Skip to main content

OPINION article

Front. Neuroanat., 13 February 2023
This article is part of the Research Topic What Makes Us Human: From Genes to Machine View all 7 articles

A brief sketch across multiscale and comparative neuroanatomical features

  • Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States

Introduction

In 2018, at the Jerusalem conference which inspired this research topic, I repeated the call for more human neuroanatomy (cf. Crick and Jones, 1993) and talked about the related question of human-specific features. Since then, considerable progress has been made in neuroanatomical techniques (e.g., 3-D visualization of whole brains with or without tissue clearing), although the database for human neuroanatomy is still limited. Increasingly apparent, however, is that human specializations are found at multiple scales, from the subcellular and genetic to the macro- and extra-neural (e.g., Axer and Amunts, 2022). This points to a multifactorial answer to “what makes us human” or, rather, to a slightly different question: how do these factors interact across temporal and spatial scales, and what about that is human-specific? As an indirect approach to this question, this opinion is a brief canvas of what may appear as separate topics, although one can sense the building momentum in the research community toward a unified broad systems-level approach. I have neglected important topics of comparative oscillatory signatures as well as prenatal and early developmental processes and confined discussion largely to features of the cerebral cortex.

Cortical architecture

A widely shared neocortical feature is the organization of six cellular layers. In humans and also in nonhuman primates (NHP), the cellular supragranular layers are thicker, perhaps in accord with a greater abundance and role of the ipsilateral and contralateral corticocortical connections originating preferentially (but not exclusively) from the upper layers (DeFelipe, 2011; Rockland, 2019).

There are other large-brain species with sophisticated behaviors and social structures; but in striking contrast with humans and NHP, cortical areas in elephants, whales, and other cetaceans are pyramidalized, without a small cell layer 4 (Graic et al., 2022). This is a major difference in terms of input organization. Thalamocortical and other inputs, instead of targeting excitatory local neurons in layer 4 together with dendritic elements in the passage, can be assumed to directly target basal or apical dendrites of pyramidal cells. In addition to the reduction of layer 4, layer 1 is thicker in both elephants and cetaceans.

A pronounced verticality, conspicuous in cellular and myelin preparations, is typical of neocortical areas but with species, area, and individual variations that have not been widely systematized (Molnar and Rockland, 2000; DeFelipe, 2011). For apical dendritic bundles, a relatively identifiable feature, quantitative measures exist for dendritic number and diameter per bundle and for bundle-to-bundle spacing but the area and species census is sporadic (Gabbott, 2003).

Cell packing density

Neuron density varies across species, where average cell density is least in humans, and progressively more dense in chimpanzees, macaques, rats, and mice (Figure 13 in DeFelipe, 2011). The reduced cell density complements an increase in neuropil space and synaptic density, which would be consistent with more complex neural processing in larger brains. A greater number of neurons has often been posited as correlating with human cognitive abilities; but a less neuron-centric view assigns a prominent role to biochemical variations and interactions of neurotransmitters, neurotrophins, and other substances facilitating communication (cf. Saniotis et al., 2020; Chin et al., 2022).

Cell types

Broad morphological neuronal subtypes are recognizable across mammalian species; for example, cortical interneurons, cortical pyramidal neurons, and cerebellar Purkinje cells. For the typical cortical pyramidal neurons with apical dendritic tufts in layer 1, species-specific morphometric modifications have been reported in the size of the basal dendritic arbor (Elston, 2003), the length of the apical dendrite (Galloni et al., 2020), the number of dendritic branches, and the number of postsynaptic dendritic spines. These parameters are all significant for electrical properties and microcircuitry processing and are increased in humans and NHP by comparison with rodents (Gilman et al., 2017).

Layer 3 magnocellular pyramidal neurons in the cognitively important human prefrontal cortex have a protracted multi-stage maturational process of the dendritic arbor, with an additional phase of extensive dendritic growth during early childhood (Petanjek et al., 2019). One consequence of the protracted circuitry reorganization with synaptic overproduction and pruning is a great opportunity for gene–environment interactions.

