全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Axon Guidance Mechanisms for Establishment of Callosal Connections

DOI: 10.1155/2013/149060

Full-Text   Cite this paper   Add to My Lib

Abstract:

Numerous studies have investigated the formation of interhemispheric connections which are involved in high-ordered functions of the cerebral cortex in eutherian animals, including humans. The development of callosal axons, which transfer and integrate information between the right/left hemispheres and represent the most prominent commissural system, must be strictly regulated. From the beginning of their growth, until reaching their targets in the contralateral cortex, the callosal axons are guided mainly by two environmental cues: (1) the midline structures and (2) neighboring? axons. Recent studies have shown the importance of axona guidance by such cues and the underlying molecular mechanisms. In this paper, we review these guidance mechanisms during the development of the callosal neurons. Midline populations express and secrete guidance molecules, and “pioneer” axons as well as interactions between the medial and lateral axons are also involved in the axon pathfinding of the callosal neurons. Finally, we describe callosal dysgenesis in humans and mice, that results from a disruption of these navigational mechanisms. 1. Introduction Interhemispheric connections are essential components of the complex neural network in eutherian animals [1, 2]. Among such connections, the corpus callosum (CC) is the most prominent commissural connection, composed of callosal axons, in the brain. In humans, the corpus callosum consists of about 200 million axons, making it the most prominent fiber tract within the central nervous system [3, 4]. Many studies have clarified the molecular mechanism involved in the development of the CC in humans using mouse experiments [5]. Callosal neurons are mostly found in layers II/III and layer V of the cerebral cortex in rodents [6]. Recently, molecules related to the identities of the general or subtypes of cortical neurons have been disclosed. Alcamo et al. reported that Satb2, a DNA-binding protein, has a key role in the specification of callosal neurons and the formation of corticocortical connections [7]. Developmentally, callosal axons from layer V first start to project to the contralateral targets, and callosal axons from the upper layers follow the preexisting axons. After the callosal axons start to elongate, they are guided by many cues within their pathfinding route [6]. Although the importance of such cues in the development of callosal axons has been known for over 30 years [8], it still remained unclear until recently how these cues help callosal axons encountering them to project precisely to their targets. Recent

References

[1]  B. D. Mitchell and J. D. Macklis, “Large-scale maintenance of dual projections by callosal and frontal cortical projection neurons in adult mice,” Journal of Comparative Neurology, vol. 482, no. 1, pp. 17–32, 2005.
[2]  R. Mihrshahi, “The corpus callosum as an evolutionary innovation,” Journal of Experimental Zoology B, vol. 306, no. 1, pp. 8–17, 2006.
[3]  A. Biegon, J. L. Eberling, B. C. Richardson et al., “Human corpus callosum in aging and Alzheimer's disease: a magnetic resonance imaging study,” Neurobiology of Aging, vol. 15, no. 4, pp. 393–397, 1994.
[4]  M. G. Funnell, P. M. Corballis, and M. Gazzaniga, “Cortical and subcortical interhemispheric interactions following partial and complete callosotomy,” Archives of Neurology, vol. 57, no. 2, pp. 185–189, 2000.
[5]  D. Kamnasaran, “Agenesis of the corpus callosum: lessons from humans and mice,” Clinical and Investigative Medicine, vol. 28, no. 5, pp. 267–282, 2005.
[6]  R. M. Fame, J. L. MacDonald, and J. D. Macklis, “Development, specification, and diversity of callosal projection neurons,” Trends in Neurosciences, vol. 34, no. 1, pp. 41–50, 2011.
[7]  E. A. Alcamo, L. Chirivella, M. Dautzenberg et al., “Satb2 regulates callosal projection neuron identity in the developing cerebral cortex,” Neuron, vol. 57, no. 3, pp. 364–377, 2008.
[8]  J. Silver, S. E. Lorenz, D. Wahlstein, and J. Coughlin, “Axonal guidance during development of the great cerebral commissures: descriptive and experimental studies, in vivo, on the role of preformed glial pathways,” Journal of Comparative Neurology, vol. 210, no. 1, pp. 10–29, 1982.
[9]  L. J. Richards, C. Plachez, and T. Ren, “Mechanisms regulating the development of the corpus callosum and its agenesis in mouse and human,” Clinical Genetics, vol. 66, no. 4, pp. 276–289, 2004.
