Skip to main content

EDITORIAL article

Front. Neuroanat., 07 July 2023
Volume 17 - 2023 | https://doi.org/10.3389/fnana.2023.1242684

Editorial: Molecular and cellular logic of cerebral cortex development, evolution, and disease

  • 1Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
  • 2Department of Cellular, Computational and Integrated Biology, University of Trento, Trento, Italy
  • 3Department of Biology, University of Pisa, Pisa, Italy

Among central nervous system structures, the cerebral cortex is widely recognized as the hub of higher cognitive functions that distinctly characterize humans. With its intricate network of connections, bewildering variety of cell types, and peculiar multilaminated structure, the cerebral cortex has undergone a dramatic evolution over time accompanied primarily by an increase in proportions in relation to body size. Indeed, the gyrification process is a result of the expansion of novel classes of progenitor cells, especially the outer radial glia, that produce and provide migration guidance for upper-layer cortical neurons, many of which are characteristic of the most evolved species and define the neocortex, underlining its recent phylogenetic origin.

This Research Topic aims to present a comprehensive overview of the latest advancements in cerebral cortex development. Special attention is given to the role of the molecular mechanisms that coordinate its assembly, the involvement of non-neuronal cells in its development and in the acquisition of cognitive abilities, and the evolutionary factors influencing its cytoarchitecture.

In humans, the neural tube closes ~30 days post-conception, and the neocortex forms at the rostral end of the neural tube, through the migration of neurons originating from proliferative regions near cerebral ventricles of the telencephalon (Sidman and Rakic, 1973; Marin and Rubenstein, 2003). Migration ensures that layers generate in an inside-out fashion. Therefore, layer 1 is the most external and the first to be generated, followed by deep infragranular layer 5 and 6 neurons, then granular layer 4 neurons, and, eventually, layer 2 and 3 neurons (Cadwell et al., 2019). The ultimate identity of a cortical neuron and its definitive allocation are attained through the coordinated activation of crucial transcription factors (Kast and Levitt, 2019). However, the transcriptional profile per se may not be sufficiently informative, as translation into proteins may be delayed, depending on mRNA stability, localization, and editing (Zahr et al., 2018; Park et al., 2022). Cremisi and Vignali focus on the post-transcriptional control attributed to RNA-binding proteins (RBPs) and microRNAs during corticogenesis. Both RBPs and microRNAs operate as translational repressors exerting control over various processes, including neural proliferation and differentiation (Franzoni et al., 2015), layering (Shu et al., 2017), and plasticity (Letellier et al., 2014). The authors summarize how microRNAs (i.e., mir-3607, mir- 122, and mir-137) may exert a heterochronic effect in cortical neuron maturation by refining the translated protein's temporal appearance. This suggests that microRNAs also have a role in the evolution of the mammalian brain, facilitating increases in neural progenitor cells or regulating differentiation and migration (Tomasello et al., 2022), two processes that eventually sustain the enlargement of cortical areas that anatomically differentiate gyrencephalic species.

The question of whether proliferative niches persist in the adult brain has been extensively studied, with the detection of mitotic events in the adult hippocampus of rodents (Alvarez-Buylla and Lim, 2004). This area originates from the medial pallium, which has been thoroughly investigated in mammals and in anamniotes due to its involvement in learning and navigation (Salas et al., 2006; Sotelo et al., 2016). Amphibians, the only extant anamniote tetrapods, express conserved transcription factors deemed necessary for mammalian hippocampal development (Moreno et al., 2004; Lust et al., 2022; Woych et al., 2022). However, little is known about the evolution of the medial pallium transcriptional profile in amniotes and anamniotes. In a comparative analysis of the expression pattern of conserved markers in the amphibian Xenopus laevis and in the amniote Trachemys scripta, Jiménez and Moreno reported that, despite cytoarchitectural differences in the layering of the medial pallium, expression of the gene Prox1 and transcription factors Er81 and Lmo4 was shared with the mammalian dentate gyrus, thus providing evidence of a common genoarchitectonics supporting the functional involvement in memory tasks.

