Next Article in Journal
Mapping the Nicotinic Acetylcholine Receptor Nanocluster Topography at the Cell Membrane with STED and STORM Nanoscopies
Next Article in Special Issue
Altered Expression of AQP1 and AQP4 in Brain Barriers and Cerebrospinal Fluid May Affect Cerebral Water Balance during Chronic Hypertension
Previous Article in Journal
Roles of Species-Specific Legumains in Pathogenicity of the Pinewood Nematode Bursaphelenchus xylophilus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

AQP4, Astrogenesis, and Hydrocephalus: A New Neurological Perspective

by
Leandro Castañeyra-Ruiz
1,*,
Ibrahim González-Marrero
2,
Luis G. Hernández-Abad
2,
Seunghyun Lee
1,
Agustín Castañeyra-Perdomo
2,3,† and
Michael Muhonen
4,†
1
CHOC Children’s Research Institute, 1201 W, La Veta Avenue, Orange, CA 92868, USA
2
Departamento de Ciencias Médicas Basicas, Anatomía, Facultad de Medicina, Universidad de La Laguna, Ofra s/n, 38071 La Laguna, Spain
3
Instituto de Investigación y Ciencias de Puerto del Rosario, 35600 Puerto del Rosario, Spain
4
Neurosurgery Department at CHOC Children’s Hospital, 505 S Main St., Orange, CA 92868, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(18), 10438; https://doi.org/10.3390/ijms231810438
Submission received: 8 August 2022 / Revised: 3 September 2022 / Accepted: 6 September 2022 / Published: 9 September 2022
(This article belongs to the Special Issue Aquaporins in Brain Disease)

Abstract

:
Aquaporin 4 (AQP4) is a cerebral glial marker that labels ependymal cells and astrocytes’ endfeet and is the main water channel responsible for the parenchymal fluid balance. However, in brain development, AQP4 is a marker of glial stem cells and plays a crucial role in the pathophysiology of pediatric hydrocephalus. Gliogenesis characterization has been hampered by a lack of biomarkers for precursor and intermediate stages and a deeper understanding of hydrocephalus etiology is needed. This manuscript is a focused review of the current research landscape on AQP4 as a possible biomarker for gliogenesis and its influence in pediatric hydrocephalus, emphasizing reactive astrogliosis. The goal is to understand brain development under hydrocephalic and normal physiologic conditions.

1. Introduction

Although 90% of brain tissue is composed of glial cells, most cellular brain studies have focused on neuronal physiology; consequently, the glial cells have been relegated to a supportive position. Astrocytes are the most common glial cells in the CNS, accounting for 20 to 40% of total brain cells [1] and play a fundamental role in neural development, neural circuit function, neurotransmission, blood–brain barrier creation, and neural metabolic support. Astrocytes support the neurovascular systems by connecting neurons and endothelial cells, maintaining brain homeostasis, controlling water, amino acid, and neurotransmitter intake, and monitoring the local activity of synaptic circuits [2,3,4,5].
Since Ramon y Cajal proposed the neuron theory in the early 20th century [6], hundreds of neuron types and functions have been identified [7,8]. However, astrocytes are still considered a homogeneous population, only classified as protoplasmic or fibrous [9]. Astrocytes’ morphological and functional diversity, including their critical role in governing neuronal activity, is well-accepted. Therefore, defining astrogliogenesis to identify astrocytes’ functional and anatomical heterogeneity is crucial to understanding brain physiology [5,10].
Astrogenesis characterization is impeded by a lack of precursor and intermediate stages markers. Furthermore, astrocytes’ plasticity allows proliferative capabilities after being differentiated, complicating their identification [5,10]. Recently, our group has proposed the water channel AQP4 as a possible biomarker of gliogenesis, and its variations were characterized under control and pathological conditions. In-depth systematic reviews have been published, focusing on neuromyelitis optica [11] and gliomas [12,13]. This review focuses on AQP4 expression as a possible biomarker of astrogenesis development and its hypothetical relationship with pathology causing pediatric hydrocephalus.