[10]  Y. Tagawa, H. Mizuno, and T. Hirano, “Activity-dependent development of interhemispheric connections in the visual cortex,” Reviews in the Neurosciences, vol. 19, no. 1, pp. 19–28, 2008.
[11]  C. L. Wang, L. Zhang, Y. Zhou et al., “Activity-dependent development of callosal projections in the somatosensory cortex,” Journal of Neuroscience, vol. 27, no. 42, pp. 11334–11342, 2007.
[12]  H. Mizuno, T. Hirano, and Y. Tagawa, “Evidence for activity-dependent cortical wiring: formation of interhemispheric connections in neonatal mouse visual cortex requires projection neuron activity,” Journal of Neuroscience, vol. 27, no. 25, pp. 6760–6770, 2007.
[13]  R. R. Sturrock, “Identification of mitotic cells in the central nervous system by electron microscopy of re-embedded semithin sections,” Journal of Anatomy, vol. 138, no. 4, pp. 657–673, 1984.
[14]  T. Shu, Y. Li, A. Keller, and L. J. Richards, “The glial sling is a migratory population of developing neurons,” Development, vol. 130, no. 13, pp. 2929–2937, 2003.
[15]  T. Shu and L. J. Richards, “Cortical axon guidance by the glial wedge during the development of the corpus callosum,” Journal of Neuroscience, vol. 21, no. 8, pp. 2749–2758, 2001.
[16]  T. Shu, A. C. Puche, and L. J. Richards, “Development of midline glial populations at the corticoseptal boundary,” Journal of Neurobiology, vol. 57, no. 1, pp. 81–94, 2003.
[17]  J. Silver, M. A. Edwards, and P. Levitt, “Immunocytochemical demonstration of early appearing astroglial structures that form boundaries and pathways along axon tracts in the fetal brain,” Journal of Comparative Neurology, vol. 328, no. 3, pp. 415–436, 1993.
[18]  T. Shu, K. G. Butz, C. Plachez, R. M. Gronostajski, and L. J. Richards, “Abnormal development of forebrain midline glia and commissural projections in Nfia knock-out mice,” Journal of Neuroscience, vol. 23, no. 1, pp. 203–212, 2003.
[19]  S. Tole, G. Gutin, L. Bhatnagar, R. Remedios, and J. M. Hébert, “Development of midline cell types and commissural axon tracts requires Fgfr1 in the cerebrum,” Developmental Biology, vol. 289, no. 1, pp. 141–151, 2006.
[20]  S. E. Bilasy, T. Satoh, T. Terashima, and T. Kataoka, “RA-GEF-1 (Rapgef2) is essential for proper development of the midline commissures,” Neuroscience Research, vol. 71, no. 3, pp. 200–209, 2011.
[21]  C. Sánchez-Camacho, J. A. Ortega, I. Oca?a, S. Alcántara, and P. Bovolenta, “Appropriate Bmp7 levels are required for the differentiation of midline guidepost cells involved in corpus callosum formation,” Developmental Neurobiology, vol. 71, no. 5, pp. 337–350, 2011.
[22]  M. Piper, R. X. Moldrich, C. Lindwall et al., “Multiple non-cell-autonomous defects underlie neocortical callosal dysgenesis in Nfib-deficient mice,” Neural Development, vol. 4, no. 1, p. 43, 2009.
[23]  M. Piper, G. Barry, J. Hawkins et al., “NFIA controls telencephalic progenitor cell differentiation through repression of the Notch effector Hes1,” Journal of Neuroscience, vol. 30, no. 27, pp. 9127–9139, 2010.
[24]  A. Bagri, O. Marín, A. S. Plump et al., “Slit proteins prevent midline crossing and determine the dorsoventral position of major axonal pathways in the mammalian forebrain,” Neuron, vol. 33, no. 2, pp. 233–248, 2002.
[25]  W. Andrews, A. Liapi, C. Plachez et al., “Robo1 regulates the development of major axon tracts and interneuron migration in the forebrain,” Development, vol. 133, no. 11, pp. 2243–2252, 2006.