Morphologically distinct cell types, such as Von Economo cells, Betz cells, and Meynert cells, occur in humans but are also reported in NHP, as well as in some non-primate species (Jacobs et al., 2018). Thus, human-specific features may be primarily quantitative in nature and/or at the subcellular level of receptors, ion channels, and input distribution (cf. Hodge et al., 2019). At the transcriptomic level of analysis, still ongoing results so far indicate a high degree of diversity for both interneurons and pyramidal cells (e.g., Galakhova et al., 2022 for a recent discussion). An apparently human-specific GABAeric cell type has been identified in layer 1 (rosehip: Boldog et al., 2018).

In contrast with the relatively subtle morphological differences in neurons, pronounced structural differentiations have been identified for glia; namely, humans have larger and structurally more complex protoplasmic and fibrous astrocytes, and several distinct glia subclasses not so far identified in rodents (Oberheim et al., 2009; Falcone et al., 2021). These variations are functionally significant, as astrocytes are involved in the maintenance of the blood–brain barrier; synapse formation, elimination, and plasticity; calcium flux regulation; and modulation of neural activity. Astrocytes communicate by gap junctions. The increased volume of an astrocytic domain in humans carries an increased synaptic number (humans: 270,000–2 million synapses vs. 20,000–120,000 synapses in rodents); and a larger glial-neuronal unit can be hypothesized as adapted for more complex processing (Oberheim et al., 2009).

Nano-architecture

The subcellular level of organization has been relatively under-investigated but is an important source of functionally relevant comparative data (Garcia-Etxarri and Yuste, 2021). Ultrastructural analyses have already indicated synaptic structural specializations in humans. Cortical synapses in layer 5 of the human temporal cortex have 2–3 fold larger active zones, a larger pool of synaptic vesicles (releasable, recycling, and resting), and tighter astrocytic ensheathment—all features compatible with greater efficiency (Rollenhagen et al., 2020). Strong and reliable synaptic communication, as compared with the mouse, has been verified by in vitro electrophysiology of surgical samples from the human temporal cortex (Hunt et al., 2022).

Cytoskeletal features are being revealed to a new degree of intricacy and diversity, impacting the understanding of neuronal trafficking and axonal transport mechanisms (Leterrier et al., 2017). The axon initial segment, readily identifiable across species, has specific morpho-functional features which are not fixed but changeable in neural activity or pathological conditions (Anton-Fernandez et al., 2022).

The extracellular space is increasingly appreciated as an integral component of brain architecture, forming a complex 3D structure that occupies about 20% of brain volume (Hrabetova et al., 2018). Area and species differences will impact brain fluid dynamics and homeostasis (Rasmussen et al., 2022).

Myelination provides a further rich field for the investigation of species-specific specializations (Fields, 2015). A human-specific hallmark is developmentally prolonged cortical myelination, allowing time for several epochs of reorganization. Data from rodents strongly support the idea of differences in myelin thickness and spacing of nodes of Ranvier in both activity-dependent and -independent conditions (de Faria et al., 2021). Transcriptomic profiles in humans and mice, an important model of human myelin disorders, have been reported as well-correlating but with a divergent expression of distinct genes (Gargareta et al., 2022).

Asymmetries

Hemispheric asymmetries (e.g., Thiebaut de Schotten and Beckmann, 2022) have been closely associated with human-specific features such as manual dexterity and language; and a left hemisphere (LH) prefrontal hyperscaling is put forth as a neural adaptive shift distinguishing the ape and monkey radiations (Smaers et al., 2011). Other asymmetries, including in humans (Toga and Thompson, 2003), are developmental (the right hemisphere (RH) develops earlier and faster) and neurochemical (higher dopamine levels in the LH and higher adrenaline in the RH). There is a preponderance of cilia-associated genes in the RH but cell adhesion-associated genes in the LH (Lubben et al., 2021). Hemispheric asymmetry has been attributed to multiple mechanisms, both genetic and environmental (Buckner et al., 2009). The right-left asymmetry of frontal and parieto-occipital regions has been referenced to a lateralized gradient of embryological development, specifically in humans (Xiang et al., 2019); and a genetic association has been reported in relation to the typical counterclockwise brain torque (Zhao et al., 2022).

