151
|
Lewitus E, Kelava I, Huttner WB. Conical expansion of the outer subventricular zone and the role of neocortical folding in evolution and development. Front Hum Neurosci 2013; 7:424. [PMID: 23914167 PMCID: PMC3729979 DOI: 10.3389/fnhum.2013.00424] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 07/14/2013] [Indexed: 12/01/2022] Open
Abstract
THERE IS A BASIC RULE TO MAMMALIAN NEOCORTICAL EXPANSION as it expands, so does it fold. The degree to which it folds, however, cannot strictly be attributed to its expansion. Across species, cortical volume does not keep pace with cortical surface area, but rather folds appear more rapidly than expected. As a result, larger brains quickly become disproportionately more convoluted than smaller brains. Both the absence (lissencephaly) and presence (gyrencephaly) of cortical folds is observed in all mammalian orders and, while there is likely some phylogenetic signature to the evolutionary appearance of gyri and sulci, there are undoubtedly universal trends to the acquisition of folds in an expanding neocortex. Whether these trends are governed by conical expansion of neocortical germinal zones, the distribution of cortical connectivity, or a combination of growth- and connectivity-driven forces remains an open question. But the importance of cortical folding for evolution of the uniquely mammalian neocortex, as well as for the incidence of neuropathologies in humans, is undisputed. In this hypothesis and theory article, we will summarize the development of cortical folds in the neocortex, consider the relative influence of growth- vs. connectivity-driven forces for the acquisition of cortical folds between and within species, assess the genetic, cell-biological, and mechanistic implications for neocortical expansion, and discuss the significance of these implications for human evolution, development, and disease. We will argue that evolutionary increases in the density of neuron production, achieved via maintenance of a basal proliferative niche in the neocortical germinal zones, drive the conical migration of neurons toward the cortical surface and ultimately lead to the establishment of cortical folds in large-brained mammal species.
Collapse
Affiliation(s)
| | | | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsDresden, Germany
| |
Collapse
|
152
|
Barzilai A. The interrelations between malfunctioning DNA damage response (DDR) and the functionality of the neuro-glio-vascular unit. DNA Repair (Amst) 2013; 12:543-57. [DOI: 10.1016/j.dnarep.2013.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
153
|
Medina FS, Hunt GR, Gray RD, Wild JM, Kubke MF. Perineuronal satellite neuroglia in the telencephalon of New Caledonian crows and other Passeriformes: evidence of satellite glial cells in the central nervous system of healthy birds? PeerJ 2013; 1:e110. [PMID: 23904989 PMCID: PMC3728766 DOI: 10.7717/peerj.110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 07/02/2013] [Indexed: 12/24/2022] Open
Abstract
Glia have been implicated in a variety of functions in the central nervous system, including the control of the neuronal extracellular space, synaptic plasticity and transmission, development and adult neurogenesis. Perineuronal glia forming groups around neurons are associated with both normal and pathological nervous tissue. Recent studies have linked reduction in the number of perineuronal oligodendrocytes in the prefrontal cortex with human schizophrenia and other psychiatric disorders. Therefore, perineuronal glia may play a decisive role in homeostasis and normal activity of the human nervous system. Here we report on the discovery of novel cell clusters in the telencephala of five healthy Passeriforme, one Psittaciform and one Charadriiforme bird species, which we refer to as Perineuronal Glial Clusters (PGCs). The aim of this study is to describe the structure and distribution of the PGCs in a number of avian species. PGCs were identified with the use of standard histological procedures. Heterochromatin masses visible inside the nuclei of these satellite glia suggest that they may correspond to oligodendrocytes. PGCs were found in the brains of nine New Caledonian crows, two Japanese jungle crows, two Australian magpies, two Indian mynah, three zebra finches (all Passeriformes), one Southern lapwing (Charadriiformes) and one monk parakeet (Psittaciformes). Microscopic survey of the brain tissue suggests that the largest PGCs are located in the hyperpallium densocellulare and mesopallium. No clusters were found in brain sections from one Gruiform (purple swamphen), one Strigiform (barn owl), one Trochiliform (green-backed firecrown), one Falconiform (chimango caracara), one Columbiform (pigeon) and one Galliform (chick). Our observations suggest that PGCs in Aves are brain region- and taxon-specific and that the presence of perineuronal glia in healthy human brains and the similar PGCs in avian gray matter is the result of convergent evolution. The discovery of PGCs in the zebra finch is of great importance because this species has the potential to become a robust animal model in which to study the function of neuron-glia interactions in healthy and diseased adult brains.