[26]  T. R. Keeble, M. M. Halford, C. Seaman et al., “The Wnt receptor Ryk is required for Wnt5a-mediated axon guidance on the contralateral side of the corpus callosum,” Journal of Neuroscience, vol. 26, no. 21, pp. 5840–5848, 2006.
[27]  T. Shu, K. M. Valentino, C. Seaman, H. M. Cooper, and L. J. Richards, “Expression of the netrin-1 receptor, deleted in colorectal cancer (DCC), is largely confined to projecting neurons in the developing forebrain,” Journal of Comparative Neurology, vol. 416, no. 2, pp. 201–212, 2000.
[28]  T. Serafini, S. A. Colamarino, E. D. Leonardo et al., “Netrin-1 is required for commissural axon guidance in the developing vertebrate nervous system,” Cell, vol. 87, no. 6, pp. 1001–1014, 1996.
[29]  S. M. Islam, Y. Shinmyo, T. Okafuji et al., “Draxin, a repulsive guidance protein for spinal cord and forebrain commissures,” Science, vol. 323, no. 5912, pp. 388–393, 2009.
[30]  H. Zhao, T. Maruyama, Y. Hattori et al., “A molecular mechanism that regulates medially oriented axonal growth of upper layer neurons in the developing neocortex,” Journal of Comparative Neurology, vol. 519, no. 5, pp. 834–848, 2011.
[31]  D. K. Unni, M. Piper, R. X. Moldrich et al., “Multiple Slits regulate the development of midline glial populations and the corpus callosum,” Developmental Biology, vol. 365, no. 1, pp. 36–49, 2012.
[32]  L. Li, B. I. Hutchins, and K. Kalil, “Wnt5a induces simultaneous cortical axon outgrowth and repulsive axon guidance through distinct signaling mechanisms,” Journal of Neuroscience, vol. 29, no. 18, pp. 5873–5883, 2009.
[33]  L. Li, B. I. Hutchins, and K. Kalil, “Wnt5a induces simultaneous cortical axon outgrowth and repulsive turning through distinct signaling mechanisms,” Science Signaling, vol. 3, no. 147, p. pt2, 2010.
[34]  M. Niquille, S. Garel, F. Mann et al., “Transient neuronal populations are required to guide callosal axons: a role for semaphorin 3C,” PLoS Biology, vol. 7, no. 10, Article ID e1000230, 2009.
[35]  Y. Choe, J. A. Siegenthaler, and S. J. Pleasure, “A cascade of morphogenic signaling initiated by the meninges controls corpus callosum formation,” Neuron, vol. 73, no. 4, pp. 698–712, 2012.
[36]  A. Martínez and E. Soriano, “Functions of ephrin/Eph interactions in the development of the nervous system: emphasis on the hippocampal system,” Brain Research Reviews, vol. 49, no. 2, pp. 211–226, 2005.
[37]  J. O. Bush and P. Soriano, “Ephrin-B1 regulates axon guidance by reverse signaling through a PDZ-dependent mechanism,” Genes and Development, vol. 23, no. 13, pp. 1586–1599, 2009.
[38]  A. Davy and P. Soriano, “Ephrin signaling in vivo: look both ways,” Developmental Dynamics, vol. 232, no. 1, pp. 1–10, 2005.
[39]  C. Lindwall, T. Fothergill, and L. J. Richards, “Commissure formation in the mammalian forebrain,” Current Opinion in Neurobiology, vol. 17, no. 1, pp. 3–14, 2007.
[40]  S. W. Mendes, M. Henkemeyer, and D. J. Liebl, “Multiple Eph receptors and B-class ephrins regulate midline crossing of corpus callosum fibers in the developing mouse forebrain,” Journal of Neuroscience, vol. 26, no. 3, pp. 882–892, 2006.
[41]  C. Plachez and L. J. Richards, “Mechanisms of axon guidance in the developing nervous system,” Current Topics in Developmental Biology, vol. 69, pp. 267–346, 2005.
[42]  S. E. Koester and D. D. M. O'Leary, “Axons of early generated neurons in cingulate cortex pioneer the corpus callosum,” Journal of Neuroscience, vol. 14, no. 11, pp. 6608–6620, 1994.
[43]  B. G. Rash and L. J. Richards, “A role for cingulate pioneering axons in the development of the corpus callosum,” Journal of Comparative Neurology, vol. 434, no. 2, pp. 147–157, 2001.