A left-right cerebral torque appears to be human-specific, where the RH has an anterior protrusion but the LH protrudes posteriorly. There is individual variability (40% of subjects exhibit torque), and the torque is reported to be less common in females (Xiang et al., 2019).

Several brainstem nuclei are identified as specific to humans, with species-variable neurochemical differences in homologous nuclei and pronounced left-right asymmetry in humans (Baizer, 2014). These changes in brainstem organization were attributed to the unique human motor and cognitive abilities, as associated with the parallel expansions of cortical and cerebellar structures (Refer to Smaers and Soligo, 2013 on “brain reorganization not relative brain size.”).

Connectivity

Comparisons between NHP and humans are a rich source of inquiry on human-specific brain organization. Recently, a large proportion of this research is from functional imaging or tractography, not treated here (but refer, for recent review and references, to Thiebaut de Schotten and Forkel, 2022). New data on differential human-specific features of connectivity can be expected from three-dimensional multimodal atlases of large cohorts of human subjects, as compatible with cross-species comparisons (Thiebaut de Schotten et al., 2019).

The clinically important corticospinal (CS) tract serves as a representative example of species differences. The CS tract contains a wide spectrum of fiber diameters, of which a subpopulation of large-diameter fibers (up to 22 vs. 12 μm in macaques) is characteristic of humans (Lemon, 2021). Fast-conducting large fibers are associated with reduced conduction delays which are considered important in movement and skilled grasp transitions (summarized in Lemon, 2021). For a comparable discussion of the corpus callosum, refer to Innocenti et al., 2022.

Another variable is the proportion of crossed fibers: 80–95% in rodents but 75–90% in humans, with noticeable individual variability (Welniarz et al., 2017). Furthermore, a large number of cortical areas contribute to the CS tract in humans, from parts of the frontal, cingulate, and parietal cortices in addition to the primary motor (the latter estimated as 50%; Lemon, 2021). Convergence of these diverse inputs in the spinal cord and their communication by corticocortical connections is likely to subserve other than purely motor functions.

The gray matter terminations of long-distance axons are difficult to investigate in large brains and especially in humans, where standard anatomical techniques cannot be used. There are some data from labeling by antibodies or, increasingly, dense ultrastructural analyses of small tissue volumes in postmortem material or surgical samples. Otherwise, results in experimental animals need to serve as a provisional guide (Rockland, 2020). Likely homologs might be thalamocortical axons in the primary sensory areas, the highly conserved pattern of corticothalamic projections from layers 5 and 6 to non-sensory thalamic nuclei (Rouiller and Welker, 2000), and the divergent collateralization pattern of layer 5 cortico-brainstem neurons (Coude et al., 2018). Organizational differences (e.g., larger arbors and more boutons) can be expected since even homologous structures have modified characteristics. In primates, but not in rodents, the thalamus is densely innervated by dopaminergic fibers (Garcia-Cabezas et al., 2009) and the primate thalamus has both excitatory projection neurons (positive for calbindin and parvalbumin) and inhibitory interneurons, the latter of which are only sparsely present in the rodent thalamus (Arcelli et al., 1997).

In mice, connectivity analysis is approaching routine at the level of whole neuron/whole axon (and Xu et al., 2021 in macaque; McElvain et al., 2021; Gao et al., 2022). A general conclusion from this approach is that individual axons within the same source–target system are highly variable in branching patterns, size of single arbors, and the number of synaptic boutons. This is consistent with smaller sample sizes from previous work in NHP (refer to references in Rockland, 2020).

Conclusion

In this brief and selective opinion, I have attempted to highlight several examples of human-specific characteristics. There are, of course, many others, such as species differences in the cerebrospinal fluid, lifespan changes in myelination and other parameters, and species characteristics of the brain oscillations and sleep cycles. An important direction is a closer integration of cognitive and systems neuroscience with the cellular, subcellular, and molecular levels of ion channels, receptors, and other components of signal transduction and metabolic pathways (cf. Raichle, 2015 among others).

