orthologous gene mutations (e.g., DCC, L1CAM, PLP1) validates these models, thus supporting their use in dissecting the cell-type-specific roles of glia and neurons. Ongoing refinement of conditional and cell-type-specific knockout strategies will further delineate the contribution of each glial population to specific stages of CC ontogenesis.

Emerging tools and translational perspectives

A comprehensive understanding of CC development increasingly relies on tools capable of dissecting the specific roles of glial and neuronal cells. While classical studies often focused on axonal pathfinding, newer experimental approaches have shifted attention toward how astrocytes, oligodendrocytes, and microglia orchestrate axon guidance, myelination, and synaptic remodeling.

Conditional knockout mouse models have become indispensable for clarifying cell-type-specific functions. Cre-loxP systems using promoters such as Aldh1l1-Cre for astrocytes, Olig2-Cre for oligodendrocyte lineage cells, and Cx3cr1-Cre for microglia allow for targeted gene manipulation (Livet et al., 2007; Hu et al., 2020). For example, astrocyte-specific deletion of Fgfr1 or Gli3 disrupts glial wedge formation and leads to callosal agenesis (Gobius et al., 2016; Morcom et al., 2021b). Likewise, Cx3cr1-Cre mice have been used to show that altered microglial activity during prenatal and early postnatal periods results in callosal miswiring and overconnectivity (Hoshiko et al., 2012; Sahasrabuddhe and Ghosh, 2022).

In vivo imaging techniques such as time-lapse two-photon microscopy and magnetic resonance imaging (MRI) are being adapted to monitor glial behavior and CC integrity in real-time. Labeling of glial subpopulations with fluorescent reporters (e.g., Sox10-GFP for oligodendrocytes, Cx3cr1-GFP for microglia) enables the visualization of cell migration, axon interaction, and morphological remodeling in developing brains. Diffusion tensor imaging (DTI) also allows the correlation of microstructural white matter changes with genetic or environmental alterations affecting glial cell populations (Zhang B. et al., 2014).

Single-cell and spatial transcriptomics provide further resolution into glial diversity and function within the developing CC. These techniques allow the identification of transcriptional states linked to distinct roles in axon guidance, myelination, or pruning. For instance, scRNA-seq datasets from developing mouse brains have revealed temporally shifting microglial gene signatures associated with phagocytosis, complement activation, and cytokine signaling. Astrocyte and OPC populations likewise exhibit unique gene expression profiles correlated with midline signaling, extracellular matrix remodeling, and metabolic support to the neuronal pathfinding and functions (Zhang B. et al., 2014; Li et al., 2019; Allen et al., 2023; Han Z. et al., 2023).

In vitro models also offer controlled environments to dissect molecular pathways. Co-culture systems of neurons with glia, especially astrocytes or OPCs, permit functional assays of axon outgrowth, myelination, and synaptogenesis (Shimizu et al., 2011; Roqué and Costa, 2017; Goshi et al., 2020). Application of recombinant proteins (e.g., Slit2, Netrin-1, IL-33, TGF-β) or pharmacological inhibitors (e.g., PDGFR blockers, Notch antagonists) further refines these models. Additionally, patient-derived iPSC lines can be differentiated into glial subtypes to evaluate the functional impact of disease-associated variants on CC-relevant processes (Frati et al., 2018).

Recent advances in brain organoids and assembloids technologies allow partial modeling of interhemispheric connections and glial development in a human cellular context (Wu and Nowakowski, 2025). Although callosum-like axonal tracts remain incomplete in these systems, improvements in regional patterning and glial differentiation protocols are enhancing their translational value. Transplantation of oligodendrocyte progenitors or astrocyte precursors into mouse brains or cerebral organoids is also being explored to test the capacity of glial cells to support or restore callosal function (Luciani et al., 2024).

Overall, the integration of conditional genetics, live imaging, transcriptomics, and iPSC-based modeling is evolving the study of glial cells into cell-specific functions and signaling affecting CC development. These approaches not only deepen our understanding of cellular interactions in the developing forebrain but also help establish mechanistic links to human disorders marked by callosal dysgenesis. Moving forward, these models and tools are essential for uncovering cell-type-specific vulnerabilities and potential therapeutic strategies targeting glial dysfunction.

Concluding remarks and future directions

The development of the corpus callosum involves a complex orchestration of cellular interactions, where glial cells function not merely as passive support elements but as active, essential participants in every phase of interhemispheric connectivity, contributing to the growth and survival of callosal neuronal projections (Figure 3). Overall, the evidence presented throughout this review affirms that callosal development is not a neuron-centric process but rather a cooperative venture involving diverse glial populations.

FIGURE 3
Illustration showing a neuron connecting the right and left hemispheres. It includes labeled components: dendrites, synapse, axon, and myelin sheets. Oligodendrocytes, astrocytes, and microglia are labeled below, with corresponding structures shown in the diagram.

Figure 3. Cooperation between microglia, astrocytes, and oligodendrocytes in the postnatal brain. This schematic illustrates the dynamic interactions where microglia mediate synaptic pruning and immune surveillance, astrocytes support neuronal health and modulate synapse maturation, and oligodendrocytes facilitate myelination. Together, these glial cells coordinate to maintain neural circuit integrity and promote proper development and function of the corpus callosum. Created in BioRender (https://app.biorender.com/citation/6904bbd3cf3d6e0fe32c2298).

Comparative studies in rodents and humans reveal both conserved pathways and species-specific adaptations in CC development. Importantly, environmental factors such as inflammation, hypoxia, or endocrine disruption interact with glial programs to influence outcomes, underlining the plasticity and vulnerability of this system.

The convergence of animal models and human data strongly supports the hypothesis that defects in glial function contribute to various callosal pathologies, from congenital agenesis to white matter disorders associated with neurodevelopmental syndromes. These pathologies often involve disruptions in fetal brain remodeling processes, and/or in neuron-glia signaling pathways such as Netrin-DCC, Slit-Robo, Neuregulin-ErbB, and CX3CL1-CX3CR1, suggesting a central role for glia in modulating axonal guidance, synaptic refinement, and structural maturation.

Despite significant advances, critical questions remain unanswered. The specific molecular programs that define regional glial heterogeneity within the CC, the temporal coordination of neuron-glia communication across developmental windows, and the degree to which these mechanisms can be targeted for therapeutic intervention remain active areas of research. The functional consequences of glial dysfunction in neuropsychiatric disorders also warrant deeper investigation. Future directions include the integration of multimodal single-cell datasets (transcriptomic, proteomic, etc.) with cell-type-specific gene editing tools to map causal mechanisms. Longitudinal studies in human cohorts, supported by iPSC and organoid/assembloid models, are expected to refine translational applications.