[44]  L. C. deAzevedo, C. Hedin-Pereira, and R. Lent, “Callosal neurons in the cingulate cortical plate and subplate of human fetuses,” Journal of Comparative Neurology, vol. 386, no. 1, pp. 60–70, 1997.
[45]  M. Piper, C. Plachez, O. Zalucki et al., “Neuropilin 1-Sema signaling regulates crossing of cingulate pioneering axons during development of the corpus callosum,” Cerebral Cortex, vol. 19, supplement 1, pp. i11–i21, 2009.
[46]  H. S. Ozaki and D. Wahlsten, “Timing and origin of the first cortical axons to project through the corpus callosum and the subsequent emergence of callosal projection cells in mouse,” Journal of Comparative Neurology, vol. 400, no. 2, pp. 197–206, 1998.
[47]  S. O. Chan and K. Y. Chung, “Changes in axon arrangement in the retinofugal [correction of retinofungal] pathway of mouse embryos: confocal microscopy study using single- and double-dye label,” Journal of Comparative Neurology, vol. 406, no. 2, pp. 251–262, 1999.
[48]  B. W. Gallarda, D. Bonanomi, D. Müller et al., “Segregation of axial motor and sensory pathways via heterotypic trans-axonal signaling,” Science, vol. 320, no. 5873, pp. 233–236, 2008.
[49]  D. T. Plas, J. E. Lopez, and M. C. Crair, “Pretarget sorting of retinocollicular axons in the mouse,” Journal of Comparative Neurology, vol. 491, no. 4, pp. 305–319, 2005.
[50]  T. Imai, T. Yamazaki, R. Kobayakawa et al., “Pre-Target axon sorting establishes the neural map topography,” Science, vol. 325, no. 5940, pp. 585–590, 2009.
[51]  C. Kudo, I. Ajioka, Y. Hirata, and K. Nakajima, “Expression profiles of EphA3 at both the RNA and protein level in the developing mammalian forebrain,” Journal of Comparative Neurology, vol. 487, no. 3, pp. 255–269, 2005.
[52]  M. Nishikimi, K. Oishi, H. Tabata, K. Torii, and K. Nakajima, “Segregation and pathfinding of callosal axons through EphA3 signaling,” Journal of Neuroscience, vol. 31, no. 45, pp. 16251–16260, 2011.
[53]  H. Tabata and K. Nakajima, “Efficient in utero gene transfer system to the developing mouse brain using electroporation: visualization of neuronal migration in the developing cortex,” Neuroscience, vol. 103, no. 4, pp. 865–872, 2001.
[54]  B. Kn?ll, C. Weinl, A. Nordheim, and F. Bonhoeffer, “Stripe assay to examine axonal guidance and cell migration,” Nature Protocols, vol. 2, no. 5, pp. 1216–1224, 2007.
[55]  T. Maruyama, M. Matsuura, K. Suzuki, and N. Yamamoto, “Cooperative activity of multiple upper layer proteins for thalamocortical axon growth,” Developmental Neurobiology, vol. 68, no. 3, pp. 317–331, 2008.
[56]  G. M. Innocenti, “Growth and reshaping of axons in the establishment of visual callosal connections,” Science, vol. 212, no. 4496, pp. 824–827, 1981.
[57]  G. M. Innocenti and D. J. Price, “Exuberance in the development of cortical networks,” Nature Reviews Neuroscience, vol. 6, no. 12, pp. 955–965, 2005.
[58]  J. H. Leslie and E. Nedivi, “Activity-regulated genes as mediators of neural circuit plasticity,” Progress in Neurobiology, vol. 94, no. 3, pp. 223–237, 2011.
[59]  J. Olavarria and R. C. Van Sluyters, “Callosal connections of the posterior neocortex in normal-eyed, congenitally anophthalmic, and neonatally enucleated mice,” Journal of Comparative Neurology, vol. 230, no. 2, pp. 249–268, 1984.
[60]  G. M. Innocenti, D. O. Frost, and J. Illes, “Maturation of visual callosal connections in visually deprived kittens: a challenging critical period,” Journal of Neuroscience, vol. 5, no. 2, pp. 255–267, 1985.