1.1. AQP4: A Possible Astrogenesis Marker

Even though specific markers do not exist for the different stages of astrogenesis, it has been suggested that astrogenesis progresses trough at least four cellular stages: a radial glial cell (RGC), an intermediate progenitor cell, a maturing postnatal astrocyte, and an adult astrocyte. RGCs are fundamental in early brain development, serving as a scaffold for intermediate progenitors and neuro-glial precursors. After the cessation of neurogenesis, the RGCs become gliogenic (gliogenic switch) and the intermediate progenitors and astrocyte precursors migrate away from the germinal areas to differentiate into astrocytes [5,10,14].
AQP4 is a water channel linked to a glial lineage in the brain since it is expressed in astrocytes (mainly in the endfeet) and the ependymal cells [15,16]. AQP4 is expressed in neural stem cells (NSC) and their glial progeny [17] and modulates the proliferation, survival, migration, and neuronal differentiation of adult NSCs [18,19,20]. Interestingly, in adult lesser hedgehog tenrec (Echinops telfairi), immature radial glial cells persist through adulthood without expressing AQP4 [21]. In zebrafish, another form of astroglial cells continues through adulthood as RGCs expressing AQP4 [22]. In primates, cortical interlaminar astrocytes seem to have an RGC origin since they express typical RGC markers and adult astrocytes markers such as AQP4 [23]. All the animal findings support that AQP4 is associated with an astroglial lineage that may remain undifferentiated as RGCs in adulthood. In utero, AQP4 labels RGCs committed to the astrocyte lineage in humans [24,25] and mice [26]. It has been proposed that unpolarized AQP4-positive cells in the brain show proliferative and regenerative properties as neural stem cells [27]. In mice, the unpolarized expression of AQP4 in RGCs is not detected at the end of in utero life (E16), and the expression of AQP4 is restricted to the astroglial endfeet postnatally (polarized), at P1–3 [26] However, in humans, the AQP4-positive RGCs are present at the beginning of the second trimester of the pregnancy (13–14 postconceptional weeks, PCW), and the polarized expression of AQP4 forming the neurovascular unit is detected at the beginning of the third trimester (25 PCW) [24,25]. Therefore, the maturation of the expression of AQP4 seems to be delayed in mice compared to humans. Our group reported that unpolarized expression of AQP4 is explicitly found in a subpopulation of RGCs that do not progress radially toward the cortical plate but curve to follow white matter tracks, thus serving as a scaffold for glioblasts to develop and populate the white matter tracks, which in turn, provides functionality and maturity to the axons. Thus, these AQP4-positive RGCs were coined as glial stem cells (GSCs) because the projections are not radial and are committed to astrocytes [25].
Interestingly, GSCs follow a temporospatial expression pattern that may indicate the end of neurogenesis and the beginning of gliogenesis [24,25]. GSCs’ first detection is in the glioepithelium of the fimbria of the archicortex and progresses toward fibrous tracts, such as the corpus callosum (CC), the fornix, and the internal capsule in medial areas of the brain. From 25 PCW onward, AQP4 is expressed throughout the neocortex, mainly in the intermediate zone or subplate, giving rise to the cerebral cortex’s white matter. AQP4 is expressed primarily on the main fibrous tracts of the isocortex, possibly to provide homeostasis to facilitate neural impulse transmission to incipient fiber tracts. In general terms, AQP4-positive GSC expression progresses from medial to lateral, starting in the dorsal and ventral hippocampus archicortex, followed by the CC and the ganglionic eminences (transitory structures that contribute to the development of first neurogenic, later gliogenic, and ultimately degenerate), and finally, terminating in the intermediate zone of the resting brain. AQP4-dependent maturity progresses from medial to polar, initially in the perisylvian regions and finally in the occipital and prefrontal zones [24,25,28] (see Figure 1). This correlates with CC maturation. According to classic neuroanatomic studies and recent human embryology neuroimaging, the colossal connections begin centrally in the hippocampal primordium and progress bidirectionally both anteriorly and posteriorly [29,30,31,32,33], with more prominent anterior growth [34,35]. The expression pattern of AQP4-dependent GSCs could define a developmental pathway for cortical neuron functionality, with the archicortex (primitive cortex) acquiring functionality in early gestation, while the frontal and occipital poles acquire functionality at the end of gestation. Therefore, there is an ontogenic logic in which occipital and prefrontal areas achieve functionality and maturity late in brain development since they are responsible for the vision and complex behaviors such as the expression of personality, respectively.
In summary, unpolarized expression of AQP4 is found in proliferative cells with different morphologies: GSCs (with a long projection that is used for other cells to migrate and populate the white matter tracks), and intermediate progenitor cells (without a long projection and oval shape). We hypothesize that the expression of AQP4 in these progenitor cells indicates a gliogenic switch that represents the early stages of astrogenesis. Finally, when AQP4 expression is polarized (astrocytes’ endfeet), this indicates astrocyte maturity as a final step of the astrogenesis. Currently, three specific markers are accepted to identify astrocyte precursors: GLAST, FABP7/BLBP/, and FGFR3 [36,37,38,39]. GLAST is a glutamate transporter active in astrocytes, and its expression initiates with the gliogenic switch indicating specifically astrogenesis precursors. However, FABP7/BLBP/and FGFR3 are also expressed during neurogenic stages, making them unspecific markers [14]. In humans, AQP4 initiates its expression at the gliogenic switch, labeling different astrocyte precursors [24,25]. Thus, this water channel shows a similar expression pattern to GLAST and could be a relevant marker of astrogenesis. Further studies in experimental models should be conducted to confirm this.

1.2. AQP4 in Pediatric Hydrocephalus: Neurodevelopmental Implications

Hydrocephalus is an abnormal build-up of cerebrospinal fluid associated with distension of the ventricular system due to impairments in CSF circulation [40,41]. Inflammatory response plays a fundamental role in acquired conditions such as postinfectious and posthemorrhagic hydrocephalus [42,43,44,45,46]. Reactive astrogliosis (one of the primary expressions of neuroinflammation) is associated with both congenital and acquired hydrocephalus [45,47,48,49,50,51,52,53,54]. Historically, the neurodevelopmental consequences of pediatric hydrocephalus are attributed to parenchymal stretch and periventricular white matter injury secondary to ventricular enlargement and elevated intracranial pressure [55]. Recent evidence supports that the alterations in brain development (neuro-gliogenesis) are responsible for the etiology of some congenital hydrocephalus cases. Thus, genetic disruption of the neuro-gliogenesis in early brain development without CSF dynamic impairment could be a primary pathological factor in patients with congenital hydrocephalus [56]. In addition, GRC and neural progenitor cell loss due to VZ disruption are associated with abnormalities in neurogenesis such as abnormal neuroblast migration as intrinsic mechanisms of congenital [57,58,59] and posthemorrhagic hydrocephalus [60,61]. AQPs have been highly studied in brain development under control [24,28,62] and hydrocephalic conditions [63,64,65,66,67,68,69,70,71]. It has been proposed that the AQP variations are associated with hydrocephalus-compensatory mechanisms to decrease the production and increase the absorption of CSF [66,67,68,69,70,71]. AQPs have been proposed as possible CSF biomarkers for the diagnosis and prognosis of hydrocephalus [63,64,65,66].
Interestingly, AQP4 modulates the neurogenesis associated with neuroinflammation [20] and the glial proliferation in astrocyte cultures [72]. Thus, variations of the expression of AQP4 in the early stages of fetal-onset hydrocephalus may induce neurodevelopmental disorders associated with decreased neurogenesis. Our group reported an early differentiation of AQP4 positive GSCs into reactive astrocytes, implying impaired scaffold functionality in congenital hydrocephalus with spina bifida [25]. The GSCs’ early differentiation into reactive astrocytes could be considered a complementary neurodevelopmental disorder since the GSCs lose their projection, and the glioblasts are no longer migrating toward the white matter tracks. A similar mechanism of premature progenitor differentiation into astrocytes has been recently confirmed in a rat model with spina bifida [73]. This early differentiation into astrocytes could explain the characteristic white matter alterations found in pediatric hydrocephalus [74,75,76]. In spina bifida patients, the CC shows partial agenesis in 65% of the cases and hypoplasia in 35%. In obstructive hydrocephalus due to mesencephalic aqueduct obstruction, 35% of the cases showed partial agenesis and 50% hypoplasia of the CC. Finally, 100% of patients with posthemorrhagic hydrocephalus showed CC hypoplasia [76]. Therefore, this premature differentiation of GSCs into reactive astrocytes may contribute to the neuro-gliogenesis disorders in pediatric hydrocephalus (See Figure 2).
In experiments with neural stem cell cultures from mice that differentiate into ependymal cells, a similar mechanism of premature gliogenesis was proposed under posthemorrhagic conditions [77,78]. In these experiments, neural stem cells committed to becoming ependymal cells differentiated into reactive astrocytes changing the final fate of the cells. These findings support a unified mechanism of neurodevelopmental disorder associated with early astrocyte differentiation. This differentiation in congenital and acquired hydrocephalus conditions may result in primary white matter alterations.