Author contributions

JC: Investigation, Visualization, Writing – original draft, Writing – review & editing. MZ: Investigation, Visualization, Writing – original draft, Writing – review & editing, Resources. AB: Investigation, Visualization, Writing – original draft, Writing – review & editing. SS: Conceptualization, Funding acquisition, Resources, Supervision, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This research was funded by the Polish National Science Centre (Narodowe Centrum Nauki, NCN), project OPUS No. 2020/39/B/NZ4/02105 to SS.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare Generative AI was used in the creation of this manuscript. Generative AI was used to correct English grammar and improve readability.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Akinlaja, Y. O., and Nishiyama, A. (2024). Glial modulation of synapse development and plasticity: Oligodendrocyte precursor cells as a new player in the synaptic quintet. Front. Cell Dev. Biol. 12:1418100. doi: 10.3389/fcell.2024.1418100

PubMed Abstract | Crossref Full Text | Google Scholar

Allen, W. E., Blosser, T. R., Sullivan, Z. A., Dulac, C., and Zhuang, X. (2023). Molecular and spatial signatures of mouse brain aging at single-cell resolution. Cell 186, 194–208.e18. doi: 10.1016/j.cell.2022.12.010

PubMed Abstract | Crossref Full Text | Google Scholar

Alliot, F., Godin, I., and Pessac, B. (1999). Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 117, 145–152. doi: 10.1016/s0165-3806(99)00113-3

PubMed Abstract | Crossref Full Text | Google Scholar

Anderson, C., Gerding, W. M., Fraenz, C., Schlüter, C., Friedrich, P., Raane, M., et al. (2018). PLP1 and CNTN1 gene variation modulates the microstructure of human white matter in the corpus callosum. Brain Struct. Funct. 223, 3875–3887. doi: 10.1007/s00429-018-1729-7

PubMed Abstract | Crossref Full Text | Google Scholar

Andrews, W., Liapi, A., Plachez, C., Camurri, L., Zhang, J., Mori, S., et al. (2006). Robo1 regulates the development of major axon tracts and interneuron migration in the forebrain. Development 133, 2243–2252. doi: 10.1242/dev.02379

PubMed Abstract | Crossref Full Text | Google Scholar

Arinrad, S., Depp, C., Siems, S. B., Sasmita, A. O., Eichel, M. A., Ronnenberg, A., et al. (2023). Isolated catatonia-like executive dysfunction in mice with forebrain-specific loss of myelin integrity. eLife 12:e70792. doi: 10.7554/elife.70792

PubMed Abstract | Crossref Full Text | Google Scholar

Armstrong, R. C., Le, T. Q., Frost, E. E., Borke, R. C., and Vana, A. C. (2002). Absence of fibroblast growth factor 2 promotes oligodendroglial repopulation of demyelinated white matter. J. Neurosci. 22, 8574–8585. doi: 10.1523/jneurosci.22-19-08574

Crossref Full Text | Google Scholar

Back, S. A. (2017). White matter injury in the preterm infant: Pathology and mechanisms. Acta Neuropathol. 134, 331–349. doi: 10.1007/s00401-017-1718-6

PubMed Abstract | Crossref Full Text | Google Scholar

Ball, J. B., Green-Fulgham, S. M., and Watkins, L. R. (2022). Mechanisms of microglia-mediated synapse turnover and synaptogenesis. Progr. Neurobiol. 218:102336. doi: 10.1016/j.pneurobio.2022.102336

PubMed Abstract | Crossref Full Text | Google Scholar

Bathina, S., and Das, U. N. (2015). Brain-derived neurotrophic factor and its clinical implications. Aoms 6, 1164–1178. doi: 10.5114/aoms.2015.56342

PubMed Abstract | Crossref Full Text | Google Scholar

Bedeschi, M. F., Bonaglia, M. C., Grasso, R., Pellegri, A., Garghentino, R. R., Battaglia, M. A., et al. (2006). Agenesis of the corpus callosum: Clinical and genetic study in 63 young patients. Pediatric Neurol. 34, 186–193. doi: 10.1016/j.pediatrneurol.2005.08.008

PubMed Abstract | Crossref Full Text | Google Scholar

Birnbaum, R., Barzilay, R., Brusilov, M., Wolman, I., and Malinger, G. (2020). The early pattern of human corpus callosum development: A transvaginal 3D neurosonographic study. Prenatal Diagnosis 40, 1239–1245. doi: 10.1002/pd.5735

PubMed Abstract | Crossref Full Text | Google Scholar

Bohlen, M. O., Bailoo, J. D., Jordan, R. L., and Wahlsten, D. (2012). Hippocampal commissure defects in crosses of four inbred mouse strains with absent corpus callosum. Genes Brain Behav. 11, 757–766. doi: 10.1111/j.1601-183x.2012.00802.x

PubMed Abstract | Crossref Full Text | Google Scholar

Bonetto, G., Belin, D., and Káradóttir, R. T. (2021). Myelin: A gatekeeper of activity-dependent circuit plasticity? Science 374:eaba6905. doi: 10.1126/science.aba6905

PubMed Abstract | Crossref Full Text | Google Scholar

Brown, A. M., Baltan Tekkök, S., and Ransom, B. R. (2004). Energy transfer from astrocytes to axons: The role of CNS glycogen. Neurochem. Int. 45, 529–536. doi: 10.1016/j.neuint.2003.11.005

PubMed Abstract | Crossref Full Text | Google Scholar

Buchanan, J., Da Costa, N. M., and Cheadle, L. (2023). Emerging roles of oligodendrocyte precursor cells in neural circuit development and remodeling. Trends Neurosci. 46, 628–639. doi: 10.1016/j.tins.2023.05.007

PubMed Abstract | Crossref Full Text | Google Scholar

Calver, A. R., Hall, A. C., Yu, W.-P., Walsh, F. S., Heath, J. K., Betsholtz, C., et al. (1998). Oligodendrocyte population dynamics and the role of PDGF in vivo. Neuron 20, 869–882. doi: 10.1016/S0896-6273(00)80469-9

PubMed Abstract | Crossref Full Text | Google Scholar

Cargnin, F., Kwon, J.-S., Katzman, S., Chen, B., Lee, J. W., and Lee, S.-K. (2018). FOXG1 orchestrates neocortical organization and cortico-cortical connections. Neuron 100, 1083–1096.e5. doi: 10.1016/j.neuron.2018.10.016

PubMed Abstract | Crossref Full Text | Google Scholar

Carlstrom, L. P., Hines, J. H., Henle, S. J., and Henley, J. R. (2011). Bidirectional remodeling of β1-integrin adhesions during chemotropic regulation of nerve growth. BMC Biol. 9:82. doi: 10.1186/1741-7007-9-82

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, T.-J., Kula, B., Nagy, B., Barzan, R., Gall, A., Ehrlich, I., et al. (2018). In vivo regulation of oligodendrocyte precursor cell proliferation and differentiation by the AMPA-receptor subunit GluA2. Cell Rep. 25, 852–861.e7. doi: 10.1016/j.celrep.2018.09.066

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, Y., Luan, P., Liu, J., Wei, Y., Wang, C., Wu, R., et al. (2024). Spatiotemporally selective astrocytic ATP dynamics encode injury information sensed by microglia following brain injury in mice. Nat. Neurosci. 27, 1522–1533. doi: 10.1038/s41593-024-01680-w

PubMed Abstract | Crossref Full Text | Google Scholar

Choudhry, Z., Rikani, A. A., Choudhry, A. M., Tariq, S., Zakaria, F., Asghar, M. W., et al. (2014). Sonic hedgehog signalling pathway: A complex network. ANS 21, 28–31. doi: 10.5214/ans.0972.7531.210109

PubMed Abstract | Crossref Full Text | Google Scholar

Christopherson, K. S., Ullian, E. M., Stokes, C. C. A., Mullowney, C. E., Hell, J. W., Agah, A., et al. (2005). Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell 120, 421–433. doi: 10.1016/j.cell.2004.12.020

PubMed Abstract | Crossref Full Text | Google Scholar

Clarke, L. E., and Barres, B. A. (2013). Emerging roles of astrocytes in neural circuit development. Nat. Rev. Neurosci. 14, 311–321. doi: 10.1038/nrn3484

PubMed Abstract | Crossref Full Text | Google Scholar

Clegg, J. M., Parkin, H. M., Mason, J. O., and Pratt, T. (2019). Heparan sulfate sulfation by Hs2st restricts astroglial precursor somal translocation in developing mouse forebrain by a non-cell-autonomous mechanism. J. Neurosci. 39, 1386–1404. doi: 10.1523/JNEUROSCI.1747-17.2018

PubMed Abstract | Crossref Full Text | Google Scholar

Constantin, G., Piccio, L., Bussini, S., Pizzuti, A., Scarpini, E., Baron, P., et al. (1999). Induction of adhesion molecules on human schwann cells by proinflammatory cytokines, an immunofluorescence study. J. Neurol. Sci. 170, 124–130. doi: 10.1016/s0022-510x(99)00202-6