Collapse
Affiliation(s)
- Felipe S Medina
- School of Psychology, University of Auckland , New Zealand ; Department of Anatomy with Radiology, University of Auckland , New Zealand
| | | | | | | | | |
Collapse
|
154
|
Jensen CJ, Massie A, De Keyser J. Immune players in the CNS: the astrocyte. J Neuroimmune Pharmacol 2013; 8:824-39. [PMID: 23821340 DOI: 10.1007/s11481-013-9480-6] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 05/26/2013] [Indexed: 12/20/2022]
Abstract
In the finely balanced environment of the central nervous system astrocytes, the most numerous cell type, play a role in regulating almost every physiological system. First found to regulate extracellular ions and pH, they have since been shown to regulate neurotransmitter levels, cerebral blood flow and energy metabolism. There is also growing evidence for an essential role of astrocytes in central immunity, which is the topic of this review. In the healthy state, the central nervous system is potently anti-inflammatory but under threat astrocytes readily respond to pathogens and to both sterile and pathogen-induced cell damage. In response, astrocytes take on some of the roles of immune cells, releasing cyto- and chemokines to influence effector cells, modulating the blood-brain barrier and forming glial scars. To date, much of the data supporting a role for astrocytes in immunity have been obtained from in vitro systems; however data from experimental models and clinical samples support the suggestion that astrocytes perform similar roles in more complex environments. This review will discuss some aspects of the role of astrocytes in central nervous system immunity.
Collapse
Affiliation(s)
- Cathy J Jensen
- Department of Neurology, Universitair Ziekenhuis Brussel, Center for Neurosciences, Vrije Universiteit Brussel-VUB, Brussels, Belgium.
| | | | | |
Collapse
|
155
|
Freas CA, Roth TC, LaDage LD, Pravosudov VV. Hippocampal neuron soma size is associated with population differences in winter climate severity in food-caching chickadees. Funct Ecol 2013. [DOI: 10.1111/1365-2435.12125] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Cody A. Freas
- Department of Biology; MS 314, University of Nevada at Reno; Reno Nevada 89557 USA
| | - Timothy C. Roth
- Department of Psychology; Franklin & Marshall College; PO Box 3003 Lancaster Pennsylvania 17604 USA
| | - Lara D. LaDage
- Department of Biology; MS 314, University of Nevada at Reno; Reno Nevada 89557 USA
| | | |
Collapse
|
156
|
Yokel R, Grulke E, MacPhail R. Metal-based nanoparticle interactions with the nervous system: the challenge of brain entry and the risk of retention in the organism. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2013; 5:346-73. [PMID: 23568784 DOI: 10.1002/wnan.1202] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This review of metal-based nanoparticles focuses on factors influencing their distribution into the nervous system, evidence they enter brain parenchyma, and nervous system responses. Gold is emphasized as a model metal-based nanoparticle and for risk assessment in the companion review. The anatomy and physiology of the nervous system, basics of colloid chemistry, and environmental factors that influence what cells see are reviewed to provide background on the biological, physical-chemical, and internal milieu factors that influence nervous system nanoparticle uptake. The results of literature searches reveal little nanoparticle research included the nervous system, which about equally involved in vitro and in vivo methods, and very few human studies. The routes of uptake into the nervous system and mechanisms of nanoparticle uptake by cells are presented with examples. Brain nanoparticle uptake inversely correlates with size. The influence of shape has not been reported. Surface charge has not been clearly shown to affect flux across the blood-brain barrier. There is very little evidence for metal-based nanoparticle distribution into brain parenchyma. Metal-based nanoparticle disruption of the blood-brain barrier and adverse brain changes have been shown, and are more pronounced for spheres than rods. Study concentrations need to be put in exposure contexts. Work with dorsal root ganglion cells and brain cells in vitro show the potential for metal-based nanoparticles to produce toxicity. Interpretation of these results must consider the ability of nanoparticles to distribute across the barriers protecting the nervous system. Effects of the persistence of poorly soluble metal-based nanoparticles are of particular concern.
Collapse
Affiliation(s)
- Robert Yokel
- Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA.
| | | | | |
Collapse
|
157
|
Amiri M, Montaseri G, Bahrami F. A phase plane analysis of neuron-astrocyte interactions. Neural Netw 2013; 44:157-65. [PMID: 23685459 DOI: 10.1016/j.neunet.2013.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 01/24/2013] [Accepted: 03/31/2013] [Indexed: 10/27/2022]
Abstract
Intensive experimental studies have shown that astrocytes are active partners in modulation of synaptic transmission. In the present research, we study neuron-astrocyte signaling using a biologically inspired model of one neuron synapsing one astrocyte. In this model, the firing dynamics of the neuron is described by the Morris-Lecar model and the Ca(2+) dynamics of a single astrocyte explained by a functional model introduced by Postnov and colleagues. Using the coupled neuron-astrocyte model and based on the results of the phase plane analyses, it is demonstrated that the astrocyte is able to activate the silent neuron or change the neuron spiking frequency through bidirectional communication. This suggests that astrocyte feedback signaling is capable of modulating spike transmission frequency by changing neuron spiking frequency. This effect is described by a saddle-node on invariant circle bifurcation in the coupled neuron-astrocyte model. In this way, our results suggest that the neuron-astrocyte crosstalk has a fundamental role in producing diverse neuronal activities and therefore enhances the information processing capabilities of the brain.