2. Future Work

According to the bibliographic evidence reported in this manuscript, AQP4 is a specific gliogenesis marker that labels glial stem cells associated with white matter tracks. It also affects the degree of glial development as more polarization is associated with maturity (endfeet in neurovascular unit), while no polarization is associated with undifferentiated cells (GSCs). The glial stem cells and neuroblasts seem to show reactivity under hydrocephalic conditions. This reactivity triggers an early differentiation into astrocytes affecting cell migration and normal neural development, possibly explaining primary white matter alterations such as agenesis or hypoplasia of the corpus callosum. Current treatments cannot improve this alteration since it is related to primary neurological development disorders and not to intracranial pressure. Therefore, complementary treatments should not only focus on treating intracranial hypertension but also on preventing cellular neurodevelopmental impairments. Since the inflammatory component plays a fundamental role in reactive astrogliosis, several anti-inflammatory drugs have been proposed to decrease the glial response under hydrocephalic conditions to improve linked cellular alterations. For example, decorin, a proteoglycan that plays a fundamental role in immune regulation and inflammatory diseases [79], reduces white matter-dependent alterations and cytopathology associated with juvenile hydrocephalus [80,81,82].

3. Search Strategy and Selection Criteria

Searches on Pubmed and Google Scholar from 1950 to June 2022 and pertinent papers were used to find references for this review. The searching terms were “hydrocephalus and gliogenesis”, “reactive astrogliosis and white matter and hydrocephalus”, “AQP4 and gliogenesis”, “AQP4 and brain development”, and “AQP4 and neurogenesis”. There were no language restrictions.

4. Limitations

This review did not strictly adhere to PRISMA guidelines [83]. This review focused primarily on AQP4, gliogenesis, and hydrocephalus, other pathologies associated with AQP4 such as neuromyelitis optica or gliomas were not covered.