PubMed Abstract | Crossref Full Text | Google Scholar

Cunningham, C. L., Martínez-Cerdeño, V., and Noctor, S. C. (2013). Microglia regulate the number of neural precursor cells in the developing cerebral cortex. J. Neurosci. 33, 4216–4233. doi: 10.1523/JNEUROSCI.3441-12.2013

PubMed Abstract | Crossref Full Text | Google Scholar

De León Reyes, N. S., Bragg-Gonzalo, L., and Nieto, M. (2020). Development and plasticity of the corpus callosum. Development 147:dev189738. doi: 10.1242/dev.189738

PubMed Abstract | Crossref Full Text | Google Scholar

Debernardi, R., Pierre, K., Lengacher, S., Magistretti, P. J., and Pellerin, L. (2003). Cell-specific expression pattern of monocarboxylate transporters in astrocytes and neurons observed in different mouse brain cortical cell cultures. J. Neurosci. Res. 73, 141–155. doi: 10.1002/jnr.10660

PubMed Abstract | Crossref Full Text | Google Scholar

Dyer, A. H., Vahdatpour, C., Sanfeliu, A., and Tropea, D. (2016). The role of Insulin-Like growth factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience 325, 89–99. doi: 10.1016/j.neuroscience.2016.03.056

PubMed Abstract | Crossref Full Text | Google Scholar

Eid, T., Lee, T. W., Patrylo, P., and Zaveri, H. P. (2019). Astrocytes and glutamine synthetase in epileptogenesis. J. Neurosci. Res. 97, 1345–1362. doi: 10.1002/jnr.24267

PubMed Abstract | Crossref Full Text | Google Scholar

Ellison, J. A., Scully, S. A., and De Vellis, J. (1996). Evidence for neuronal regulation of oligodendrocyte development: Cellular localization of platelet-derived growth factor? receptor and A-chain mRNA during cerebral cortex development in the rat. J. Neurosci. Res. 45, 28–39. doi: 10.1002/(sici)1097-4547(19960701)45

Crossref Full Text | Google Scholar

Faraji, R., Ganji, Z., Zamanpour, S. A., Nikparast, F., Akbari-Lalimi, H., and Zare, H. (2023). Impaired white matter integrity in infants and young children with autism spectrum disorder: What evidence does diffusion tensor imaging provide? Psychiatry Res. Neuroimaging 335:111711. doi: 10.1016/j.pscychresns.2023.111711

PubMed Abstract | Crossref Full Text | Google Scholar

Fayol, L., Baud, O., Monier, A., Pellerin, L., Magistretti, P., Evrard, P., et al. (2004). Immunocytochemical expression of monocarboxylate transporters in the human visual cortex at midgestation. Dev. Brain Res. 148, 69–76. doi: 10.1016/j.devbrainres.2003.10.010

PubMed Abstract | Crossref Full Text | Google Scholar

Feigenson, K., Reid, M., See, J., Crenshaw, E. B., and Grinspan, J. B. (2009). Wnt signaling is sufficient to perturb oligodendrocyte maturation. Mol. Cell. Neurosci. 42, 255–265. doi: 10.1016/j.mcn.2009.07.010

PubMed Abstract | Crossref Full Text | Google Scholar

Ferent, J., Zimmer, C., Durbec, P., Ruat, M., and Traiffort, E. (2013). Sonic hedgehog signaling is a positive oligodendrocyte regulator during demyelination. J. Neurosci. 33, 1759–1772. doi: 10.1523/jneurosci.3334-12.2013

PubMed Abstract | Crossref Full Text | Google Scholar

Filipello, F., Morini, R., Corradini, I., Zerbi, V., Canzi, A., Michalski, B., et al. (2018). The microglial innate immune receptor TREM2 is required for synapse elimination and normal brain connectivity. Immunity 48, 979–991.e8. doi: 10.1016/j.immuni.2018.04.016

PubMed Abstract | Crossref Full Text | Google Scholar

Fothergill, T., Donahoo, A.-L. S., Douglass, A., Zalucki, O., Yuan, J., Shu, T., et al. (2014). Netrin-DCC signaling regulates corpus callosum formation through attraction of pioneering axons and by modulating Slit2-mediated repulsion. Cereb. Cortex 24, 1138–1151. doi: 10.1093/cercor/bhs395

PubMed Abstract | Crossref Full Text | Google Scholar

Fouquet, C., Di Meglio, T., Ma, L., Kawasaki, T., Long, H., Hirata, T., et al. (2007). Robo1 and Robo2 control the development of the lateral olfactory tract. J. Neurosci. 27, 3037–3045. doi: 10.1523/jneurosci.0172-07.2007

PubMed Abstract | Crossref Full Text | Google Scholar

Fransen, E., Lemmon, V., Van Camp, G., Vits, L., Coucke, P., and Willems, P. J. (1995). CRASH syndrome: Clinical spectrum of corpus callosum hypoplasia, retardation, adducted thumbs, spastic paraparesis and hydrocephalus due to mutations in one single gene, L1. Eur. J. Hum. Genet. 3, 273–284. doi: 10.1159/000472311

PubMed Abstract | Crossref Full Text | Google Scholar

Frati, G., Luciani, M., Meneghini, V., De Cicco, S., Ståhlman, M., Blomqvist, M., et al. (2018). Human iPSC-based models highlight defective glial and neuronal differentiation from neural progenitor cells in metachromatic leukodystrophy. Cell Death Dis. 9:698. doi: 10.1038/s41419-018-0737-0

PubMed Abstract | Crossref Full Text | Google Scholar

Fruttiger, M., Karlsson, L., Hall, A. C., Abramsson, A., Calver, A. R., Boström, H., et al. (1999). Defective oligodendrocyte development and severe hypomyelination in PDGF-A knockout mice. Development 126, 457–467. doi: 10.1242/dev.126.3.457

PubMed Abstract | Crossref Full Text | Google Scholar

Fujita, Y., and Yamashita, T. (2021). Mechanisms and significance of microglia-axon interactions in physiological and pathophysiological conditions. Cell. Mol. Life Sci. 78, 3907–3919. doi: 10.1007/s00018-021-03758-1

PubMed Abstract | Crossref Full Text | Google Scholar

Fujita, Y., Nakanishi, T., Ueno, M., Itohara, S., and Yamashita, T. (2020). Netrin-G1 regulates microglial accumulation along axons and supports the survival of layer V neurons in the postnatal mouse brain. Cell Rep. 31:107580. doi: 10.1016/j.celrep.2020.107580

PubMed Abstract | Crossref Full Text | Google Scholar

Galichet, C., Clayton, R. W., and Lovell-Badge, R. (2021). Novel tools and investigative approaches for the study of oligodendrocyte precursor cells (NG2-Glia) in CNS development and disease. Front. Cell. Neurosci. 15:673132. doi: 10.3389/fncel.2021.673132

PubMed Abstract | Crossref Full Text | Google Scholar

Gavrish, M., Kustova, A., Celis Suescún, J. C., Bessa, P., Mitina, N., and Tarabykin, V. (2024). Molecular mechanisms of corpus callosum development: A four-step journey. Front. Neuroanat. 17:1276325. doi: 10.3389/fnana.2023.1276325

PubMed Abstract | Crossref Full Text | Google Scholar

Gibson, E. M., Purger, D., Mount, C. W., Goldstein, A. K., Lin, G. L., Wood, L. S., et al. (2014). Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 344:1252304. doi: 10.1126/science.1252304

PubMed Abstract | Crossref Full Text | Google Scholar

Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., et al. (2010). Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845. doi: 10.1126/science.1194637