Collapse
Affiliation(s)
- Mahmood Amiri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | | | | |
Collapse
|
158
|
Chinea A, Korutcheva E. Intelligence and embodiment: A statistical mechanics approach. Neural Netw 2013; 40:52-72. [DOI: 10.1016/j.neunet.2013.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 01/11/2013] [Accepted: 01/11/2013] [Indexed: 10/27/2022]
|
159
|
Killeen PR, Russell VA, Sergeant JA. A behavioral neuroenergetics theory of ADHD. Neurosci Biobehav Rev 2013; 37:625-57. [PMID: 23454637 DOI: 10.1016/j.neubiorev.2013.02.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 02/02/2013] [Accepted: 02/18/2013] [Indexed: 02/02/2023]
Abstract
Energetic insufficiency in neurons due to inadequate lactate supply is implicated in several neuropathologies, including attention-deficit/hyperactivity disorder (ADHD). By formalizing the mechanism and implications of such constraints on function, the behavioral Neuroenergetics Theory (NeT) predicts the results of many neuropsychological tasks involving individuals with ADHD and kindred dysfunctions, and entails many novel predictions. The associated diffusion model predicts that response times will follow a mixture of Wald distributions from the attentive state, and ex-Wald distributions after attentional lapses. It is inferred from the model that ADHD participants can bring only 75-85% of the neurocognitive energy to bear on tasks, and allocate only about 85% of the cognitive resources of comparison groups. Parameters derived from the model in specific tasks predict performance in other tasks, and in clinical conditions often associated with ADHD. The primary action of therapeutic stimulants is to increase norepinephrine in active regions of the brain. This activates glial adrenoceptors, increasing the release of lactate from astrocytes to fuel depleted neurons. The theory is aligned with other approaches and integrated with more general theories of ADHD. Therapeutic implications are explored.
Collapse
Affiliation(s)
- Peter R Killeen
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, USA.
| | | | | |
Collapse
|
160
|
Abstract
What evolutionary events led to the emergence of human cognition? Although the genetic differences separating modern humans from both non-human primates (for example, chimpanzees) and archaic hominins (Neanderthals and Denisovans) are known, linking human-specific mutations to the cognitive phenotype remains a challenge. One strategy is to focus on human-specific changes at the level of intermediate phenotypes, such as gene expression and metabolism, in conjunction with evolutionary changes in gene regulation involving transcription factors, microRNA and proximal regulatory elements. In this Review we show how this strategy has yielded some of the first hints about the mechanisms of human cognition.
Collapse
|
161
|
Freas C, Bingman K, LaDage L, Pravosudov V. Untangling Elevation-Related Differences in the Hippocampus in Food-Caching Mountain Chickadees: The Effect of a Uniform Captive Environment. BRAIN, BEHAVIOR AND EVOLUTION 2013; 82:199-209. [DOI: 10.1159/000355503] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/03/2013] [Indexed: 11/19/2022]
|
162
|
Maseko BC, Jacobs B, Spocter MA, Sherwood CC, Hof PR, Manger PR. Qualitative and Quantitative Aspects of the Microanatomy of the African Elephant Cerebellar Cortex. BRAIN, BEHAVIOR AND EVOLUTION 2013; 81:40-55. [DOI: 10.1159/000345565] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 09/14/2012] [Indexed: 11/19/2022]
|
163
|
Mack AF, Tiedemann K. Cultures of astroglial cells derived from brain of adult cichlid fish. J Neurosci Methods 2013; 212:269-75. [DOI: 10.1016/j.jneumeth.2012.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 10/22/2012] [Accepted: 11/08/2012] [Indexed: 10/27/2022]
|
164
|
González de la Aleja J, Ramos A, Mato-Abad V, Martínez-Salio A, Hernández-Tamames JA, Molina JA, Hernández-Gallego J, Álvarez-Linera J. Higher Glutamate to Glutamine Ratios in Occipital Regions in Women With Migraine During the Interictal State. Headache 2012; 53:365-75. [DOI: 10.1111/head.12030] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2012] [Indexed: 01/25/2023]
|
165
|
Shulha HP, Crisci JL, Reshetov D, Tushir JS, Cheung I, Bharadwaj R, Chou HJ, Houston IB, Peter CJ, Mitchell AC, Yao WD, Myers RH, Chen JF, Preuss TM, Rogaev EI, Jensen JD, Weng Z, Akbarian S. Human-specific histone methylation signatures at transcription start sites in prefrontal neurons. PLoS Biol 2012; 10:e1001427. [PMID: 23185133 PMCID: PMC3502543 DOI: 10.1371/journal.pbio.1001427] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 10/12/2012] [Indexed: 11/18/2022] Open
Abstract
Cognitive abilities and disorders unique to humans are thought to result from adaptively driven changes in brain transcriptomes, but little is known about the role of cis-regulatory changes affecting transcription start sites (TSS). Here, we mapped in human, chimpanzee, and macaque prefrontal cortex the genome-wide distribution of histone H3 trimethylated at lysine 4 (H3K4me3), an epigenetic mark sharply regulated at TSS, and identified 471 sequences with human-specific enrichment or depletion. Among these were 33 loci selectively methylated in neuronal but not non-neuronal chromatin from children and adults, including TSS at DPP10 (2q14.1), CNTN4 and CHL1 (3p26.3), and other neuropsychiatric susceptibility genes. Regulatory sequences at DPP10 and additional loci carried a strong footprint of hominid adaptation, including elevated nucleotide substitution rates and regulatory motifs absent in other primates (including archaic hominins), with evidence for selective pressures during more recent evolution and adaptive fixations in modern populations. Chromosome conformation capture at two neurodevelopmental disease loci, 2q14.1 and 16p11.2, revealed higher order chromatin structures resulting in physical contact of multiple human-specific H3K4me3 peaks spaced 0.5-1 Mb apart, in conjunction with a novel cis-bound antisense RNA linked to Polycomb repressor proteins and downregulated DPP10 expression. Therefore, coordinated epigenetic regulation via newly derived TSS chromatin could play an important role in the emergence of human-specific gene expression networks in brain that contribute to cognitive functions and neurological disease susceptibility in modern day humans.