Another hallmark of superior brain function is reciprocal connections between the cerebral cortex and the thalamus. The cortico-thalamic and thalamo-cortical circuits elaborate essential tasks such as wakefulness, sensory processing, learning and memory, plasticity, and consciousness. Disorders that affect higher brain functions, including schizophrenia, bipolar disorder, and autism spectrum disorders, impact this system. Angulo Salavarria et al. summarize cortico-thalamic formation, starting with the prosomeric model of neurodevelopment (Rubenstein et al., 1994; Puelles et al., 2013). The cerebral cortex and the thalamus operate as a single unit, and the establishment of their reciprocal connections was observed in the human embryo at ~7.5/8 post-conceptional weeks. Various models have been used to study the molecular and cellular mechanisms of cortico-thalamic development. In parallel to animal models, which remain fundamental for unveiling neural network establishment, the authors critically discuss advanced in vitro platforms, e.g., brain organoids and assembloids derived from human pluripotent stem cells. These innovative tools have substantial potential for basic research in brain development and dysfunctions. Furthermore, in silico techniques are used to mimic composite brain circuitry, simulating realistic inputs/outputs and clarifying neuron interaction in complex networks.

The arrangement of minicolumns is another crucial aspect of cortical structure that has undergone evolutionary changes across different species (Buxhoeveden and Casanova, 2002). Morphologically defined as strings of interconnected neurons extending radially across layers 2–6 (Rakic, 1988), minicolumns are the elemental processing unit of the neocortex and have been detected in diverse cortical areas. The iterative repetition of these structures is thought to be fundamental to the neocortical expansion that has characterized brain size augmentation with evolution and the increase in computational power (Rakic, 1995, 2008). Here, Wallace et al. explore features of minicolumns present in the primary visual cortex (V-1) in five mammalian orders: human and non-human primates (Homo sapiens, Pan troglodytes, and Gorilla gorilla), rodents (Cavia porcellus, Mus musculus, and Rattus rattus), Eulipotyphla (Erinaceous europeus), Artiodactyla (Sus scrofa), and Carnivora (Mustela putorius). The authors describe a spatial arrangement of minicolumnar bundles of the primary visual cortex (V-1) in linear or branched strings with variable intra-layer length, density, and intracolumnar distance, depending on the species (Wallace et al.). In general, V-1 minicolumns spanned from the base of layer 3 to the white matter in all great apes including humans, and carnivores, whereas other mammalian orders had a diverse structure made of repeating modules or microcolumns with a shorter layer extension and more irregularity in the spatial patterning. There was a strong association between the abundance of minicolumns and visual acuity, thus indicating the existence of a relationship between a numerical parameter in cortical cytoarchitecture and an indicative function of the computational power.

Several observations have supported the notion that non-neuronal cells, specifically astroglia, may affect cortex development and evolution. Findings included unique characteristics of primate astroglia, implying a potential contribution to cortical processes (Oberheim et al., 2009; Zhang et al., 2016; Vasile et al., 2017; Falcone and Martinez-Cerdeno, 2023). The study by Degl'Innocenti and Dell'Anno summarized prevalent astrocyte differences between mice and humans in an overview that starts with cortical astrogliogenesis and includes morphological and functional differences. Compared with rodents, human astrocytes have a higher degree of complexity in size, morphology, and extension of intercellular interactions, all aspects that reflect their increased capacity in fostering synaptic transmission and increasing mouse cognitive capacities upon engraftment (Han et al., 2013). Human astrocytes are generated from the ventricular and subventricular zones, similarly to the mouse. However, human-specific glial precursors have been identified in the basal or outer radial glia cells, the prominent gliogenic capacity of which is responsible for the thickening of the cortex and the development of convolutions (Rash et al., 2019).

In summary, the findings in this Research Topic offer a comprehensive overview of factors participating in cortex development and key divergences that have led to the acquisition of distinctive species-specific features. These elements collectively contribute to our understanding of the intricate processes shaping the cortex and its evolutionary trajectory. The data in these five articles, in conjunction with studies outside this Research Topic, should provide clues for uncovering the logic behind the vast heterogeneity of the human cerebral cortex, not only to reveal underlying mechanisms in neurological or psychiatric disorders but also to disclose the neurobiological elements conferring the uniqueness, multifaceted talents, and capacities of the human brain.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Acknowledgments

The editors acknowledge and thank the respective authors for their contributions to this special collection. The editors also thank Prof. John P. Kastelic for his support and critical reading of the manuscript.

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.