PubMed Abstract | Crossref Full Text | Google Scholar

Gobius, I., Morcom, L., Suárez, R., Bunt, J., Bukshpun, P., Reardon, W., et al. (2016). Astroglial-mediated remodeling of the interhemispheric midline is required for the formation of the corpus callosum. Cell Rep. 17, 735–747. doi: 10.1016/j.celrep.2016.09.033

PubMed Abstract | Crossref Full Text | Google Scholar

Gobius, I., Suárez, R., Morcom, L., Paolino, A., Edwards, T. J., Kozulin, P., et al. (2017). Astroglial-mediated remodeling of the interhemispheric midline during telencephalic development is exclusive to eutherian mammals. Neural Dev. 12:9. doi: 10.1186/s13064-017-0086-1

PubMed Abstract | Crossref Full Text | Google Scholar

Gómez, M., Hernández, M. L., Pazos, M. R., Tolón, R. M., Romero, J., and Fernández-Ruiz, J. (2008). Colocalization of CB1 receptors with L1 and GAP-43 in forebrain white matter regions during fetal rat brain development: Evidence for a role of these receptors in axonal growth and guidance. Neuroscience 153, 687–699. doi: 10.1016/j.neuroscience.2008.02.038

PubMed Abstract | Crossref Full Text | Google Scholar

Gonda, Y., Namba, T., and Hanashima, C. (2020). Beyond axon guidance: Roles of slit-robo signaling in neocortical formation. Front. Cell Dev. Biol. 8:607415. doi: 10.3389/fcell.2020.607415

PubMed Abstract | Crossref Full Text | Google Scholar

Goshi, N., Morgan, R. K., Lein, P. J., and Seker, E. (2020). A primary neural cell culture model to study neuron, astrocyte, and microglia interactions in neuroinflammation. J. Neuroinflammation 17:155. doi: 10.1186/s12974-020-01819-z

PubMed Abstract | Crossref Full Text | Google Scholar

Goursaud, S., Kozlova, E. N., Maloteaux, J., and Hermans, E. (2009). Cultured astrocytes derived from corpus callosum or cortical grey matter show distinct glutamate handling properties. J. Neurochem. 108, 1442–1452. doi: 10.1111/j.1471-4159.2009.05889.x

PubMed Abstract | Crossref Full Text | Google Scholar

Groeschel, S., Kehrer, C., Engel, C., Dali, C., Bley, A., Steinfeld, R., et al. (2011). Metachromatic leukodystrophy: Natural course of cerebral MRI changes in relation to clinical course. J. Inher. Metab. Dis. 34, 1095–1102. doi: 10.1007/s10545-011-9361-1

PubMed Abstract | Crossref Full Text | Google Scholar

Guedes, J. R., Ferreira, P. A., Costa, J. M., Cardoso, A. L., and Peça, J. (2022). Microglia-dependent remodeling of neuronal circuits. J. Neurochem. 163, 74–93. doi: 10.1111/jnc.15689

PubMed Abstract | Crossref Full Text | Google Scholar

Guo, C., Chom, K. S., Li, Y., Tchedre, K., Antolik, C., Ma, J., et al. (2018). IGFBPL1 regulates axon growth through IGF-1-mediated signaling cascades. Sci. Rep. 8:2054. doi: 10.1038/s41598-018-20463-5

PubMed Abstract | Crossref Full Text | Google Scholar

Gzielo, K., and Nikiforuk, A. (2021). Astroglia in autism spectrum disorder. IJMS 22:11544. doi: 10.3390/ijms222111544

PubMed Abstract | Crossref Full Text | Google Scholar

Haigh, S. M., Eack, S. M., Keller, T., Minshew, N. J., and Behrmann, M. (2019). White matter structure in schizophrenia and autism: Abnormal diffusion across the brain in schizophrenia. Neuropsychologia 135:107233. doi: 10.1016/j.neuropsychologia.2019.107233

PubMed Abstract | Crossref Full Text | Google Scholar

Han, R. T., Vainchtein, I. D., Schlachetzki, J. C. M., Cho, F. S., Dorman, L. C., Ahn, E., et al. (2023). Microglial pattern recognition via IL-33 promotes synaptic refinement in developing corticothalamic circuits in mice. J. Exp. Med. 220:e20220605. doi: 10.1084/jem.20220605

PubMed Abstract | Crossref Full Text | Google Scholar

Han, Z., Luo, N., Ma, W., Liu, X., Cai, Y., Kou, J., et al. (2023). AAV11 enables efficient retrograde targeting of projection neurons and enhances astrocyte-directed transduction. Nat. Commun. 14:3792. doi: 10.1038/s41467-023-39554-7

PubMed Abstract | Crossref Full Text | Google Scholar

Hoshiko, M., Arnoux, I., Avignone, E., Yamamoto, N., and Audinat, E. (2012). Deficiency of the microglial receptor CX3CR1 impairs postnatal functional development of thalamocortical synapses in the barrel cortex. J. Neurosci. 32, 15106–15111. doi: 10.1523/JNEUROSCI.1167-12.2012

PubMed Abstract | Crossref Full Text | Google Scholar

Hu, N.-Y., Chen, Y.-T., Wang, Q., Jie, W., Liu, Y.-S., You, Q.-L., et al. (2020). Expression patterns of inducible cre recombinase driven by differential astrocyte-specific promoters in transgenic mouse lines. Neurosci. Bull. 36, 530–544. doi: 10.1007/s12264-019-00451-z

PubMed Abstract | Crossref Full Text | Google Scholar

Hu, X., Xiao, G., He, L., Niu, X., Lou, T., Hu, Q., et al. (2021). Sustained ErbB activation causes demyelination and hypomyelination by driving necroptosis of mature oligodendrocytes and apoptosis of oligodendrocyte precursor cells. J. Neurosci. 41, 9872–9890. doi: 10.1523/JNEUROSCI.2922-20.2021

PubMed Abstract | Crossref Full Text | Google Scholar

Hu, X., Zhu, Q., Lou, T., Hu, Q., Li, H., Xu, Y., et al. (2024). Pan-ErbB inhibition impairs cognition via disrupting myelination and aerobic glycolysis in oligodendrocytes. Proc. Natl. Acad. Sci. U.S.A. 121:e2405152121. doi: 10.1073/pnas.2405152121

PubMed Abstract | Crossref Full Text | Google Scholar

Hu, Z., Yue, X., Shi, G., Yue, Y., Crockett, D. P., Blair-Flynn, J., et al. (2003). Corpus callosum deficiency in transgenic mice expressing a truncated Ephrin-A receptor. J. Neurosci. 23, 10963–10970. doi: 10.1523/JNEUROSCI.23-34-10963.2003

PubMed Abstract | Crossref Full Text | Google Scholar

Hughes, A. N. (2021). Glial cells promote myelin formation and elimination. Front. Cell Dev. Biol. 9:661486. doi: 10.3389/fcell.2021.661486

PubMed Abstract | Crossref Full Text | Google Scholar

Irfan, M., Evonuk, K. S., and DeSilva, T. M. (2022). Microglia phagocytose oligodendrocyte progenitor cells and synapses during early postnatal development: Implications for white versus gray matter maturation. FEBS J. 289, 2110–2127. doi: 10.1111/febs.16190

PubMed Abstract | Crossref Full Text | Google Scholar

Islam, S. M., Shinmyo, Y., Okafuji, T., Su, Y., Naser, I. B., Ahmed, G., et al. (2009). Draxin, a repulsive guidance protein for spinal cord and forebrain commissures. Science 323, 388–393. doi: 10.1126/science.1165187

PubMed Abstract | Crossref Full Text | Google Scholar

Kang, D., Yang, Y. R., Lee, C., Park, B., Park, K. I., Seo, J. K., et al. (2018). Netrin-1/ DCC -mediated PLC γ1 activation is required for axon guidance and brain structure development. EMBO Rep. 19:e46250. doi: 10.15252/embr.201846250