Collapse
Affiliation(s)
- Hennady P. Shulha
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jessica L. Crisci
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Denis Reshetov
- Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Moscow, Russian Federation
| | - Jogender S. Tushir
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Iris Cheung
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Rahul Bharadwaj
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Hsin-Jung Chou
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Isaac B. Houston
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Cyril J. Peter
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Amanda C. Mitchell
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Wei-Dong Yao
- New England Primate Center, Southboro, Massachusetts, United States of America
| | - Richard H. Myers
- Department of Neurology, Boston University, Boston, Massachusetts, United States of America
| | - Jiang-fan Chen
- Department of Neurology, Boston University, Boston, Massachusetts, United States of America
| | - Todd M. Preuss
- Yerkes National Primate Research Center/Emory University, Atlanta, Georgia, United States of America
| | - Evgeny I. Rogaev
- Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Moscow, Russian Federation
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Research Center of Mental Health, Russian Academy of Medical Sciences, Moscow, Russian Federation
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Russian Federation
| | - Jeffrey D. Jensen
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Schahram Akbarian
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Departments of Psychiatry and Neuroscience, Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York, United States of America
| |
Collapse
|
166
|
Spocter MA, Hopkins WD, Barks SK, Bianchi S, Hehmeyer AE, Anderson SM, Stimpson CD, Fobbs AJ, Hof PR, Sherwood CC. Neuropil distribution in the cerebral cortex differs between humans and chimpanzees. J Comp Neurol 2012; 520:2917-29. [PMID: 22350926 DOI: 10.1002/cne.23074] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Increased connectivity of high-order association regions in the neocortex has been proposed as a defining feature of human brain evolution. At present, however, there are limited comparative data to examine this claim fully. We tested the hypothesis that the distribution of neuropil across areas of the neocortex of humans differs from that of one of our closest living relatives, the common chimpanzee. The neuropil provides a proxy measure of total connectivity within a local region because it is composed mostly of dendrites, axons, and synapses. Using image analysis techniques, we quantified the neuropil fraction from both hemispheres in six cytoarchitectonically defined regions including frontopolar cortex (area 10), Broca's area (area 45), frontoinsular cortex (area FI), primary motor cortex (area 4), primary auditory cortex (area 41/42), and the planum temporale (area 22). Our results demonstrate that humans exhibit a unique distribution of neuropil in the neocortex compared to chimpanzees. In particular, the human frontopolar cortex and the frontoinsular cortex had a significantly higher neuropil fraction than the other areas. In chimpanzees these prefrontal regions did not display significantly more neuropil, but the primary auditory cortex had a lower neuropil fraction than other areas. Our results support the conclusion that enhanced connectivity in the prefrontal cortex accompanied the evolution of the human brain. These species differences in neuropil distribution may offer insight into the neural basis of human cognition, reflecting enhancement of the integrative capacity of the prefrontal cortex.