As a broad systems-level approach will include cultural and environmental influences, a key question is how external factors access and influence neuronal substrates. The influence of external factors has commonly been assayed by changes in dendritic spine density in response to environmental stressors, or aging and pathology (e.g., Jacobs et al., 2022) but can be expected to impact a large number of interacting elements across scales.

Author contributions

The author confirms being the sole contributor of this work and has approved it for publication.

Funding

Publication fees were generously contributed by the Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine.

Conflict of interest

The author declares 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

Anton-Fernandez, A., Leon-Espinosa, G., DeFelipe, J., and Munoz, A. (2022). Pyramidal cell axon initial segment in Alzheimer's disease. Sci. Rep. 12, 8722. doi: 10.1038/s41598-022-12700-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Arcelli, P., Frassoni, C., Regondi, M. C., De Biasi, S., and Spreafico, R. (1997). GABAergic neurons in the mammalian thalamus: a marker of thalamic complexity? Brain Res. Bull. 42, 27–37. doi: 10.1016/S0361-9230(96)00107-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Axer, M., and Amunts, K. (2022). Scale matters: the nested human connectome. Science 378, 500–504. doi: 10.1126/science.abq2599

PubMed Abstract | CrossRef Full Text | Google Scholar

Baizer, J. S. (2014). Unique features of the human brainstem and cerebellum. Front. Hum. Neurosci. 8, 1–17. doi: 10.3389/fnhum.2014.00202

PubMed Abstract | CrossRef Full Text | Google Scholar

Boldog, E., Bakken, T. E., Hodge, R. D., Novotny, M., Aevermann, B. D., Baka, J., et al. (2018). Transcriptomic and morphophysiological evidence for a specialized human cortical GABAergic cell type. Nat. Neurosci. 21, 1185–1195. doi: 10.1038/s41593-018-0205-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Buckner, R. L., Sepulcre, J., Talukdar, T., Krienen, F. M., Liu, H., Hedden, T., et al. (2009). Cortical hubs revealed by intrinsic functional connectivity: mapping, assessment of stability, and relation to Alzheimer's disease. J. Neurosci. 29, 1869–1873. doi: 10.1523/JNEUROSCI.5062-08.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Chin, R., Chang, S. W. C., and Holmes, A. J. (2022). Beyond cortex: the evolution of the human brain. Psychol. Rev. 14, 1–23. doi: 10.1037/rev0000361

PubMed Abstract | CrossRef Full Text | Google Scholar

Coude, D., Parent, A., and Parent, M. (2018). Single-axon tracing of the corticosubthalamic hyperdirect pathway in primates. Brain Struct Funct. 223, 3959–3973. doi: 10.1007/s00429-018-1726-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Crick, F., and Jones, E. (1993). Backwardness of human neuroantomy. Nature 361, 109–110. doi: 10.1038/361109a0

PubMed Abstract | CrossRef Full Text | Google Scholar

de Faria, O., Pivonkova, H., Varga, B., Timmler, S., Evans, K. A., and Káradóttir, R. T. (2021). Periods of synchronized myelin changes shape brain function and plasticity. Nat. Neurosci. 24, 1508–1521. doi: 10.1038/s41593-021-00917-2

PubMed Abstract | CrossRef Full Text | Google Scholar

DeFelipe, J. (2011). The evolution of the brain, the human nature of cortical circuits, and intellectual creativity. Front. Neuroanatomy 5, 29. doi: 10.3389/fnana.2011.00029

PubMed Abstract | CrossRef Full Text | Google Scholar

Elston, G. N. (2003). Cortex, cognition and the cell: new insights into the pyramidal neuron and prefrontal function. Cereb. Cortex 13, 1124–1138. doi: 10.1093/cercor/bhg093

PubMed Abstract | CrossRef Full Text | Google Scholar

Falcone, C., Penna, E., Hong, T., Tarantal, A. F., Hof, P. R., Hopkins, W. D., et al. (2021). Cortical interlaminar astrocytes are generated prenatally, mature postnatally, and express unique markers in human and nonhuman primates. Cereb. Cortex 31, 379–395. doi: 10.1093/cercor/bhaa231