PubMed Abstract | Crossref Full Text | Google Scholar

Katz, M. J., Lasek, R. J., and Silver, J. (1983). Ontophyletics of the nervous system: Development of the corpus callosum and evolution of axon tracts. Proc. Natl. Acad. Sci. U.S.A. 80, 5936–5940. doi: 10.1073/pnas.80.19.5936

PubMed Abstract | Crossref Full Text | Google Scholar

Kennedy, L., Morland, C., Narum, M., Bergersen, L. H., and Rinholm, J. E. (2025). Lactate receptor HCAR1 affects axonal development and contributes to Lactate’s protection of axons and myelin in experimental neonatal hypoglycemia. eNeuro 12:ENEURO.0563-24.2025. doi: 10.1523/eneuro.0563-24.2025

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, J. Y., Sun, Q., Oglesbee, M., and Yoon, S. O. (2003). The role of ErbB2 signaling in the onset of terminal differentiation of oligodendrocytes In Vivo. J. Neurosci. 23, 5561–5571. doi: 10.1523/jneurosci.23-13-05561.2003

PubMed Abstract | Crossref Full Text | Google Scholar

Knowles, J. K., Batra, A., Xu, H., and Monje, M. (2022). Adaptive and maladaptive myelination in health and disease. Nat. Rev. Neurol. 18, 735–746. doi: 10.1038/s41582-022-00737-3

PubMed Abstract | Crossref Full Text | Google Scholar

Kucukdereli, H., Allen, N. J., Lee, A. T., Feng, A., Ozlu, M. I., Conatser, L. M., et al. (2011). Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc. Natl. Acad. Sci. U.S.A. 108, E440–E449. doi: 10.1073/pnas.1104977108

PubMed Abstract | Crossref Full Text | Google Scholar

Kuhn, S., Gritti, L., Crooks, D., and Dombrowski, Y. (2019). Oligodendrocytes in development, myelin generation and beyond. Cells 8:1424. doi: 10.3390/cells8111424

PubMed Abstract | Crossref Full Text | Google Scholar

Lagos-Cabré, R., Burgos-Bravo, F., Avalos, A. M., and Leyton, L. (2020). Connexins in astrocyte migration. Front. Pharmacol. 10:1546. doi: 10.3389/fphar.2019.01546

PubMed Abstract | Crossref Full Text | Google Scholar

Laukka, J. J., Makki, M. I., Lafleur, T., Stanley, J., Kamholz, J., and Garbern, J. Y. (2014). Diffusion tensor imaging of patients with proteolipid protein 1 gene mutations. J. Neurosci. Res. 92, 1723–1732. doi: 10.1002/jnr.23458

PubMed Abstract | Crossref Full Text | Google Scholar

Lawrence, A. R., Canzi, A., Bridlance, C., Olivié, N., Lansonneur, C., Catale, C., et al. (2024). Microglia maintain structural integrity during fetal brain morphogenesis. Cell 187, 962–980.e19. doi: 10.1016/j.cell.2024.01.012

PubMed Abstract | Crossref Full Text | Google Scholar

Levitt, P. (2003). Structural and functional maturation of the developing primate brain. J. Pediatr. 143, 35–45. doi: 10.1067/S0022-3476(03)00400-1

PubMed Abstract | Crossref Full Text | Google Scholar

Li, Q., Cheng, Z., Zhou, L., Darmanis, S., Neff, N. F., Okamoto, J., et al. (2019). Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing. Neuron 101, 207–223.e10. doi: 10.1016/j.neuron.2018.12.006

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, J., Xiao, Q., Xiao, J., Niu, C., Li, Y., Zhang, X., et al. (2022). Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Sig. Transduct Target Ther. 7:3. doi: 10.1038/s41392-021-00762-6

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, Y., Shen, X., Zhang, Y., Zheng, X., Cepeda, C., Wang, Y., et al. (2023). Interactions of glial cells with neuronal synapses, from astrocytes to microglia and oligodendrocyte lineage cells. Glia 71, 1383–1401. doi: 10.1002/glia.24343

PubMed Abstract | Crossref Full Text | Google Scholar

Livet, J., Weissman, T. A., Kang, H., Draft, R. W., Lu, J., Bennis, R. A., et al. (2007). Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature 450, 56–62. doi: 10.1038/nature06293

PubMed Abstract | Crossref Full Text | Google Scholar

Long, H., Sabatier, C., Le, Ma, Plump, A., Yuan, W., et al. (2004). Conserved roles for slit and robo proteins in midline commissural axon guidance. Neuron 42, 213–223. doi: 10.1016/s0896-6273(04)00179-5

PubMed Abstract | Crossref Full Text | Google Scholar

Luciani, M., Garsia, C., Beretta, S., Cifola, I., Peano, C., Merelli, I., et al. (2024). Human iPSC-derived neural stem cells displaying radial glia signature exhibit long-term safety in mice. Nat. Commun. 15:9433. doi: 10.1038/s41467-024-53613-7

PubMed Abstract | Crossref Full Text | Google Scholar

Luo, P., Chu, S.-F., Zhang, Z., Xia, C.-Y., and Chen, N.-H. (2019). Fractalkine/CX3CR1 is involved in the cross-talk between neuron and glia in neurological diseases. Brain Res. Bull. 146, 12–21. doi: 10.1016/j.brainresbull.2018.11.017

PubMed Abstract | Crossref Full Text | Google Scholar

Luo, Y., and Wang, Z. (2024). The impact of microglia on neurodevelopment and brain function in autism. Biomedicines 12:210. doi: 10.3390/biomedicines12010210

PubMed Abstract | Crossref Full Text | Google Scholar

Luongo, C., Butruille, L., Sébillot, A., Le Blay, K., Schwaninger, M., Heuer, H., et al. (2021). Absence of both thyroid hormone transporters MCT8 and OATP1C1 impairs neural stem cell fate in the adult mouse subventricular zone. Stem Cell. Rep. 16, 337–353. doi: 10.1016/j.stemcr.2020.12.009

PubMed Abstract | Crossref Full Text | Google Scholar

Magnani, D., Hasenpusch-Theil, K., Benadiba, C., Yu, T., Basson, M. A., Price, D. J., et al. (2014). Gli3 controls corpus callosum formation by positioning midline guideposts during telencephalic patterning. Cereb. Cortex 24, 186–198. doi: 10.1093/cercor/bhs303

PubMed Abstract | Crossref Full Text | Google Scholar

Marsh, A. P. L., Edwards, T. J., Galea, C., Cooper, H. M., Engle, E. C., Jamuar, S. S., et al. (2018). DCCmutation update: Congenital mirror movements, isolated agenesis of the corpus callosum, and developmental split brain syndrome. Hum. Mutation 39, 23–39. doi: 10.1002/humu.23361

PubMed Abstract | Crossref Full Text | Google Scholar

Marsh, A. P. L., Heron, D., Edwards, T. J., Quartier, A., Galea, C., Nava, C., et al. (2017). Mutations in DCC cause isolated agenesis of the corpus callosum with incomplete penetrance. Nat. Genet. 49, 511–514. doi: 10.1038/ng.3794

PubMed Abstract | Crossref Full Text | Google Scholar

Martin, L. A., Hsu, F., Herd, B., Gregg, M., Sample, H., and Kaplan, J. (2021). Executive functions in agenesis of the corpus callosum: Working memory and sustained attention in the BTBR inbred mouse strain. Brain Behav. 11:e01933. doi: 10.1002/brb3.1933

PubMed Abstract | Crossref Full Text | Google Scholar

Mason, S. (2017). Lactate shuttles in neuroenergetics—homeostasis, allostasis and beyond. Front. Neurosci. 11:43. doi: 10.3389/fnins.2017.00043