Collapse
Affiliation(s)
- Muhammad A Spocter
- Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Hemispheric asymmetry in the fusiform gyrus distinguishes Homo sapiens from chimpanzees. Brain Struct Funct 2012; 218:1391-405. [DOI: 10.1007/s00429-012-0464-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 10/06/2012] [Indexed: 12/24/2022]
|
168
|
Reato D, Cammarota M, Parra LC, Carmignoto G. Computational model of neuron-astrocyte interactions during focal seizure generation. Front Comput Neurosci 2012; 6:81. [PMID: 23091457 PMCID: PMC3467689 DOI: 10.3389/fncom.2012.00081] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 09/21/2012] [Indexed: 11/18/2022] Open
Abstract
Empirical research in the last decade revealed that astrocytes can respond to neurotransmitters with Ca2+ elevations and generate feedback signals to neurons which modulate synaptic transmission and neuronal excitability. This discovery changed our basic understanding of brain function and provided new perspectives for how astrocytes can participate not only to information processing, but also to the genesis of brain disorders, such as epilepsy. Epilepsy is a neurological disorder characterized by recurrent seizures that can arise focally at restricted areas and propagate throughout the brain. Studies in brain slice models suggest that astrocytes contribute to epileptiform activity by increasing neuronal excitability through a Ca2+-dependent release of glutamate. The underlying mechanism remains, however, unclear. In this study, we implemented a parsimonious network model of neurons and astrocytes. The model consists of excitatory and inhibitory neurons described by Izhikevich's neuron dynamics. The experimentally observed Ca2+ change in astrocytes in response to neuronal activity was modeled with linear equations. We considered that glutamate is released from astrocytes above certain intracellular Ca2+ concentrations thus providing a non-linear positive feedback signal to neurons. Propagating seizure-like ictal discharges (IDs) were reliably evoked in our computational model by repeatedly exciting a small area of the network, which replicates experimental results in a slice model of focal ID in entorhinal cortex. We found that the threshold of focal ID generation was lowered when an excitatory feedback-loop between astrocytes and neurons was included. Simulations show that astrocytes can contribute to ID generation by directly affecting the excitatory/inhibitory balance of the neuronal network. Our model can be used to obtain mechanistic insights into the distinct contributions of the different signaling pathways to the generation and propagation of focal IDs.
Collapse
Affiliation(s)
- Davide Reato
- Department of Biomedical Engineering, The City College of the City University of New York New York, NY, USA
| | | | | | | |
Collapse
|
169
|
Mesulam M. The evolving landscape of human cortical connectivity: facts and inferences. Neuroimage 2012; 62:2182-9. [PMID: 22209814 PMCID: PMC3321392 DOI: 10.1016/j.neuroimage.2011.12.033] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 11/11/2011] [Accepted: 12/13/2011] [Indexed: 10/14/2022] Open
Abstract
Human cognitive brain mapping is at a crossroads. On the one hand, it can access a rich data set of synaptic connectivity in the cerebral cortex of the monkey, an animal that lacks many of the complicated behaviors of interest. On the other hand, it is rapidly amassing an even richer data set on the functional map of the human cerebral cortex, but with relatively little hard data on underlying structural connectivity. This second point tends to be blurred in the current literature because of the multiple ways in which the term 'connection' is used in the context of the human brain. In some instances the term is used at a conceptual level, to designate a pathway that should be there if the behavior is to be performed. In other instances, it refers to the computational demonstration of a functional relationship, the structural basis of which is not necessarily known. A third usage is based on connections that are known to exist in the monkey and that are inferred to also exist in the human. The fourth and most direct usage involves connections structurally proven to exist in the human. These four usages have been invoked interchangeably to propose connectivistic mechanisms of human cognitive function. To enlarge the currently limited data set on structural connectivity is of considerable importance for conducting biologically more valid explorations of large-scale neurocognitive networks. This challenging goal will require histological laboratory investigations of the human brain to resume their former prominence and to play an increasingly more substantial role in brain mapping research.
Collapse
|
170
|
Elevation-related differences in memory and the hippocampus in mountain chickadees, Poecile gambeli. Anim Behav 2012. [DOI: 10.1016/j.anbehav.2012.04.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
171
|
García-Amado M, Prensa L. Stereological analysis of neuron, glial and endothelial cell numbers in the human amygdaloid complex. PLoS One 2012; 7:e38692. [PMID: 22719923 PMCID: PMC3374818 DOI: 10.1371/journal.pone.0038692] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 05/09/2012] [Indexed: 02/02/2023] Open
Abstract
Cell number alterations in the amygdaloid complex (AC) might coincide with neurological and psychiatric pathologies with anxiety imbalances as well as with changes in brain functionality during aging. This stereological study focused on estimating, in samples from 7 control individuals aged 20 to 75 years old, the number and density of neurons, glia and endothelial cells in the entire AC and in its 5 nuclear groups (including the basolateral (BL), corticomedial and central groups), 5 nuclei and 13 nuclear subdivisions. The volume and total cell number in these territories were determined on Nissl-stained sections with the Cavalieri principle and the optical fractionator. The AC mean volume was 956 mm3 and mean cell numbers (x106) were: 15.3 neurons, 60 glial cells and 16.8 endothelial cells. The numbers of endothelial cells and neurons were similar in each AC region and were one fourth the number of glial cells. Analysis of the influence of the individuals’ age at death on volume, cell number and density in each of these 24 AC regions suggested that aging does not affect regional size or the amount of glial cells, but that neuron and endothelial cell numbers respectively tended to decrease and increase in territories such as AC or BL. These accurate stereological measures of volume and total cell numbers and densities in the AC of control individuals could serve as appropriate reference values to evaluate subtle alterations in this structure in pathological conditions.