Author Contributions

L.C.-R. contributed to the conception, design, interpretation, drafting, and editing of the manuscript. I.G.-M. and L.G.H.-A. contributed to manuscript drafting, editing, and creating figures. S.L., A.C.-P. and M.M. contributed to manuscript editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financially supported by the Levecke Family Foundation through a donation to Children’s Hospital of Orange County.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the Levecke Family Foundation for providing the donation necessary to carry out this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Herculano-Houzel, S. The glia/neuron ratio: How it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia 2014, 62, 1377–1391. [Google Scholar] [CrossRef] [PubMed]
  2. Morel, L.; Chiang, M.S.R.; Higashimori, H.; Shoneye, T.; Iyer, L.K.; Yelick, J.; Tai, A.; Yang, Y. Molecular and Functional Properties of Regional Astrocytes in the Adult Brain. J. Neurosci. 2017, 37, 8706–8717. [Google Scholar] [CrossRef] [PubMed]
  3. Chai, H.; Diaz-Castro, B.; Shigetomi, E.; Monte, E.; Octeau, J.C.; Yu, X.; Cohn, W.; Rajendran, P.S.; Vondriska, T.M.; Whitelegge, J.P.; et al. Neural Circuit-Specialized Astrocytes: Transcriptomic, Proteomic, Morphological, and Functional Evidence. Neuron 2017, 95, 531–549.e9. [Google Scholar] [CrossRef] [PubMed]
  4. John Lin, C.-C.; Yu, K.; Hatcher, A.; Huang, T.-W.; Lee, H.K.; Carlson, J.; Weston, M.C.; Chen, F.; Zhang, Y.; Zhu, W.; et al. Identification of diverse astrocyte populations and their malignant analogs. Nat. Neurosci. 2017, 20, 396–405. [Google Scholar] [CrossRef] [PubMed]
  5. Akdemir, E.S.; Huang, A.Y.-S.; Deneen, B. Astrocytogenesis: Where, when, and how. F1000Research 2020, 9, 233. [Google Scholar] [CrossRef]
  6. López-Muñoz, F.; Boya, J.; Alamo, C. Neuron theory, the cornerstone of neuroscience, on the centenary of the Nobel Prize award to Santiago Ramón y Cajal. Brain Res. Bull. 2006, 70, 391–405. [Google Scholar] [CrossRef]
  7. Zeisel, A.; Hochgerner, H.; Lönnerberg, P.; Johnsson, A.; Memic, F.; van der Zwan, J.; Häring, M.; Braun, E.; Borm, L.E.; La Manno, G.; et al. Molecular Architecture of the Mouse Nervous System. Cell 2018, 174, 999–1014.e22. [Google Scholar] [CrossRef] [PubMed]
  8. Saunders, A.; Macosko, E.Z.; Wysoker, A.; Goldman, M.; Krienen, F.M.; de Rivera, H.; Bien, E.; Baum, M.; Bortolin, L.; Wang, S.; et al. Molecular Diversity and Specializations among the Cells of the Adult Mouse Brain. Cell 2018, 174, 1015–1030.e16. [Google Scholar] [CrossRef] [PubMed]
  9. Batiuk, M.Y.; Martirosyan, A.; Wahis, J.; de Vin, F.; Marneffe, C.; Kusserow, C.; Koeppen, J.; Viana, J.F.; Oliveira, J.F.; Voet, T.; et al. Identification of region-specific astrocyte subtypes at single cell resolution. Nat. Commun. 2020, 11, 1220. [Google Scholar] [CrossRef]
  10. Molofsky, A.V.; Krenick, R.; Ullian, E.; Tsai, H.-H.; Deneen, B.; Richardson, W.D.; Barres, B.A.; Rowitch, D.H. Astrocytes and disease: A neurodevelopmental perspective. Genes Dev. 2012, 26, 891–907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Jarius, S.; Paul, F.; Weinshenker, B.G.; Levy, M.; Kim, H.J.; Wildemann, B. Neuromyelitis Optica. Nat. Rev. Dis. Primers 2020, 6, 85. [Google Scholar] [CrossRef] [PubMed]
  12. Ding, T.; Gu, F.; Fu, L.; Ma, Y.-J. Aquaporin-4 in glioma invasion and an analysis of molecular mechanisms. J. Clin. Neurosci. 2010, 17, 1359–1361. [Google Scholar] [CrossRef] [PubMed]
  13. Lan, Y.-L.; Wang, X.; Lou, J.-C.; Ma, X.-C.; Zhang, B. The potential roles of aquaporin 4 in malignant gliomas. Oncotarget 2017, 8, 32345–32355. [Google Scholar] [CrossRef]
  14. Molofsky, A.V.; Deneen, B. Astrocyte development: A Guide for the Perplexed. Glia 2015, 63, 1320–1329. [Google Scholar] [CrossRef] [PubMed]
  15. Nielsen, S.; King, L.S.; Christensen, B.M.; Agre, P. Aquaporins in complex tissues. II. Subcellular distribution in respiratory and glandular tissues of rat. Am. J. Physiol. 1997, 273, C1549–C1561. [Google Scholar] [CrossRef]
  16. Rash, J.E.; Yasumura, T.; Hudson, C.S.; Agre, P.; Nielsen, S. Direct immunogold labeling of aquaporin-4 in square arrays of astrocyte and ependymocyte plasma membranes in rat brain and spinal cord. Proc. Natl. Acad. Sci. USA 1998, 95, 11981–11986. [Google Scholar] [CrossRef]
  17. Cavazzin, C.; Ferrari, D.; Facchetti, F.; Russignan, A.; Vescovi, A.L.; La Porta, C.A.; Gritti, A. Unique expression and localization of aquaporin- 4 and aquaporin-9 in murine and human neural stem cells and in their glial progeny. Glia 2006, 53, 167–181. [Google Scholar] [CrossRef]
  18. Kong, H.; Fan, Y.; Xie, J.; Ding, J.; Sha, L.; Shi, X.; Sun, X.; Hu, G. AQP4 knockout impairs proliferation, migration and neuronal differentiation of adult neural stem cells. J. Cell Sci. 2008, 121, 4029–4036. [Google Scholar] [CrossRef]
  19. La Porta, C.A.; Gena, P.; Gritti, A.; Fascio, U.; Svelto, M.; Calamita, G. Adult murine CNS stem cells express aquaporin channels. Biol. Cell 2006, 98, 89–94. [Google Scholar] [CrossRef]
  20. Liang, R.; Yong, S.; Huang, X.; Kong, H.; Hu, G.; Fan, Y. Aquaporin-4 Mediates the Suppressive Effect of Lipopolysaccharide on Hippocampal Neurogenesis. Neuroimmunomodulation 2016, 23, 309–317. [Google Scholar] [CrossRef]
  21. Mack, A.F.; Künzle, H.; Lange, M.; Mages, B.; Reichenbach, A.; Härtig, W. Radial glial elements in the cerebral cortex of the lesser hedgehog tenrec. Brain Struct. Funct. 2018, 223, 3909–3917. [Google Scholar] [CrossRef] [PubMed]
  22. Gleiser, C.; Wagner, A.; Fallier-Becker, P.; Wolburg, H.; Hirt, B.; Mack, A.F. Aquaporin-4 in Astroglial Cells in the CNS and Supporting Cells of Sensory Organs—A Comparative Perspective. Int. J. Mol. Sci. 2016, 17, 1411. [Google Scholar] [CrossRef] [PubMed]