[61]  M. G. Hanson and L. T. Landmesser, “Normal patterns of spontaneous activity are required for correct motor axon guidance and the expression of specific guidance molecules,” Neuron, vol. 43, no. 5, pp. 687–701, 2004.
[62]  S. Serizawa, K. Miyamichi, H. Takeuchi, Y. Yamagishi, M. Suzuki, and H. Sakano, “A neuronal identity code for the odorant receptor-specific and activity-dependent axon sorting,” Cell, vol. 127, no. 5, pp. 1057–1069, 2006.
[63]  K. Itoh, B. Stevens, M. Schachner, and R. D. Fields, “Regulated expression of the neural cell adhesion molecule L1 by specific patterns of neural impulses,” Science, vol. 270, no. 5240, pp. 1369–1372, 1995.
[64]  V. Conti, C. Marini, S. Gana, J. Sudi, W. B. Dobyns, and R. Guerrini, “Corpus callosum agenesis, severe mental retardation, epilepsy, and dyskinetic quadriparesis due to a novel mutation in the homeodomain of ARX,” American Journal of Medical Genetics, Part A, vol. 155, no. 4, pp. 892–897, 2011.
[65]  G. Friocourt, K. Poirier, S. Raki?, J. G. Parnavelas, and J. Chelly, “The role of ARX in cortical development,” European Journal of Neuroscience, vol. 23, no. 4, pp. 869–876, 2006.
[66]  T. Ren, J. Zhang, C. Plachez, S. Mori, and L. J. Richards, “Diffusion tensor magnetic resonance imaging and tract-tracing analysis of probst bundle structure in netrin1- and DCC-deficient mice,” Journal of Neuroscience, vol. 27, no. 39, pp. 10345–10349, 2007.
[67]  H. S. Ozaki and D. Wahlsten, “Cortical axon trajectories and growth cone morphologies in fetuses of acallosal mouse strains,” Journal of Comparative Neurology, vol. 336, no. 4, pp. 595–604, 1993.
[68]  R. Lent, D. Uziel, M. Baudrimont, and C. Fallet, “Cellular and molecular tunnels surrounding the forebrain commissures of human fetuses,” Journal of Comparative Neurology, vol. 483, no. 4, pp. 375–382, 2005.
[69]  A. L. S. Donahoo and L. J. Richards, “Understanding the mechanisms of callosal development through the use of transgenic mouse models,” Seminars in Pediatric Neurology, vol. 16, no. 3, pp. 127–142, 2009.
[70]  T. Ren, A. Anderson, W. B. Shen et al., “Imaging, anatomical, and molecular analysis of callosal formation in the developing human fetal brain,” Anatomical Record A, vol. 288, no. 2, pp. 191–204, 2006.
[71]  J. K. Gupta and R. J. Lilford, “Assessment and management of fetal agenesis of the corpus callosum,” Prenatal Diagnosis, vol. 15, no. 4, pp. 301–312, 1995.
[72]  H. C. Sauerwein and M. Lassonde, “Cognitive and sensori-motor functioning in the absence of the corpus callosum: neuropsychological studies in callosal agenesis and callosotomized patients,” Behavioural Brain Research, vol. 64, no. 1-2, pp. 229–240, 1994.
[73]  L. K. Paul, B. Schieffer, and W. S. Brown, “Social processing deficits in agenesis of the corpus callosum: narratives from the Thematic Apperception Test,” Archives of Clinical Neuropsychology, vol. 19, no. 2, pp. 215–225, 2004.
[74]  B. Egaas, E. Courchesne, and O. Saitoh, “Reduced size of corpus callosum in autism,” Archives of Neurology, vol. 52, no. 8, pp. 794–801, 1995.
[75]  I. K. Lyoo, G. G. Noam, C. K. Lee, H. K. Lee, B. P. Kennedy, and P. F. Renshaw, “The corpus callosum and lateral ventricles in children with attention-deficit hyperactivity disorder: a brain magnetic resonance imaging study,” Biological Psychiatry, vol. 40, no. 10, pp. 1060–1063, 1996.
[76]  O. Lungu and E. Stip, “Agenesis of corpus callosum and emotional information processing in schizophrenia,” Front Psychiatry, vol. 3, p. 1, 2012.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133