PubMed Abstract | CrossRef Full Text | Google Scholar

Fields, R. D. (2015). A new mechanism of nervous system plasticity: activity-dependent myelination. Nat. Rev Neurosci. 16, 756–767. doi: 10.1038/nrn4023

PubMed Abstract | CrossRef Full Text | Google Scholar

Gabbott, P. L. A. (2003). Radial organisation of neurons and dendrites in human cortical areas 25, 32. and 32'. Brain Res. 992, 298–304. doi: 10.1016/j.brainres.2003.08.054

PubMed Abstract | CrossRef Full Text | Google Scholar

Galakhova, A. A., Hunt, S., Wilbers, R., Heyer, D. B., de Kock, H. D, Mansvelder, C. P. J., et al. (2022). Evolution of cortical neurons supporting human cognition. Trends Cog. Sci. 26, 909–922. doi: 10.1016/j.tics.2022.08.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Galloni, A. R., Laffere, A., and Rancz, E. (2020). Apical length governs computational diversity of layer 5 pyramidal neurons. eLife 9, e55761. doi: 10.7554/eLife.55761.sa2

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, L., Liu, S., Gou, L., Hu, Y., Liu, Y., Deng, L., et al. (2022). Single-neuron projectome of mouse prefrontal cortex. Nat. Neuro. 25, 515–529. doi: 10.1038/s41593-022-01041-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia-Cabezas, M. A., Martinez-Sanchez, P., Sachez-Gonzalez, M. A., Garzon, M., and Cavada, C. (2009). dopamine innervation in the thalamus: monkey versus rat. Cereb. Cortex 9, 424–434. doi: 10.1093/cercor/bhn093

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia-Etxarri, A., and Yuste, R. (2021). Time for NANONEUro. Nat. Methods 18, 1287–1293. doi: 10.1038/s41592-021-01270-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Gargareta, V. I., Reuschenbach, J., Siems, S. B., Sun, T., Piepkorn, L., Mangana, C., et al. (2022). Conservation and divergence of myelin proteome and oligodendrocyte transcriptome profiles between humans and mice. Elife 11, e77019. doi: 10.7554/eLife.77019.sa2

PubMed Abstract | CrossRef Full Text | Google Scholar

Gilman, J. P., Medalla, M., and Luebke, J. I. (2017). Area-specific features of pyramidal neurons -a comparative study in mouse and rhesus monkey. Cerebral Cortex 27, 2078–2094. doi: 10.1093/cercor/bhw062

PubMed Abstract | CrossRef Full Text | Google Scholar

Graic, J. M., Peruffo, A., Corain, L., Finos, L., Grisan, E., Cozzi, B., et al. (2022). The primary visual cortex of Cetartiodactyls: organization, cytoarchitectonics and comparison with perissodactyls and primates. Brain Structure Funct. 227, 1195–1225. doi: 10.1007/s00429-021-02392-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Hodge, R. D., Bakken, T. E., Miller, J. A., Smith, K. A., Barkan, E. R., Graybuck, L. T., et al. (2019). Conserved cell types with divergent features in human versus mouse cortex. Nature 573, 61–68. doi: 10.1038/s41586-019-1506-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Hrabetova, S., Cognet, L., Rusakov, D. A., and Nagerl, U. V. (2018). Unveiling the extracellular space of the brain: from super-resolved microstructure to in vivo function. J. Neurosci. 38, 9355–9363. doi: 10.1523/JNEUROSCI.1664-18.2018

PubMed Abstract | CrossRef Full Text | Google Scholar

Hunt, S., Leibner, Y., Mertens, E. J., Barros-Zulaica, N., Kanari, L., Heistek, T. S., et al. (2022). Strong and reliable synaptic communication between pyramidal neurons in adult human cerebral cortex. Cerebral Cortex 1, 22. doi: 10.1093/cercor/bhac246