Collapse
Affiliation(s)
- María García-Amado
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lucía Prensa
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
172
|
Sherwood CC, Bauernfeind AL, Bianchi S, Raghanti MA, Hof PR. Human brain evolution writ large and small. PROGRESS IN BRAIN RESEARCH 2012; 195:237-54. [PMID: 22230630 DOI: 10.1016/b978-0-444-53860-4.00011-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human evolution was marked by an extraordinary increase in total brain size relative to body size. While it is certain that increased encephalization is an important factor contributing to the origin of our species-specific cognitive abilities, it is difficult to disentangle which aspects of human neural structure and function are correlated by-products of brain size expansion from those that are specifically related to particular psychological specializations, such as language and enhanced "mentalizing" abilities. In this chapter, we review evidence from allometric scaling studies demonstrating that much of human neocortical organization can be understood as a product of brain enlargement. Defining extra-allometric specializations in humans is often hampered by a severe lack of comparative data from the same neuroanatomical variables across a broad range of primates. When possible, we highlight evidence for features of human neocortical architecture and function that cannot be easily explained as correlates of brain size and, hence, might be more directly associated with the evolution of uniquely human cognitive capacities.
Collapse
Affiliation(s)
- Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC, USA.
| | | | | | | | | |
Collapse
|
173
|
Roberts D, Killiany R, Rosene D. Neuron numbers in the hypothalamus of the normal aging rhesus monkey: stability across the adult lifespan and between the sexes. J Comp Neurol 2012; 520:1181-97. [PMID: 21935936 PMCID: PMC4278435 DOI: 10.1002/cne.22761] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Normal aging is accompanied by changes in hypothalamic functions including autonomic and endocrine functions and circadian rhythms. The rhesus monkey provides an excellent model of normal aging without the potential confounds of incipient Alzheimer's disease inherent in human populations. This study examined the hypothalamus of 51 rhesus monkeys (23 male, 18 female, 6.5-31 years old) using design-based stereology to obtain unbiased estimates of neuron and glia numbers and the Cavalieri method to estimate volumes for eight reference spaces: total unilateral hypothalamus, suprachiasmatic nucleus (SCN), supraoptic nucleus (SON), paraventricular nucleus (PVN), dorsomedial nucleus (DM), ventromedial nucleus (VM), medial mammillary nucleus (MMN), and lateral hypothalamic area (LHA). The results demonstrated no age-related difference in neuron number, glia number, or volume in any area in either sex except the PVN of male monkeys, which showed a significant increase in both neuron and glia numbers with age. Comparison of males and females for sexual dimorphisms revealed no significant differences in neuron number. However, males had more glia overall as well as in the SCN, DM, and LHA and had a larger hypothalamic volume overall and in the SCN, SON, VM, DM, and MMN. These results demonstrate that hypothalamic neuron loss cannot account for age-related deficits in hypothalamic function and provides further evidence of the absence of neurodegeneration and cell death in the normal aging rhesus monkey.
Collapse
Affiliation(s)
- D.E. Roberts
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts 02127
| | - R.J. Killiany
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts 02127
| | - D.L. Rosene
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts 02127
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
174
|
Lewitus E, Hof PR, Sherwood CC. Phylogenetic comparison of neuron and glia densities in the primary visual cortex and hippocampus of carnivores and primates. Evolution 2012; 66:2551-63. [PMID: 22834752 DOI: 10.1111/j.1558-5646.2012.01601.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
A major focus of comparative neuroanatomy has been on whether the mammalian brain evolves in a concerted or a mosaic fashion. Workers have examined variation in the volume of different brain regions across taxa to test the degree to which selection is constrained by the timing of events in neural development. Whether a conserved neurogenetic program in the mammalian brain constrains the distribution of different cell types, however, has not yet been investigated. Here we tested for evidence of evolutionary constraints on the densities of different cell types in the primary visual cortex (V1) and the hippocampus in 37 primate and 21 carnivore species. Cellular densities in V1 and the hippocampus scale isometrically with respect to one another in carnivores, as predicted by the concerted evolution hypothesis. In primates, however, cellular distributions in the hippocampus and primary visual cortex show no correlations, which supports the hypothesis of mosaic brain evolution. We therefore provide evidence for the presence of constraints controlling the adult densities of different cell types in disparate regions of the mammalian brain, but also for specializations along the primate lineage. We propose that adaptations to modularity at the cellular level may carry a deep phylogenetic signal.