  23. Falcone, C.; Penna, E.; Hong, T.; Tarantal, A.F.; Hof, P.R.; Hopkins, W.D.; Sherwood, C.C.; Noctor, S.C.; Martínez-Cerdeño, V. Cortical Interlaminar Astrocytes Are Generated Prenatally, Mature Postnatally, and Express Unique Markers in Human and Nonhuman Primates. Cereb. Cortex 2021, 31, 379–395. [Google Scholar] [CrossRef] [PubMed]
  24. Holst, C.B.; Brøchner, C.B.; Vitting-Seerup, K.; Møllgård, K. Astrogliogenesis in human fetal brain: Complex spatiotemporal immunoreactivity patterns of GFAP, S100, AQP 4 and YKL -40. J. Anat. 2019, 235, 590–615. [Google Scholar] [CrossRef]
  25. Castañeyra-Ruiz, L.; González-Marrero, I.; Hernández-Abad, L.G.; Carmona-Calero, E.M.; Pardo, M.R.; Baz-Davila, R.; Lee, S.; Muhonen, M.; Borges, R.; Castañeyra-Perdomo, A. AQP4 labels a subpopulation of white matter-dependent glial radial cells affected by pediatric hydrocephalus, and its expression increased in glial microvesicles released to the cerebrospinal fluid in obstructive hydrocephalus. Acta Neuropathol. Commun. 2022, 10, 41. [Google Scholar] [CrossRef]
  26. Fallier-Becker, P.; Vollmer, J.P.; Bauer, H.; Noell, S.; Wolburg, H.; Mack, A.F. Onset of aquaporin-4 expression in the developing mouse brain. Int. J. Dev. Neurosci. 2014, 36, 81–89. [Google Scholar] [CrossRef]
  27. Mack, A.F.; Wolburg, H. A Novel Look at Astrocytes: Aquaporins, ionic homeostasis, and the role of the microenvironment for regeneration in the CNS. Neuroscientist 2013, 19, 195–207. [Google Scholar] [CrossRef]
  28. Gömöri, E.; Pál, J.; Ábrahám, H.; Vajda, Z.; Sulyok, E.; Seress, L.; Dóczi, T. Fetal development of membrane water channel proteins aquaporin-1 and aquaporin-4 in the human brain. Int. J. Dev. Neurosci. 2006, 24, 295–305. [Google Scholar] [CrossRef]
  29. Deoni, S.C.L.; Mercure, E.; Blasi, A.; Gasston, D.; Thomson, A.; Johnson, M.; Williams, S.; Murphy, D. Mapping Infant Brain Myelination with Magnetic Resonance Imaging. J. Neurosci. 2011, 31, 784–791. [Google Scholar] [CrossRef]
  30. Kier, E.L.; Truwit, C.L. The normal and abnormal genu of the corpus callosum: An evolutionary, embryologic, anatomic, and MR analysis. AJNR Am. J. Neuroradiol. 1996, 17, 1631–1641. [Google Scholar]
  31. Huang, H.; Xue, R.; Zhang, J.; Ren, T.; Richards, L.J.; Yarowsky, P.; Miller, M.I.; Mori, S. Anatomical Characterization of Human Fetal Brain Development with Diffusion Tensor Magnetic Resonance Imaging. J. Neurosci. 2009, 29, 4263–4273. [Google Scholar] [CrossRef] [PubMed]
  32. Barkovich, A.J.; Lyon, G.; Evrard, P. Formation, maturation, and disorders of white matter. AJNR Am. J. Neuroradiol. 1992, 13, 447–461. [Google Scholar] [PubMed]
  33. Paul, L.K. Developmental malformation of the corpus callosum: A review of typical callosal development and examples of developmental disorders with callosal involvement. J. Neurodev. Disord. 2011, 3, 3–27. [Google Scholar] [CrossRef]
  34. Rakic, P.; Yakovlev, P.I. Development of the corpus callosum and cavum septi in man. J. Comp. Neurol. 1968, 132, 45–72. [Google Scholar] [CrossRef] [PubMed]
  35. Luders, E.; Thompson, P.; Toga, A.W. The Development of the Corpus Callosum in the Healthy Human Brain. J. Neurosci. 2010, 30, 10985–10990. [Google Scholar] [CrossRef]
  36. Anthony, T.E.; Klein, C.; Fishell, G.; Heintz, N. Radial Glia Serve as Neuronal Progenitors in All Regions of the Central Nervous System. Neuron 2004, 41, 881–890. [Google Scholar] [CrossRef]
  37. Owada, Y.; Yoshimoto, T.; Kondo, H. Spatio-temporally differential expression of genes for three members of fatty acid binding proteins in developing and mature rat brains. J. Chem. Neuroanat. 1996, 12, 113–122. [Google Scholar] [CrossRef]
  38. Pringle, N.P.; Yu, W.-P.; Howell, M.; Colvin, J.S.; Ornitz, D.M.; Richardson, W.D. Fgfr3 expression by astrocytes and their precursors: Evidence that astrocytes and oligodendrocytes originate in distinct neuroepithelial domains. Development 2003, 130, 93–102. [Google Scholar] [CrossRef]
  39. Shibata, T.; Yamada, K.; Watanabe, M.; Ikenaka, K.; Wada, K.; Tanaka, K.; Inoue, Y. Glutamate Transporter GLAST Is Expressed in the Radial Glia–Astrocyte Lineage of Developing Mouse Spinal Cord. J. Neurosci. 1997, 17, 9212–9219. [Google Scholar] [CrossRef]
  40. Rekate, H.L. A Contemporary Definition and Classification of Hydrocephalus. Semin. Pediatr. Neurol. 2009, 16, 9–15. [Google Scholar] [CrossRef]
  41. Kahle, K.T.; Kulkarni, A.V.; Limbrick, D.D., Jr.; Warf, B.C. Hydrocephalus in children. Lancet 2016, 387, 788–799. [Google Scholar] [CrossRef]
  42. Karimy, J.K.; Zhang, J.; Kurland, D.B.; Theriault, B.C.; Duran, D.; Stokum, J.A.; Furey, C.G.; Zhou, X.; Mansuri, M.S.; Montejo, J.; et al. Inflammation-dependent cerebrospinal fluid hypersecretion by the choroid plexus epithelium in posthemorrhagic hydrocephalus. Nat. Med. 2017, 23, 997–1003. [Google Scholar] [CrossRef]
  43. Karimy, J.K.; Reeves, B.C.; Damisah, E.; Duy, P.Q.; Antwi, P.; David, W.; Wang, K.; Schiff, S.J.; Limbrick, D.D., Jr.; Alper, S.L.; et al. Inflammation in acquired hydrocephalus: Pathogenic mechanisms and therapeutic targets. Nat. Rev. Neurol. 2020, 16, 285–296. [Google Scholar] [CrossRef]
  44. Harris, C.A.; Morales, D.M.; Arshad, R.; McAllister, J.P., II; Limbrick, D.D., Jr. Cerebrospinal fluid biomarkers of neuroinflammation in children with hydrocephalus and shunt malfunction. Fluids Barriers CNS 2021, 18, 4. [Google Scholar] [CrossRef]
  45. Garcia-Bonilla, M.; Castaneyra-Ruiz, L.; Zwick, S.; Talcott, M.; Otun, A.; Isaacs, A.M.; Morales, D.M.; Limbrick, D.D.; McAllister, J.P. Acquired hydrocephalus is associated with neuroinflammation, progenitor loss, and cellular changes in the subventricular zone and periventricular white matter. Fluids Barriers CNS 2022, 19, 17. [Google Scholar] [CrossRef]