PubMed Abstract | CrossRef Full Text | Google Scholar

Innocenti, G. M., Schmidt, K., Milleret, C., Fabri, M., Knyazeva, M. G., Battaglia-Mayer, A., et al. (2022). The functional characterization of callosal connections. Prog. Neurobiol. 208, 1–24. doi: 10.1016/j.pneurobio.2021.102186

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobs, B., Garcia, M. E., Shea-Shumsky, N. B., Tennison, M. E., Schall, M., Saviano, M. S., et al. (2018). Comparative morphology of gigantopyramidal neurons in primary motor cortex across mammals. J. Comp. Neurol. 526, 496–536. doi: 10.1002/cne.24349

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobs, B., Rally, H., Doyle, C., O'Brien, L., Tennison, M., Marino, L., et al. (2022). Putative neuronal consequences of captivity for elephants and cetaceans. Rev. Neurosci. 33, 439–465. doi: 10.1515/revneuro-2021-0100

PubMed Abstract | CrossRef Full Text | Google Scholar

Lemon, R. N. (2021). The cortical “upper motoneuron” in health and disease. Brain Sci. 11, 619. doi: 10.3390/brainsci11050619

PubMed Abstract | CrossRef Full Text | Google Scholar

Leterrier, C., Dubey, P., and Ray, S. (2017). The nano-architecture of the axonal cytoskeleton. Nat. Rev. Neurosci. 18, 713–726. doi: 10.1038/nrn.2017.129

PubMed Abstract | CrossRef Full Text | Google Scholar

Lubben, N., Ensink, E., Coetzee, G. A., and Labrie, V. (2021). The enigma and implications of brain hemispheric asymmetry in neurodegenerative diseases. Brain Commun. 3, fcab211. doi: 10.1093/braincomms/fcab211

PubMed Abstract | CrossRef Full Text | Google Scholar

McElvain, L. E., Chen, Y., Moore, J. D., Brigidi, G. S., Bloodgood, B. L., Lim, B. K., et al. (2021). Specific populations of basal ganglia output neurons target distinct brain stem areas while collateralizing throughout the diencephalon. Neuron 109, 1721–1738. doi: 10.1016/j.neuron.2021.03.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Molnar, Z., and Rockland, K. S. (2000). Cortical Columns. Neural Circuit and Cognitive Development. 2nd Edn. New York, NY: Academic Press, 103–126.

Google Scholar

Oberheim, N. A., Takano, T., Han, X., He, W., Lin, J. H. C., 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

Petanjek, Z., Sedmak, D., DŽaja, D., Hladnik, A., Rašin, M. R., Jovanov-Milosevic, N., et al. (2019). The protracted maturation of associative layer IIIC pyramidal neurons in the human prefrontal cortex during childhood: a major role in cognitive development and selective alteration in autism. Front. Psychiatry 10, 1–33. doi: 10.3389/fpsyt.2019.00122

PubMed Abstract | CrossRef Full Text | Google Scholar

Raichle, M. E. (2015). The restless brain: how intrinsic activity organizes brain function. Phil. Trans. R. Soc. B 370, 1–11. doi: 10.1098/rstb.2014.0172

PubMed Abstract | CrossRef Full Text | Google Scholar

Rasmussen, M. K., and Mestre, H., and Nedergaard,. M. (2022). Fluid transport in the brain. Pysiol. Rev. 102, 1025–1151. doi: 10.1152/physrev.00031.2020

PubMed Abstract | CrossRef Full Text | Google Scholar

Rockland, K. S. (2019). What do we know about laminar connectivity? Neuroimage 197, 772–784. doi: 10.1016/j.neuroimage.2017.07.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Rockland, K. S. (2020). What we can learn from the complex architecture of single axons. Brain Struct. Funct. 225, 1327–1347. doi: 10.1007/s00429-019-02023-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Rollenhagen, A., Walkenfort, B., Yakoubi, R., Klauke, S. A., Schmuhl-Giesen, S. F., Heinen-Weiler, J., et al. (2020). Synaptic organization of the human temporal lobe neocortex as revealed by high-resolution transmission, focused ion beam scanning, and electron microscopic tomography. Int. J. Mol. Sci. 21, 558. doi: 10.3390/ijms21155558