Collapse
Affiliation(s)
- Eric Lewitus
- Department of Anthropology, University College London, London WC1H 0BW, United Kingdom.
| | | | | |
Collapse
|
175
|
Schneider E, Mayer S, El Hajj N, Jensen LR, Kuss AW, Zischler H, Kondova I, Bontrop RE, Navarro B, Fuchs E, Zechner U, Haaf T. Methylation and expression analyses of the 7q autism susceptibility locus genes MEST , COPG2, and TSGA14 in human and anthropoid primate cortices. Cytogenet Genome Res 2012; 136:278-87. [PMID: 22456293 DOI: 10.1159/000337298] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2012] [Indexed: 12/11/2022] Open
Abstract
The autism susceptibility locus on human chromosome 7q32 contains the maternally imprinted MEST and the non-imprinted COPG2 and TSGA14 genes. Autism is a disorder of the 'social brain' that has been proposed to be due to an overbalance of paternally expressed genes. To study regulation of the 7q32 locus during anthropoid primate evolution, we analyzed the methylation and expression patterns of MEST, COPG2, and TSGA14 in human, chimpanzee, Old World monkey (baboon and rhesus macaque), and New World monkey (marmoset) cortices. In all human and anthropoid primate cortices, the MEST promoter was hemimethylated, as expected for a differentially methylated imprinting control region, whereas the COPG2 and TSGA14 promoters were completely demethylated, typical for transcriptionally active non-imprinted genes. The MEST gene also showed comparable mRNA expression levels in all analyzed species. In contrast, COPG2 expression was downregulated in the human cortex compared to chimpanzee, Old and New World monkeys. TSGA14 either showed no differential regulation in the human brain compared to chimpanzee and marmoset or a slight upregulation compared to baboon. The human-specific downregulation supports a role for COPG2 in the development of a 'social brain'. Promoter methylation patterns appear to be more stable during evolution than gene expression patterns, suggesting that other mechanisms may be more important for inter-primate differences in gene expression.
Collapse
Affiliation(s)
- E Schneider
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Whitehead C. Why the behavioural sciences need the concept of the culture-ready brain. ANTHROPOLOGICAL THEORY 2012. [DOI: 10.1177/1463499612436464] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
From the conceptual gulf dividing social from biological anthropology this paper infers an ideological problem affecting science as a whole. Cultural biases have tended to inhibit or subvert appropriate theorizing and research into unique aspects of the human mind, brain and behaviour. To resolve this problem I suggest that we need a systematic anthropological critique of ‘collective deceptions’ affecting western science, and greater anthropological collaboration with neuroscience and other disciplines. I discuss recent imaging studies which may contribute to a better understanding of the culture-ready brain. Taken in conjunction with fossil and archaeological data, the findings seem more consistent with a ‘play and display’ hypothesis of hominid brain expansion than with current cognocentric hypotheses, suggesting new directions for research. Such research, I argue, could assist integration between behavioural disciplines.
Collapse
|
177
|
Venkateswaran N, Sekhar S, Thirupatchur Sanjayasarathy T, Krishnan SN, Kabaleeswaran DK, Ramanathan S, Narayanasamy N, Jagathrakshakan SS, Vignesh SR. Energetics based spike generation of a single neuron: simulation results and analysis. FRONTIERS IN NEUROENERGETICS 2012; 4:2. [PMID: 22347180 PMCID: PMC3269776 DOI: 10.3389/fnene.2012.00002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 01/12/2012] [Indexed: 11/13/2022]
Abstract
Existing current based models that capture spike activity, though useful in studying information processing capabilities of neurons, fail to throw light on their internal functioning. It is imperative to develop a model that captures the spike train of a neuron as a function of its intracellular parameters for non-invasive diagnosis of diseased neurons. This is the first ever article to present such an integrated model that quantifies the inter-dependency between spike activity and intracellular energetics. The generated spike trains from our integrated model will throw greater light on the intracellular energetics than existing current models. Now, an abnormality in the spike of a diseased neuron can be linked and hence effectively analyzed at the energetics level. The spectral analysis of the generated spike trains in a time–frequency domain will help identify abnormalities in the internals of a neuron. As a case study, the parameters of our model are tuned for Alzheimer’s disease and its resultant spike trains are studied and presented. This massive initiative ultimately aims to encompass the entire molecular signaling pathways of the neuronal bioenergetics linking it to the voltage spike initiation and propagation; due to the lack of experimental data quantifying the inter dependencies among the parameters, the model at this stage adopts a particular level of functionality and is shown as an approach to study and perform disease modeling at the spike train and the mitochondrial bioenergetics level.
Collapse
|
178
|
Pierron D, Wildman DE, Hüttemann M, Letellier T, Grossman LI. Evolution of the couple cytochrome c and cytochrome c oxidase in primates. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 748:185-213. [PMID: 22729859 DOI: 10.1007/978-1-4614-3573-0_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondrial energy metabolism has been affected by a broad set of ancient and recent evolutionary events. The oldest example is the endosymbiosis theory that led to mitochondria and a recently proposed example is adaptation to cold climate by anatomically modern human lineages. Mitochondrial energy metabolism has also been associated with an important area in anthropology and evolutionary biology, brain enlargement in human evolution. Indeed, several studies have pointed to the need for a major metabolic rearrangement to supply a sufficient amount of energy for brain development in primates.The genes encoding for the coupled cytochrome c (Cyt c) and cytochrome c oxidase (COX, complex IV, EC 1.9.3.1) seem to have an exceptional pattern of evolution in the anthropoid lineage. It has been proposed that this evolution was linked to the rearrangement of energy metabolism needed for brain enlargement. This hypothesis is reinforced by the fact that the COX enzyme was proposed to have a large role in control of the respiratory chain and thereby global energy production.After summarizing major events that occurred during the evolution of COX and cytochrome c on the primate lineage, we review the different evolutionary forces that could have influenced primate COX evolution and discuss the probable causes and consequences of this evolution. Finally, we discuss and review the co-occurring primate phenotypic evolution.