PubMed Abstract | Crossref Full Text | Google Scholar

Matthiesen, I., Nasiri, R., Tamashiro Orrego, A., Winkler, T. E., and Herland, A. (2022). Metabolic assessment of human induced pluripotent stem cells-derived astrocytes and fetal primary astrocytes: Lactate and glucose turnover. Biosensors 12:839. doi: 10.3390/bios12100839

PubMed Abstract | Crossref Full Text | Google Scholar

McNamara, N. B., Munro, D. A. D., Bestard-Cuche, N., Uyeda, A., Bogie, J. F. J., Hoffmann, A., et al. (2023). Microglia regulate central nervous system myelin growth and integrity. Nature 613, 120–129. doi: 10.1038/s41586-022-05534-y

PubMed Abstract | Crossref Full Text | Google Scholar

Meyer, N., Richter, N., Fan, Z., Siemonsmeier, G., Pivneva, T., Jordan, P., et al. (2018). Oligodendrocytes in the mouse corpus callosum maintain axonal function by delivery of glucose. Cell Rep. 22, 2383–2394. doi: 10.1016/j.celrep.2018.02.022

PubMed Abstract | Crossref Full Text | Google Scholar

Meyer-Franke, A., Shen, S., and Barres, B. A. (1999). Astrocytes induce oligodendrocyte processes to align with and adhere to axons. Mol. Cell. Neurosci. 14, 385–397. doi: 10.1006/mcne.1999.0788

PubMed Abstract | Crossref Full Text | Google Scholar

Mire, E., Hocine, M., Bazellières, E., Jungas, T., Davy, A., Chauvet, S., et al. (2018). Developmental upregulation of Ephrin-B1 silences Sema3C/Neuropilin-1 signaling during post-crossing navigation of corpus callosum axons. Curr. Biol. 28, 1768–1782.e4. doi: 10.1016/j.cub.2018.04.026

PubMed Abstract | Crossref Full Text | Google Scholar

Mitew, S., Hay, C. M., Peckham, H., Xiao, J., Koenning, M., and Emery, B. (2014). Mechanisms regulating the development of oligodendrocytes and central nervous system myelin. Neuroscience 276, 29–47. doi: 10.1016/j.neuroscience.2013.11.029

PubMed Abstract | Crossref Full Text | Google Scholar

Morcom, L., Edwards, T. J., Rider, E., Jones-Davis, D., Lim, J. W., Chen, K.-S., et al. (2021a). DRAXIN regulates interhemispheric fissure remodelling to influence the extent of corpus callosum formation. eLife 10:e61618. doi: 10.7554/elife.61618

PubMed Abstract | Crossref Full Text | Google Scholar

Morcom, L., Gobius, I., Marsh, A. P., Suárez, R., Lim, J. W., Bridges, C., et al. (2021b). DCC regulates astroglial development essential for telencephalic morphogenesis and corpus callosum formation. eLife 10:e61769. doi: 10.7554/eLife.61769

PubMed Abstract | Crossref Full Text | Google Scholar

Morrison, B. M., Lee, Y., and Rothstein, J. D. (2013). Oligodendroglia: Metabolic supporters of axons. Trends Cell Biol. 23, 644–651. doi: 10.1016/j.tcb.2013.07.007

PubMed Abstract | Crossref Full Text | Google Scholar

Mount, C. W., and Monje, M. (2017). Wrapped to adapt: Experience-dependent myelination. Neuron 95, 743–756. doi: 10.1016/j.neuron.2017.07.009

PubMed Abstract | Crossref Full Text | Google Scholar

Nemes-Baran, A. D., White, D. R., and DeSilva, T. M. (2020). Fractalkine-dependent microglial pruning of viable oligodendrocyte progenitor cells regulates myelination. Cell Rep. 32:108047. doi: 10.1016/j.celrep.2020.108047

PubMed Abstract | Crossref Full Text | Google Scholar

Neniskyte, U., and Gross, C. T. (2017). Errant gardeners: Glial-cell-dependent synaptic pruning and neurodevelopmental disorders. Nat. Rev. Neurosci. 18, 658–670. doi: 10.1038/nrn.2017.110

PubMed Abstract | Crossref Full Text | Google Scholar

Nery, S., Wichterle, H., and Fishell, G. (2001). Sonic hedgehog contributes to oligodendrocyte specification in the mammalian forebrain. Development 128, 527–540. doi: 10.1242/dev.128.4.527

PubMed Abstract | Crossref Full Text | Google Scholar

Nicholson, M., Wood, R. J., Gonsalvez, D. G., Hannan, A. J., Fletcher, J. L., Xiao, J., et al. (2022). Remodelling of myelinated axons and oligodendrocyte differentiation is stimulated by environmental enrichment in the young adult brain. Eur. J. Neurosci. 56, 6099–6114. doi: 10.1111/ejn.15840

PubMed Abstract | Crossref Full Text | Google Scholar

Paolicelli, R. C., Bisht, K., and Tremblay, M. -Ã (2014). Fractalkine regulation of microglial physiology and consequences on the brain and behavior. Front. Cell. Neurosci. 8:129. doi: 10.3389/fncel.2014.00129

PubMed Abstract | Crossref Full Text | Google Scholar

Paolicelli, R. C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M., Panzanelli, P., et al. (2011). Synaptic pruning by microglia is necessary for normal brain development. Science 333, 1456–1458. doi: 10.1126/science.1202529

PubMed Abstract | Crossref Full Text | Google Scholar

Paul, L. K. (2011). Developmental malformation of the corpus callosum: A review of typical callosal development and examples of developmental disorders with callosal involvement. J. Neurodevelop. Disord. 3, 3–27. doi: 10.1007/s11689-010-9059-y

PubMed Abstract | Crossref Full Text | Google Scholar

Pawelec, P., Ziemka-Nalecz, M., Sypecka, J., and Zalewska, T. (2020). The Impact of the CX3CL1/CX3CR1 axis in neurological disorders. Cells 9:2277. doi: 10.3390/cells9102277

PubMed Abstract | Crossref Full Text | Google Scholar

Peotter, J. L., Pustova, I., Lettman, M. M., Shatadal, S., Bradberry, M. M., Winter-Reed, A. D., et al. (2022). TFG regulates secretory and endosomal sorting pathways in neurons to promote their activity and maintenance. Proc. Natl. Acad. Sci. U.S.A. 119:e2210649119. doi: 10.1073/pnas.2210649119

PubMed Abstract | Crossref Full Text | Google Scholar

Pont-Lezica, L., Beumer, W., Colasse, S., Drexhage, H., Versnel, M., and Bessis, A. (2014). Microglia shape corpus callosum axon tract fasciculation: Functional impact of prenatal inflammation. Eur. J. Neurosci. 39, 1551–1557. doi: 10.1111/ejn.12508

PubMed Abstract | Crossref Full Text | Google Scholar

Rapti, G. (2023). Regulation of axon pathfinding by astroglia across genetic model organisms. Front. Cell. Neurosci. 17:1241957. doi: 10.3389/fncel.2023.1241957

PubMed Abstract | Crossref Full Text | Google Scholar

Relucio, J., Tzvetanova, I. D., Ao, W., Lindquist, S., and Colognato, H. (2009). Laminin alters fyn regulatory mechanisms and promotes oligodendrocyte development. J. Neurosci. 29, 11794–11806. doi: 10.1523/JNEUROSCI.0888-09.2009

PubMed Abstract | Crossref Full Text | Google Scholar

Rinholm, J. E., Hamilton, N. B., Kessaris, N., Richardson, W. D., Bergersen, L. H., and Attwell, D. (2011). Regulation of oligodendrocyte development and myelination by glucose and lactate. J. Neurosci. 31, 538–548. doi: 10.1523/JNEUROSCI.3516-10.2011