PubMed Abstract | CrossRef Full Text | Google Scholar

Rouiller, E. M., and Welker, E. (2000). A comparative analysis of the morphology of corticothalamic projections in mammals. Brain Res. Bullet. 53, 727–741. doi: 10.1016/S0361-9230(00)00364-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Saniotis, A., Grantham, J. P., Kumaratilake, J., and Henneberg, M. (2020). Neuro-hormonal regulation is a better indication of human cognition abilities than brain anatomy: the need for a new paradigm. Front. Neuroanatomy 13, 1–11. doi: 10.3389/fnana.2019.00101

PubMed Abstract | CrossRef Full Text | Google Scholar

Smaers, J. B., and Soligo, C. (2013). Brain reorganization, not relative brain size, primarily characterizes anthropoid brain evolution. Proc. Roy. Soc. London B 280, 1–8. doi: 10.1098/rspb.2013.0269

PubMed Abstract | CrossRef Full Text | Google Scholar

Smaers, J. B., Steele, J., Case, C. R., Cowper, A., Amunts, K., Zilles, K., et al. (2011). Primate prefrontal cortex evolution: human brains are the extreme of a lateralized ape trend. Brain Behav. E77, 67–78. doi: 10.1159/000323671

PubMed Abstract | CrossRef Full Text | Google Scholar

Thiebaut de Schotten, M., and Beckmann, C. F. (2022). Asymmetry of brain structure and function: 40 years after Sperry's Nobel prize. Brain Struct. Function 227, 421–424. doi: 10.1007/s00429-021-02426-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Thiebaut de Schotten, M., Croxson, P. L., and Mars., R.B. (2019). Large-scale comparative neuroimaging: Where are we and what do we need? Cortex 118, 1880202. doi: 10.1016/j.cortex.2018.11.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Thiebaut de Schotten, M., and Forkel, S. J. (2022). The emergent properties of the connected brain. Science 378, 1–6. doi: 10.1126/science.abq2591

PubMed Abstract | CrossRef Full Text | Google Scholar

Toga, A. W., and Thompson, P. M. (2003). Mapping brain asymmetry. Nature Rev. Neurosci. 4, 37–48. doi: 10.1038/nrn1009

PubMed Abstract | CrossRef Full Text | Google Scholar

Welniarz, Q., Dusart, I., and Roze, E. (2017). The corticospinal tract: evolution, development, and human disorders. Dev. Neurobol. 77, 810–829. doi: 10.1002/dneu.22455

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiang, L., Crow, T., and Roberts, N. (2019). Cerebral torque is human specific and unrelated to brain size. Brain Struct. Funct. 224, 1141–1150. doi: 10.1007/s00429-018-01818-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, F., Shen, Y., Ding, L., Yang, C. Y., Tan, H., Wang, H., et al. (2021). High-throughput mapping of a whole rhesus monkey brain at micrometer resolution. Nat. Biotech. 39, 1521–1528. doi: 10.1038/s41587-021-00986-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, L., Matloff, W., Shi, Y., Cabeen, R. P., and Toga, A. W. (2022). Mapping complex brain torque components and their genetic architecture and phenomic associations in 24,112 individuals. Biol. Pschiatry 91, 753–768. doi: 10.1016/j.biopsych.2021.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cell types, connections, reorganization, subcellular, asymmetry, homology

Citation: Rockland KS (2023) A brief sketch across multiscale and comparative neuroanatomical features. Front. Neuroanat. 17:1108363. doi: 10.3389/fnana.2023.1108363

Received: 26 November 2022; Accepted: 10 January 2023;
Published: 13 February 2023.

Edited by:

Aviv Abraham Mezer, Edmond and Lily Safra Children's Hospital, Israel

Reviewed by:

Ricardo Insausti, University of Castilla-La Mancha, Spain

Copyright © 2023 Rockland. 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: Kathleen S. Rockland, yes krock@bu.edu

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.