  46. Isaacs, A.M.; Neil, J.J.; McAllister, J.P.; Dahiya, S.; Castaneyra-Ruiz, L.; Merisaari, H.; Botteron, H.E.; Alexopoulous, D.; George, A.; Sun, P.; et al. Microstructural Periventricular White Matter Injury in Post-hemorrhagic Ventricular Dilatation. Neurology 2021, 98, e364–e375. [Google Scholar] [CrossRef]
  47. Ding, Y.; Zhang, T.; Wu, G.; McBride, D.W.; Xu, N.; Klebe, D.W.; Zhang, Y.; Li, Q.; Tang, J.; Zhang, J.H. Astrogliosis inhibition attenuates hydrocephalus by increasing cerebrospinal fluid reabsorption through the glymphatic system after germinal matrix hemorrhage. Exp. Neurol. 2019, 320, 113003. [Google Scholar] [CrossRef]
  48. Eide, P.K.; Hansson, H.-A. Astrogliosis and impaired aquaporin-4 and dystrophin systems in idiopathic normal pressure hydrocephalus. Neuropathol. Appl. Neurobiol. 2018, 44, 474–490. [Google Scholar] [CrossRef]
  49. Garcia, C.A.B.; Catalão, C.H.R.; Machado, H.; Júnior, I.M.; Romeiro, T.H.; Peixoto-Santos, J.E.; Santos, M.V.; Lopes, L.D.S. Edaravone reduces astrogliosis and apoptosis in young rats with kaolin-induced hydrocephalus. Child’s Nerv. Syst. 2016, 33, 419–428. [Google Scholar] [CrossRef]
  50. Miller, J.M.; McAllister, J.P., II. Reduction of astrogliosis and microgliosis by cerebrospinal fluid shunting in experimental hydrocephalus. Cereb. Fluid Res. 2007, 4, 10–186. [Google Scholar] [CrossRef]
  51. Suryaningtyas, W.; Parenrengi, M.A.; Bajamal, A.H.; Rantam, F.A. Lipid Peroxidation Induces Reactive Astrogliosis by Activating WNT/β-Catenin Pathway in Hydrocephalus. Malays. J. Med. Sci. 2020, 27, 34–42. [Google Scholar] [CrossRef] [PubMed]
  52. Tida, J.A.; Catalão, C.H.R.; Garcia, C.A.B.; dos Santos, A.C.; Salmon, C.E.G.; Lopes, L.D.S. Acupuncture at ST36 Exerts Neuroprotective Effects via Inhibition of Reactive Astrogliosis in Infantile Rats with Hydrocephalus. Acupunct. Med. 2018, 36, 386–393. [Google Scholar] [CrossRef] [PubMed]
  53. Deren, K.E.; Packer, M.; Forsyth, J.; Milash, B.; Abdullah, O.M.; Hsu, E.W.; McAllister, J.P. Reactive astrocytosis, microgliosis and inflammation in rats with neonatal hydrocephalus. Exp. Neurol. 2010, 226, 110–119. [Google Scholar] [CrossRef] [PubMed]
  54. Roales-Buján, R.; Páez, P.; Guerra, M.; Rodríguez, S.; Vío, K.; Ho-Plagaro, A.; García-Bonilla, M.; Rodríguez-Pérez, L.-M.; Domínguez-Pinos, M.-D.; Rodríguez, E.-M.; et al. Astrocytes acquire morphological and functional characteristics of ependymal cells following disruption of ependyma in hydrocephalus. Acta Neuropathol. 2012, 124, 531–546. [Google Scholar] [CrossRef] [PubMed]
  55. Del Bigio, M.R. Neuropathology and structural changes in hydrocephalus. Dev. Disabil. Res. Rev. 2010, 16, 16–22. [Google Scholar] [CrossRef]
  56. Jin, S.C.; Dong, W.; Kundishora, A.J.; Panchagnula, S.; Moreno-De-Luca, A.; Furey, C.G.; Allocco, A.A.; Walker, R.L.; Nelson-Williams, C.; Smith, H.; et al. Exome sequencing implicates genetic disruption of prenatal neuro-gliogenesis in sporadic congenital hydrocephalus. Nat. Med. 2020, 26, 1754–1765. [Google Scholar] [CrossRef]
  57. Guerra, M.M.; Henzi, R.; Ortloff, A.; Lichtin, N.; Vío, K.; Jiménez, A.J.; Dominguez-Pinos, M.D.; González, C.; Jara, M.C.; Hinostroza, F.; et al. Cell Junction Pathology of Neural Stem Cells Is Associated With Ventricular Zone Disruption, Hydrocephalus, and Abnormal Neurogenesis. J. Neuropathol. Exp. Neurol. 2015, 74, 653–671. [Google Scholar] [CrossRef]
  58. Rodríguez, E.M.; Guerra, M.M.; Vío, K.; González, C.; Ortloff, A.; Bátiz, L.F.; Rodríguez, S.; Jara, M.C.; Muñoz, R.I.; Ortega, E.; et al. A cell junction pathology of neural stem cells leads to abnormal neurogenesis and hydrocephalus. Biol. Res. 2012, 45, 231–241. [Google Scholar] [CrossRef]
  59. Domínguez-Pinos, M.D.; Páez, P.; Jiménez, A.-J.; Weil, B.; Arráez, M.-A.; Pérez-Fígares, J.-M.; Rodríguez, E.-M. Ependymal Denudation and Alterations of the Subventricular Zone Occur in Human Fetuses With a Moderate Communicating Hydrocephalus. J. Neuropathol. Exp. Neurol. 2005, 64, 595–604. [Google Scholar] [CrossRef]
  60. McAllister, J.P.; Guerra, M.M.; Ruiz, L.C.; Jimenez, A.J.; Dominguez-Pinos, D.; Sival, D.; Dunnen, W.D.; Morales, D.; Schmidt, R.E.; Rodríguez, E.; et al. Ventricular Zone Disruption in Human Neonates with Intraventricular Hemorrhage. J. Neuropathol. Exp. Neurol. 2017, 76, 358–375. [Google Scholar] [CrossRef]
  61. Fernández-Muñoz, B.; Rosell-Valle, C.; Ferrari, D.; Alba-Amador, J.; Montiel, M.; Campos-Cuerva, R.; Lopez-Navas, L.; Muñoz-Escalona, M.; Martín-López, M.; Profico, D.C.; et al. Retrieval of germinal zone neural stem cells from the cerebrospinal fluid of premature infants with intraventricular hemorrhage. Stem Cells Transl. Med. 2020, 9, 1085–1101. [Google Scholar] [CrossRef] [PubMed]
  62. Castañeyra-Ruiz, L.; González-Marrero, I.; Hernández-Abad, L.G.; Carmona-Calero, E.M.; Meyer, G.; Castañeyra-Perdomo, A. A Distal to Proximal Gradient of Human Choroid Plexus Development, with Antagonistic Expression of Glut1 and AQP1 in Mature Cells vs. Calbindin and PCNA in Proliferative Cells. Front. Neuroanat. 2016, 10, 87. [Google Scholar] [CrossRef] [PubMed]
  63. Castañeyra-Ruiz, L.; González-Marrero, I.; González-Toledo, J.M.; Castañeyra-Ruiz, A.; de Paz-Carmona, H.; Castañeyra-Perdomo, A.; Carmona-Calero, E.M. Aquaporin-4 expression in the cerebrospinal fluid in congenital human hydrocephalus. Fluids Barriers CNS 2013, 10, 18. [Google Scholar] [CrossRef]
  64. Limbrick, D.D., Jr.; Castaneyra-Ruiz, L.; Han, R.H.; Berger, D.; McAllister, J.P.; Morales, D.M. Cerebrospinal Fluid Biomarkers of Pediatric Hydrocephalus. Pediatr. Neurosurg. 2017, 52, 426–435. [Google Scholar] [CrossRef] [PubMed]