PubMed Abstract | Crossref Full Text | Google Scholar

Rodríguez-Pérez, L. M., López-de-San-Sebastián, J., De Diego, I., Smith, A., Roales-Buján, R., Jiménez, A. J., et al. (2024). A selective defect in the glial wedge as part of the neuroepithelium disruption in hydrocephalus development in the mouse hyh model is associated with complete corpus callosum dysgenesis. Front. Cell. Neurosci. 18:1330412. doi: 10.3389/fncel.2024.1330412

PubMed Abstract | Crossref Full Text | Google Scholar

Roqué, P. J., and Costa, L. G. (2017). Co-culture of neurons and microglia. CP Toxicol. 74, 11.24.1–11.24.17. doi: 10.1002/cptx.32

PubMed Abstract | Crossref Full Text | Google Scholar

Rusin, D., Vahl Becirovic, L., Lyszczarz, G., Krueger, M., Benmamar-Badel, A., Vad Mathiesen, C., et al. (2024). Microglia-derived insulin-like growth factor 1 is critical for neurodevelopment. Cells 13:184. doi: 10.3390/cells13020184

PubMed Abstract | Crossref Full Text | Google Scholar

Sabo, J. K., Aumann, T. D., Merlo, D., Kilpatrick, T. J., and Cate, H. S. (2011). Remyelination is altered by bone morphogenic protein signaling in demyelinated lesions. J. Neurosci. 31, 4504–4510. doi: 10.1523/JNEUROSCI.5859-10.2011

PubMed Abstract | Crossref Full Text | Google Scholar

Sagi-Dain, L., Kurolap, A., Ilivitzki, A., Mory, A., Paperna, T., Regeneron Genetics Center, et al. (2020). A novel heterozygous loss-of-function DCC Netrin 1 receptor variant in prenatal agenesis of corpus callosum and review of the literature. Am. J. Med. Genet. Pt A 182, 205–212. doi: 10.1002/ajmg.a.61404

PubMed Abstract | Crossref Full Text | Google Scholar

Sahasrabuddhe, V., and Ghosh, H. S. (2022). Cx3Cr1-Cre induction leads to microglial activation and IFN-1 signaling caused by DNA damage in early postnatal brain. Cell Rep. 38:110252. doi: 10.1016/j.celrep.2021.110252

PubMed Abstract | Crossref Full Text | Google Scholar

Santos, E. N., and Fields, R. D. (2021). Regulation of myelination by microglia. Sci. Adv. 7:eabk1131. doi: 10.1126/sciadv.abk1131

PubMed Abstract | Crossref Full Text | Google Scholar

Shimizu, S., Abt, A., and Meucci, O. (2011). Bilaminar co-culture of primary rat cortical neurons and glia. J. Vis. Exp. 57:3257. doi: 10.3791/3257

PubMed Abstract | Crossref Full Text | Google Scholar

Shu, T., and Richards, L. J. (2001). Cortical axon guidance by the glial wedge during the development of the corpus callosum. J. Neurosci. 21, 2749–2758. doi: 10.1523/JNEUROSCI.21-08-02749.2001

PubMed Abstract | Crossref Full Text | Google Scholar

Shu, T., Puche, A. C., and Richards, L. J. (2003). Development of midline glial populations at the corticoseptal boundary. J. Neurobiol. 57, 81–94. doi: 10.1002/neu.10252

PubMed Abstract | Crossref Full Text | Google Scholar

Silver, J., Lorenz, S. E., Wahlsten, D., and Coughlin, J. (1982). Axonal guidance during development of the great cerebral commissures: Descriptive and experimental studies, in vivo, on the role of preformed glial pathways. J. Comp. Neurol. 210, 10–29. doi: 10.1002/cne.902100103

PubMed Abstract | Crossref Full Text | Google Scholar

Simons, M., and Nave, K.-A. (2016). Oligodendrocytes: Myelination and axonal support. Cold Spring Harb. Perspect. Biol. 8:a020479. doi: 10.1101/cshperspect.a020479

PubMed Abstract | Crossref Full Text | Google Scholar

Simons, M., Gibson, E. M., and Nave, K.-A. (2024). Oligodendrocytes: Myelination, plasticity, and axonal support. Cold Spring Harb. Perspect. Biol. 16:a041359. doi: 10.1101/cshperspect.a041359

PubMed Abstract | Crossref Full Text | Google Scholar

Smith, J. A., Das, A., Ray, S. K., and Banik, N. L. (2012). Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res. Bull. 87, 10–20. doi: 10.1016/j.brainresbull.2011.10.004

PubMed Abstract | Crossref Full Text | Google Scholar

Song, I., and Dityatev, A. (2018). Crosstalk between glia, extracellular matrix and neurons. Brain Res. Bull. 136, 101–108. doi: 10.1016/j.brainresbull.2017.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

Squarzoni, P., Oller, G., Hoeffel, G., Pont-Lezica, L., Rostaing, P., Low, D., et al. (2014). Microglia modulate wiring of the embryonic forebrain. Cell Rep. 8, 1271–1279. doi: 10.1016/j.celrep.2014.07.042

PubMed Abstract | Crossref Full Text | Google Scholar

Stevens, B., Allen, N. J., Vazquez, L. E., Howell, G. R., Christopherson, K. S., Nouri, N., et al. (2007). The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178. doi: 10.1016/j.cell.2007.10.036

PubMed Abstract | Crossref Full Text | Google Scholar

Stogsdill, J. A., Harwell, C. C., and Goldman, S. A. (2023). Astrocytes as master modulators of neural networks: Synaptic functions and disease-associated dysfunction of astrocytes. Ann. N. Y. Acad. Sci. 1525, 41–60. doi: 10.1111/nyas.15004

PubMed Abstract | Crossref Full Text | Google Scholar

Suarez, R., Gobius, I., and Richards, L. J. (2014). Evolution and development of interhemispheric connections in the vertebrate forebrain. Front. Hum. Neurosci. 8:497. doi: 10.3389/fnhum.2014.00497

PubMed Abstract | Crossref Full Text | Google Scholar

Sullivan, G. M., Knutsen, A. K., Peruzzotti-Jametti, L., Korotcov, A., Bosomtwi, A., Dardzinski, B. J., et al. (2020). Transplantation of induced neural stem cells (iNSCs) into chronically demyelinated corpus callosum ameliorates motor deficits. Acta Neuropathol. Commun. 8:84. doi: 10.1186/s40478-020-00960-3

PubMed Abstract | Crossref Full Text | Google Scholar

Szepesi, Z., Manouchehrian, O., Bachiller, S., and Deierborg, T. (2018). Bidirectional microglia–neuron communication in health and disease. Front. Cell. Neurosci. 12:323. doi: 10.3389/fncel.2018.00323

PubMed Abstract | Crossref Full Text | Google Scholar

Tamada, A., Kumada, T., Zhu, Y., Matsumoto, T., Hatanaka, Y., Muguruma, K., et al. (2008). Crucial roles of Robo proteins in midline crossing of cerebellofugal axons and lack of their up-regulation after midline crossing. Neural Dev. 3:29. doi: 10.1186/1749-8104-3-29

PubMed Abstract | Crossref Full Text | Google Scholar

Taveggia, C., Thaker, P., Petrylak, A., Caporaso, G. L., Toews, A., Falls, D. L., et al. (2008). Type III neuregulin-1 promotes oligodendrocyte myelination. Glia 56, 284–293. doi: 10.1002/glia.20612

PubMed Abstract | Crossref Full Text | Google Scholar

Thion, M. S., Ginhoux, F., and Garel, S. (2018). Microglia and early brain development: An intimate journey. Science 362, 185–189. doi: 10.1126/science.aat0474

PubMed Abstract | Crossref Full Text | Google Scholar

Tripathi, R. B., Jackiewicz, M., McKenzie, I. A., Kougioumtzidou, E., Grist, M., and Richardson, W. D. (2017). Remarkable stability of myelinating oligodendrocytes in mice. Cell Rep. 21, 316–323. doi: 10.1016/j.celrep.2017.09.050