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

Alvarez-Buylla, A., and Lim, D. A. (2004). For the long run: maintaining germinal niches in the adult brain. Neuron. 41, 683–686. doi: 10.1016/S0896-6273(04)00111-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Buxhoeveden, D. P., and Casanova, M. F. (2002). The minicolumn and evolution of the brain. Brain Behav. Evol. 60, 125–151. doi: 10.1159/000065935

PubMed Abstract | CrossRef Full Text | Google Scholar

Cadwell, C. R., Bhaduri, A., Mostajo-Radji, M. A., Keefe, M. G., and Nowakowski, T. J. (2019). Development and arealization of the cerebral cortex. Neuron 103, 980–1004. doi: 10.1016/j.neuron.2019.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Falcone, C., and Martinez-Cerdeno, V. (2023). Astrocyte evolution and human specificity. Neural Regen. Res. 18, 131–132. doi: 10.4103/1673-5374.340405

PubMed Abstract | CrossRef Full Text | Google Scholar

Franzoni, E., Booker, S. A., Parthasarathy, S., Rehfeld, F., Grosser, S., Srivatsa, S., et al. (2015). miR-128 regulates neuronal migration, outgrowth and intrinsic excitability via the intellectual disability gene Phf6. Elife. 4, e04263. doi: 10.7554/eLife.04263

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, X., Chen, M., Wang, F., Windrem, M., Wang, S., Shanz, S., et al. (2013). Forebrain engraftment by human glial progenitor cells enhances synaptic plasticity and learning in adult mice. Cell Stem Cell. 12, 342–353. doi: 10.1016/j.stem.2012.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Kast, R. J., and Levitt, P. (2019). Precision in the development of neocortical architecture: from progenitors to cortical networks. Prog. Neurobiol. 175, 77–95. doi: 10.1016/j.pneurobio.2019.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Letellier, M., Elramah, S., Mondin, M., Soula, A., Penn, A., Choquet, D., et al. (2014). miR-92a regulates expression of synaptic GluA1-containing AMPA receptors during homeostatic scaling. Nat. Neurosci. 17, 1040–1042. doi: 10.1038/nn.3762

PubMed Abstract | CrossRef Full Text | Google Scholar

Lust, K., Maynard, A., Gomes, T., Fleck, J. S., Camp, J. G., Tanaka, E. M., et al. (2022). Single-cell analyses of axolotl telencephalon organization, neurogenesis, and regeneration. Science 377, eabp9262. doi: 10.1126/science.abp9262

PubMed Abstract | CrossRef Full Text | Google Scholar

Marin, O., and Rubenstein, J. L. (2003). Cell migration in the forebrain. Annu. Rev. Neurosci. 26, 441–483. doi: 10.1146/annurev.neuro.26.041002.131058

PubMed Abstract | CrossRef Full Text | Google Scholar

Moreno, N., Bachy, I., Retaux, S., and Gonzalez, A. (2004). LIM-homeodomain genes as developmental and adult genetic markers of Xenopus forebrain functional subdivisions. J. Comp. Neurol. 472, 52–72. doi: 10.1002/cne.20046

PubMed Abstract | CrossRef Full Text | Google Scholar

Oberheim, N. A., Takano, T., Han, X., He, W., Lin, J. H., Wang, F., et al. (2009). Uniquely hominid features of adult human astrocytes. J. Neurosci. 29, 3276–3287. doi: 10.1523/JNEUROSCI.4707-08.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, Y., Page, N., Salamon, I., Li, D., and Rasin, M. R. (2022). Making sense of mRNA landscapes: translation control in neurodevelopment. Wiley Interdiscip. Rev. RNA 13, e1674. doi: 10.1002/wrna.1674

PubMed Abstract | CrossRef Full Text | Google Scholar

Puelles, L., Harrison, M., Paxinos, G., and Watson, C. (2013). A developmental ontology for the mammalian brain based on the prosomeric model. Trends Neurosci. 36, 570–578. doi: 10.1016/j.tins.2013.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Rakic, P. (1988). Specification of cerebral cortical areas. Science 241, 170–176. doi: 10.1126/science.3291116

PubMed Abstract | CrossRef Full Text | Google Scholar

Rakic, P. (1995). A small step for the cell, a giant leap for mankind: a hypothesis of neocortical expansion during evolution. Trends Neurosci. 18, 383–388. doi: 10.1016/0166-2236(95)93934-P

PubMed Abstract | CrossRef Full Text | Google Scholar

Rakic, P. (2008). Confusing cortical columns. Proc. Natl. Acad. Sci. U. S. A. 105, 12099–12100. doi: 10.1073/pnas.0807271105