  65. Castañeyra-Ruiz, L.; González-Marrero, I.; Carmona-Calero, E.M.; Abreu-Gonzalez, P.; Lecuona, M.; Brage, L.; Rodríguez, E.M.; Castañeyra-Perdomo, A. Cerebrospinal fluid levels of tumor necrosis factor alpha and aquaporin 1 in patients with mild cognitive impairment and idiopathic normal pressure hydrocephalus. Clin. Neurol. Neurosurg. 2016, 146, 76–81. [Google Scholar] [CrossRef] [PubMed]
  66. Castañeyra-Ruiz, L.; Hernández-Abad, L.G.; Carmona-Calero, E.M.; Castañeyra-Perdomo, A.; González-Marrero, I. AQP1 Overexpression in the CSF of Obstructive Hydrocephalus and Inversion of Its Polarity in the Choroid Plexus of a Chiari Malformation Type II Case. J. Neuropathol. Exp. Neurol. 2019, 78, 641–647. [Google Scholar] [CrossRef]
  67. Jeon, T.; Park, K.-S.; Park, S.-H.; Hwang, J.-H.; Hwang, S.K. Expression of Aquaporin 1 and 4 in the Choroid Plexus and Brain Parenchyma of Kaolin-Induced Hydrocephalic Rats. Korean J. Neurotrauma 2017, 13, 68–75. [Google Scholar] [CrossRef]
  68. Kim, J.; Jung, Y. Increased aquaporin-1 and Na+-K+-2Cl- cotransporter 1 expression in choroid plexus leads to blood-cerebrospinal fluid barrier disruption and necrosis of hippocampal CA1 cells in acute rat models of hyponatremia. J. Neurosci. Res. 2012, 90, 1437–1444. [Google Scholar] [CrossRef]
  69. Paul, L.; Madan, M.; Rammling, M.; Chigurupati, S.; Chan, S.L.; Pattisapu, J.V. Expression of Aquaporin 1 and 4 in a Congenital Hydrocephalus Rat Model. Neurosurgery 2011, 68, 462–473. [Google Scholar] [CrossRef]
  70. Carmona-Calero, E.M.; Gonzalez-Toledo, J.M.; Castañeyra-Ruiz, L.; González-Marrero, I.; Castañeyra-Ruiz, M.; Rancel-Torres, N.; Castañeyra-Ruiz, A.; Castañeyra-Perdomo, A. Expression of aquaporin-1 in the choroid plexus in communicating and non-communicating hydrocephalic rats. Eur. J. Anat. 2015, 19, 125–129. [Google Scholar]
  71. Castañeyra-Ruiz, L.; González-Marrero, I.; Castañeyra-Ruiz, M.; González-Toledo, J.M.; Carmona-Calero, E.M. Los canales de agua. Acuaporinas 1 y 4 en el sistema nervioso central y su relación con la hidrocefalia. Majorensis 2014, 10, 1–6. [Google Scholar]
  72. Küppers, E.; Gleiser, C.; Brito, V.; Wachter, B.; Pauly, T.; Hirt, B.; Grissmer, S. AQP4 expression in striatal primary cultures is regulated by dopamine—Implications for proliferation of astrocytes. Eur. J. Neurosci. 2008, 28, 2173–2182. [Google Scholar] [CrossRef] [PubMed]
  73. Oria, M.; Pathak, B.; Li, Z.; Bakri, K.; Gouwens, K.; Varela, M.F.; Lampe, K.; Murphy, K.P.; Lin, C.-Y.; Peiro, J.L. Premature Neural Progenitor Cell Differentiation Into Astrocytes in Retinoic Acid-Induced Spina Bifida Rat Model. Front. Mol. Neurosci. 2022, 15, 888351. [Google Scholar] [CrossRef] [PubMed]
  74. Tully, H.M.; Dobyns, W.B. Infantile hydrocephalus: A review of epidemiology, classification and causes. Eur. J. Med. Genet. 2014, 57, 359–368. [Google Scholar] [CrossRef]
  75. Demyanenko, G.P.; Tsai, A.Y.; Maness, P.F. Abnormalities in Neuronal Process Extension, Hippocampal Development, and the Ventricular System of L1 Knockout Mice. J. Neurosci. 1999, 19, 4907–4920. [Google Scholar] [CrossRef]
  76. Hannay, H.J. Functioning of the corpus callosum in children with early hydrocephalus. J. Int. Neuropsychol. Soc. 2000, 6, 351–361. [Google Scholar] [CrossRef]
  77. Castaneyra-Ruiz, L.; Morales, M.D.M.; McAllister, J.P.; Brody, S.L.; Isaacs, A.M.; Strahle, J.M.; Dahiya, S.M.; Limbrick, D.D. Blood Exposure Causes Ventricular Zone Disruption and Glial Activation In Vitro. J. Neuropathol. Exp. Neurol. 2018, 77, 803–813. [Google Scholar] [CrossRef]
  78. Castaneyra-Ruiz, L.; McAllister, J.P., II; Morales, D.M.; Brody, S.L.; Isaacs, A.M.; Limbrick, D.D. Preterm intraventricular hemorrhage in vitro: Modeling the cytopathology of the ventricular zone. Fluids Barriers CNS 2020, 17, 46. [Google Scholar] [CrossRef]
  79. Dong, Y.; Zhong, J.; Dong, L. The Role of Decorin in Autoimmune and Inflammatory Diseases. J. Immunol. Res. 2022, 2022, 1283383. [Google Scholar] [CrossRef]
  80. Botfield, H.; Gonzalez, A.M.; Abdullah, O.; Skjolding, A.D.; Berry, M.; McAllister, J.P.; Logan, A. Decorin prevents the development of juvenile communicating hydrocephalus. Brain 2013, 136, 2842–2858. [Google Scholar] [CrossRef]
  81. Aojula, A.; Botfield, H.; McAllister, J.P., II; Gonzalez, A.M.; Abdullah, O.; Logan, A.; Sinclair, A. Diffusion tensor imaging with direct cytopathological validation: Characterisation of decorin treatment in experimental juvenile communicating hydrocephalus. Fluids Barriers CNS 2016, 13, 9. [Google Scholar] [CrossRef] [PubMed]
  82. McAllister, J.P.; Aojula, A.; Botfield, H.; Abdullah, O.; Gonzalez, A.M.; Ragan, D.; Logan, A.; Sinclair, A. Decorin reduces white matter pathology in experimental hydrocephalus: A diffusion tensor imaging and immunohistochemical study. Fluids Barriers CNS 2015, 12, O59. [Google Scholar] [CrossRef]
  83. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Syst. Rev. 2021, 74, 790–799. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the expression of AQP4 in brain development. In the telencephalon, AQP4 expression starts in GSCs and GBs and progresses from the para hippocampal VZ toward the glioepithelium of the fimbria of the dorsal hippocampus at 12–13 PCW. At 21 PCW, AQP4 has advanced medial to lateral in the coronal view, and medial to polar in the sagittal view. At this point, AQP4 is expressed in the ventricular zone adjacent to the medial portion of the CC, where the GSCs project their cellular process toward CC fibers. GSCs are also found in the LGE and the lenticular nucleus projecting toward the internal capsule. At 25 PCW, the AQP4 is patent cortically, and GSC processes are found projecting from the SVZ to the IZ. Polarized AQP4 expression is located in the astrocyte’s endfeet as a part of the neurovascular unit at the IZ, indicating maturity and functionality. LV, lateral ventricle; MZ, marginal zone; CP, cortical plate; SCP, subcortical plate; IZ, intermediate zone; SVZ, subventricular zone; VZ, ventricular zone; CC, corpus callosum; IC, internal capsule; DHip, dorsal hippocampus; Tha, thalamus; GE ganglionic eminence; LGE, lateral ganglionic eminence; MGE, medial ganglionic eminence; Ca, caudate; LN, lenticular nucleus; GP, glial progenitor; A, astrocyte; BV, blood vessel; AQP4, aquaporin 4.