PubMed Abstract | Crossref Full Text | Google Scholar

Unni, D. K., Piper, M., Moldrich, R. X., Gobius, I., Liu, S., Fothergill, T., et al. (2012). Multiple Slits regulate the development of midline glial populations and the corpus callosum. Dev. Biol. 365, 36–49. doi: 10.1016/j.ydbio.2012.02.004

PubMed Abstract | Crossref Full Text | Google Scholar

Vainchtein, I. D., Chin, G., Cho, F. S., Kelley, K. W., Miller, J. G., Chien, E. C., et al. (2018). Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science 359, 1269–1273. doi: 10.1126/science.aal3589

PubMed Abstract | Crossref Full Text | Google Scholar

van der Knaap, M. S., and Bugiani, M. (2017). Leukodystrophies: A proposed classification system based on pathological changes and pathogenetic mechanisms. Acta neuropathologica 134, 351–382. doi: 10.1007/s00401-017-1739-1

PubMed Abstract | Crossref Full Text | Google Scholar

Vega-Pons, S., Olivetti, E., Avesani, P., Dodero, L., Gozzi, A., and Bifone, A. (2017). Differential effects of brain disorders on structural and functional connectivity. Front. Neurosci. 10:605. doi: 10.3389/fnins.2016.00605

PubMed Abstract | Crossref Full Text | Google Scholar

Verkhratsky, A., and Nedergaard, M. (2018). Physiology of astroglia. Physiol. Rev. 98, 239–389. doi: 10.1152/physrev.00042.2016

PubMed Abstract | Crossref Full Text | Google Scholar

Viscomi, M. P., Czyrska, J., Winiarczyk, D., Ziętek, M. M., and Sampino, S. (2025). The BTBR T+ Itpr3tf/J mouse strain as a model to study the genetic, immune, and metabolic origins of neurodevelopmental disorders. Res. Autism Spectr. Disord. 119:102526. doi: 10.1016/j.rasd.2024.102526

Crossref Full Text | Google Scholar

Wahlsten, D. (1987). Defects of the fetal forebrain in mice with hereditary agenesis of the corpus callosum. J. Comp. Neurol. 262, 227–241. doi: 10.1002/cne.902620205

PubMed Abstract | Crossref Full Text | Google Scholar

Wahlsten, D. (1989). Deficiency of the corpus callosum: Incomplete penetrance and substrain differentiation in BALB/c mice. J. Neurogenet. 5, 61–76. doi: 10.3109/01677068909167265

PubMed Abstract | Crossref Full Text | Google Scholar

Wahlsten, D., and Schalomon, P. M. (1994). A new hybrid mouse model for agenesis of the corpus callosum. Behav. Brain Res. 64, 111–117. doi: 10.1016/0166-4328(94)90123-6

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, W., Wang, J., Tang, Q., Zhu, X., Zhu, R., Cui, D., et al. (2021). CX3CR1 deficiency aggravates brain white matter injury and affects expression of the CD36/15LO/NR4A1 signal. Biochem. Biophys. Res. Commun. 549, 47–53. doi: 10.1016/j.bbrc.2021.02.053

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, S.-R., and Nowakowski, T. J. (2025). Exploring human brain development and disease using assembloids. Neuron 113, 1133–1150. doi: 10.1016/j.neuron.2025.02.010

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, W., and Chan, J. R. (2020). Myelin plasticity: Sculpting circuits in learning and memory. Nat. Rev. Neurosci. 21, 682–694. doi: 10.1038/s41583-020-00379-8

PubMed Abstract | Crossref Full Text | Google Scholar

Yamasaki, M., Thompson, P., and Lemmon, V. (1997). CRASH syndrome: Mutations in L1CAM correlate with severity of the disease. Neuropediatrics 28, 175–178. doi: 10.1055/s-2007-973696

PubMed Abstract | Crossref Full Text | Google Scholar

Yamazaki, R., and Ohno, N. (2025). The potential of repurposing clemastine to promote remyelination. Front. Cell. Neurosci. 19:1582902. doi: 10.3389/fncel.2025.1582902

PubMed Abstract | Crossref Full Text | Google Scholar

Yan, S., Wang, L., Samsom, J. N., Ujic, D., and Liu, F. (2024). PolyI:C maternal immune activation on E9.5 causes the deregulation of microglia and the complement system in mice, leading to decreased synaptic spine density. IJMS 25:5480. doi: 10.3390/ijms25105480

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, J., Ruchti, E., Petit, J. M., Jourdain, P., Grenningloh, G., Allaman, I., et al. (2014). Lactate promotes plasticity gene expression by potentiating NMDA signaling in neurons. Proc. Natl. Acad. Sci. U.S.A. 111, 12228–12233. doi: 10.1073/pnas.1322912111

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, S. M., Michel, K., Jokhi, V., Nedivi, E., and Arlotta, P. (2020). Neuron class–specific responses govern adaptive myelin remodeling in the neocortex. Science 370:eabd2109. doi: 10.1126/science.abd2109

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, B., Xu, Y., Zhu, B., and Kantarci, K. (2014). The role of diffusion tensor imaging in detecting microstructural changes in prodromal Alzheimer’s disease. CNS Neurosci. Ther. 20, 3–9. doi: 10.1111/cns.12166

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, J., Zhang, L., Yi, S., Jiang, X., Qiao, Y., Zhang, Y., et al. (2025). Mouse astrocytes promote microglial ramification by releasing TGF-β and forming glial fibers. Front. Cell. Neurosci. 14:195. doi: 10.3389/fncel.2020.00195

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, W., Luo, P., Liu, X., Cheng, R., Zhang, S., Qian, X., et al. (2023). Roles of fibroblast growth factors in the axon guidance. IJMS 24:10292.

Google Scholar

Zhang, X., Dong, N., and Hu, X. (2023). Wnt/β-catenin signaling inhibitors. CTMC 23, 880–896. doi: 10.2174/1568026623666230303101810

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Argaw, A. T., Gurfein, B. T., Zameer, A., Snyder, B. J., Ge, C., et al. (2009). Notch1 signaling plays a role in regulating precursor differentiation during CNS remyelination. Proc. Natl. Acad. Sci. U. S. A. 106, 19162–19167. doi: 10.1073/pnas.0902834106

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Chen, K., Sloan, S. A., Bennett, M. L., Scholze, A. R., O’Keeffe, S., et al. (2014). An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947. doi: 10.1523/JNEUROSCI.1860-14.2014

PubMed Abstract | Crossref Full Text | Google Scholar

Zhi, Y., Yu, J., Zhong, Y., Fu, H., Zhou, X., Yi, W., et al. (2025). WDR62 controls cortical radial migration and callosal projection of neurons in the developing cerebral cortex. Neurobiol. Dis. 211:106951. doi: 10.1016/j.nbd.2025.106951

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: corpus callosum, neurons, astrocytes, oligodendrocytes, microglia, animal models

Citation: Czyrska J, Ziętek MM, Bernat A and Sampino S (2025) Glial interactions in the formation and plasticity of the corpus callosum. Front. Cell. Neurosci. 19:1690400. doi: 10.3389/fncel.2025.1690400

Received: 21 August 2025; Revised: 31 October 2025; Accepted: 10 November 2025;
Published: 27 November 2025.

Edited by:

Bo Hu, Houston Methodist Research Institute, United States

Reviewed by:

Veronica Lopez-Virgen, University of Colima, Mexico
Maria Sergeevna Gavrish, Lobachevsky State University of Nizhny Novgorod, Russia

Copyright © 2025 Czyrska, Ziętek, Bernat and Sampino. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Silvestre Sampino, cy5zYW1waW5vQGlnYnpwYW4ucGw=

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.