PubMed Abstract | CrossRef Full Text | Google Scholar

Rash, B. G., Duque, A., Morozov, Y. M., Arellano, J. I., Micali, N., Rakic, P., et al. (2019). Gliogenesis in the outer subventricular zone promotes enlargement and gyrification of the primate cerebrum. Proc. Natl. Acad. Sci. U. S. A. 116, 7089–7094. doi: 10.1073/pnas.1822169116

PubMed Abstract | CrossRef Full Text | Google Scholar

Rubenstein, J. L., Martinez, S., Shimamura, K., and Puelles, L. (1994). The embryonic vertebrate forebrain: the prosomeric model. Science 266, 578–580. doi: 10.1126/science.7939711

PubMed Abstract | CrossRef Full Text | Google Scholar

Salas, C., Broglio, C., Duran, E., Gomez, A., Ocana, F. M., Jimenez-Moya, F., et al. (2006). Neuropsychology of learning and memory in teleost fish. Zebrafish. 3, 157–171. doi: 10.1089/zeb.2006.3.157

PubMed Abstract | CrossRef Full Text | Google Scholar

Shu, P., Fu, H., Zhao, X., Wu, C., Ruan, X., Zeng, Y., et al. (2017). MicroRNA-214 modulates neural progenitor cell differentiation by targeting Quaking during cerebral cortex development. Sci. Rep. 7, 8014. doi: 10.1038/s41598-017-08450-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Sidman, R. L., and Rakic, P. (1973). Neuronal migration, with special reference to developing human brain: a review. Brain Res. 62, 1–35. doi: 10.1016/0006-8993(73)90617-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Sotelo, M. I., Daneri, M. F., Bingman, V. P., and Muzio, R. N. (2016). Telencephalic neuronal activation associated with spatial memory in the terrestrial toad Rhinella arenarum: participation of the medial pallium during navigation by geometry. Brain Behav. Evol. 88, 149–160. doi: 10.1159/000447441

PubMed Abstract | CrossRef Full Text | Google Scholar

Tomasello, U., Klingler, E., Niquille, M., Mule, N., Santinha, A. J., de Vevey, L., et al. (2022). miR-137 and miR-122, two outer subventricular zone non-coding RNAs, regulate basal progenitor expansion and neuronal differentiation. Cell Rep. 38, 110381. doi: 10.1016/j.celrep.2022.110381

PubMed Abstract | CrossRef Full Text | Google Scholar

Vasile, F., Dossi, E., and Rouach, N. (2017). Human astrocytes: structure and functions in the healthy brain. Brain Struct. Funct. 222, 2017–2029. doi: 10.1007/s00429-017-1383-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Woych, J., Ortega Gurrola, A., Deryckere, A., Jaeger, E. C. B., Gumnit, E., Merello, G., et al. (2022). Cell-type profiling in salamanders identifies innovations in vertebrate forebrain evolution. Science 377, eabp9186. doi: 10.1126/science.abp9186

PubMed Abstract | CrossRef Full Text | Google Scholar

Zahr, S. K., Yang, G., Kazan, H., Borrett, M. J., Yuzwa, S. A., Voronova, A., et al. (2018). A translational repression complex in developing mammalian neural stem cells that regulates neuronal specification. Neuron 97, 520–37.e6. doi: 10.1016/j.neuron.2017.12.045

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Sloan, S. A., Clarke, L. E., Caneda, C., Plaza, C. A., Blumenthal, P. D., et al. (2016). Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53. doi: 10.1016/j.neuron.2015.11.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cortical development, cortical evolution, astrogenesis, minicolumn, cortico-thalamic circuit

Citation: Dell'Anno MT, Conti L and Onorati M (2023) Editorial: Molecular and cellular logic of cerebral cortex development, evolution, and disease. Front. Neuroanat. 17:1242684. doi: 10.3389/fnana.2023.1242684

Received: 19 June 2023; Accepted: 22 June 2023;
Published: 07 July 2023.

Edited and reviewed by: Gavin John Clowry, Newcastle University, United Kingdom

Copyright © 2023 Dell'Anno, Conti and Onorati. 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: Maria Teresa Dell'Anno, mt.dellanno@fpscience.it; Marco Onorati, marco.onorati@unipi.it

Download