Figure 1. Schematic representation of the expression of AQP4 in brain development. In the telencephalon, AQP4 expression starts in GSCs and GBs and progresses from the para hippocampal VZ toward the glioepithelium of the fimbria of the dorsal hippocampus at 12–13 PCW. At 21 PCW, AQP4 has advanced medial to lateral in the coronal view, and medial to polar in the sagittal view. At this point, AQP4 is expressed in the ventricular zone adjacent to the medial portion of the CC, where the GSCs project their cellular process toward CC fibers. GSCs are also found in the LGE and the lenticular nucleus projecting toward the internal capsule. At 25 PCW, the AQP4 is patent cortically, and GSC processes are found projecting from the SVZ to the IZ. Polarized AQP4 expression is located in the astrocyte’s endfeet as a part of the neurovascular unit at the IZ, indicating maturity and functionality. LV, lateral ventricle; MZ, marginal zone; CP, cortical plate; SCP, subcortical plate; IZ, intermediate zone; SVZ, subventricular zone; VZ, ventricular zone; CC, corpus callosum; IC, internal capsule; DHip, dorsal hippocampus; Tha, thalamus; GE ganglionic eminence; LGE, lateral ganglionic eminence; MGE, medial ganglionic eminence; Ca, caudate; LN, lenticular nucleus; GP, glial progenitor; A, astrocyte; BV, blood vessel; AQP4, aquaporin 4.
Ijms 23 10438 g001
Figure 2. Schematic representation of astrocyte’s premature differentiation in hydrocephalus. In non-hydrocephalic conditions, after mid-gestation at the periventricular white matter areas, the ventricular zone is mainly composed of ECs, RGCs, GSCs, GPs, and NPs (1). When hydrocephalus pathology develops, a ventricular lining disruption occurs, associated with reactive astrogliosis (2). This representation proposes that white matter-associated GSCs and GPs become activated under hydrocephalic conditions suffering a premature differentiation into astrocytes (a,b). This early GSC differentiation into astrocytes impairs its normal function as a scaffold to guide cells into the white matter, implying a lack of cell migration resulting in the characteristic hypoplasia or dysgenesis of the white matter tracks. In turn, the incapability of NPs to migrate triggers periventricular heterotopias. Finally, a premature differentiation of GPs into astrocytes as a reaction to an inflammatory response may alter the final fate of the cells and affect their migration (3). LV, lateral ventricle; MZ, marginal zone; CP, cortical plate; SCP, subcortical plate; IZ, intermediate zone; SVZ, subventricular zone; VZ, ventricular zone; VZD, ventricular zone disruption; CC, corpus callosum; IC, internal capsule; GSC, glial stem cell; RGSC, reactive glial stem cell; GP, glial progenitor; RGP, reactive glial progenitor; A, astrocyte; RGC, radial glial cell; NP, neural progenitor; PVH, periventricular heterotopia.
Figure 2. Schematic representation of astrocyte’s premature differentiation in hydrocephalus. In non-hydrocephalic conditions, after mid-gestation at the periventricular white matter areas, the ventricular zone is mainly composed of ECs, RGCs, GSCs, GPs, and NPs (1). When hydrocephalus pathology develops, a ventricular lining disruption occurs, associated with reactive astrogliosis (2). This representation proposes that white matter-associated GSCs and GPs become activated under hydrocephalic conditions suffering a premature differentiation into astrocytes (a,b). This early GSC differentiation into astrocytes impairs its normal function as a scaffold to guide cells into the white matter, implying a lack of cell migration resulting in the characteristic hypoplasia or dysgenesis of the white matter tracks. In turn, the incapability of NPs to migrate triggers periventricular heterotopias. Finally, a premature differentiation of GPs into astrocytes as a reaction to an inflammatory response may alter the final fate of the cells and affect their migration (3). LV, lateral ventricle; MZ, marginal zone; CP, cortical plate; SCP, subcortical plate; IZ, intermediate zone; SVZ, subventricular zone; VZ, ventricular zone; VZD, ventricular zone disruption; CC, corpus callosum; IC, internal capsule; GSC, glial stem cell; RGSC, reactive glial stem cell; GP, glial progenitor; RGP, reactive glial progenitor; A, astrocyte; RGC, radial glial cell; NP, neural progenitor; PVH, periventricular heterotopia.
Ijms 23 10438 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Castañeyra-Ruiz, L.; González-Marrero, I.; Hernández-Abad, L.G.; Lee, S.; Castañeyra-Perdomo, A.; Muhonen, M. AQP4, Astrogenesis, and Hydrocephalus: A New Neurological Perspective. Int. J. Mol. Sci. 2022, 23, 10438. https://doi.org/10.3390/ijms231810438

AMA Style

Castañeyra-Ruiz L, González-Marrero I, Hernández-Abad LG, Lee S, Castañeyra-Perdomo A, Muhonen M. AQP4, Astrogenesis, and Hydrocephalus: A New Neurological Perspective. International Journal of Molecular Sciences. 2022; 23(18):10438. https://doi.org/10.3390/ijms231810438

Chicago/Turabian Style

Castañeyra-Ruiz, Leandro, Ibrahim González-Marrero, Luis G. Hernández-Abad, Seunghyun Lee, Agustín Castañeyra-Perdomo, and Michael Muhonen. 2022. "AQP4, Astrogenesis, and Hydrocephalus: A New Neurological Perspective" International Journal of Molecular Sciences 23, no. 18: 10438. https://doi.org/10.3390/ijms231810438

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

Article Metrics

Back to TopTop