Collapse
Affiliation(s)
- Denis Pierron
- Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
179
|
Bastir M, Rosas A, Gunz P, Peña-Melian A, Manzi G, Harvati K, Kruszynski R, Stringer C, Hublin JJ. Evolution of the base of the brain in highly encephalized human species. Nat Commun 2011; 2:588. [PMID: 22158443 DOI: 10.1038/ncomms1593] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 11/14/2011] [Indexed: 01/14/2023] Open
Abstract
The increase of brain size relative to body size-encephalization-is intimately linked with human evolution. However, two genetically different evolutionary lineages, Neanderthals and modern humans, have produced similarly large-brained human species. Thus, understanding human brain evolution should include research into specific cerebral reorganization, possibly reflected by brain shape changes. Here we exploit developmental integration between the brain and its underlying skeletal base to test hypotheses about brain evolution in Homo. Three-dimensional geometric morphometric analyses of endobasicranial shape reveal previously undocumented details of evolutionary changes in Homo sapiens. Larger olfactory bulbs, relatively wider orbitofrontal cortex, relatively increased and forward projecting temporal lobe poles appear unique to modern humans. Such brain reorganization, beside physical consequences for overall skull shape, might have contributed to the evolution of H. sapiens' learning and social capacities, in which higher olfactory functions and its cognitive, neurological behavioral implications could have been hitherto underestimated factors.
Collapse
Affiliation(s)
- Markus Bastir
- Paleoanthropology Group, Department of Paleobiology, Museo Nacional de Ciencias Naturales, CSIC. J. G. Abascal 2, 28006 Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Stenovec M, Milošević M, Petrušić V, Potokar M, Stević Z, Prebil M, Kreft M, Trkov S, Andjus PR, Zorec R. Amyotrophic lateral sclerosis immunoglobulins G enhance the mobility of Lysotracker-labelled vesicles in cultured rat astrocytes. Acta Physiol (Oxf) 2011; 203:457-71. [PMID: 21726417 DOI: 10.1111/j.1748-1716.2011.02337.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIM We examined the effect of purified immunoglobulins G (IgG) from patients with amyotrophic lateral sclerosis (ALS) on the mobility and exocytotic release from Lysotracker-stained vesicles in cultured rat astrocytes. METHODS Time-lapse confocal images were acquired, and vesicle mobility was analysed before and after the application of ALS IgG. The vesicle counts were obtained to assess cargo exocytosis from stained organelles. RESULTS At rest, when mobility was monitored for 2 min in bath with Ca(2+), two vesicle populations were discovered: (1) non-mobile vesicles (6.1%) with total track length (TL) < 1 μm, averaging at 0.33 ± 0.01 μm (n = 1305) and (2) mobile vesicles (93.9%) with TL > 1 μm, averaging at 3.03 ± 0.01 μm (n = 20,200). ALS IgG (0.1 mg mL(-1)) from 12 of 13 patients increased the TL of mobile vesicles by approx. 24% and maximal displacement (MD) by approx. 26% within 4 min, while the IgG from control group did not alter the vesicle mobility. The mobility enhancement by ALS IgG was reduced in extracellular solution devoid of Ca(2+), indicating that ALS IgG vesicle mobility enhancement involves changes in Ca(2+) homeostasis. To examine whether enhanced mobility relates to elevated Ca(2+) activity, cells were stimulated by 1 mm ATP, a cytosolic Ca(2+) increasing agent, in the presence (2 mm) and in the absence of extracellular Ca(2+). ATP stimulation triggered an increase in TL by approx. 7% and 12% and a decrease in MD by approx. 11% and 1%, within 4 min respectively. Interestingly, none of the stimuli triggered the release of vesicle cargo. CONCLUSION Amyotrophic lateral sclerosis-IgG-enhanced vesicle mobility in astrocytes engages changes in calcium homeostasis.
Collapse
Affiliation(s)
- M Stenovec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Medical Faculty, Institute of Pathophysiology, University of Ljubljana, Slovenia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Chareyron LJ, Lavenex PB, Amaral DG, Lavenex P. Stereological analysis of the rat and monkey amygdala. J Comp Neurol 2011; 519:3218-39. [PMID: 21618234 PMCID: PMC4342351 DOI: 10.1002/cne.22677] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The amygdala is part of a neural network that contributes to the regulation of emotional behaviors. Rodents, especially rats, are used extensively as model organisms to decipher the functions of specific amygdala nuclei, in particular in relation to fear and emotional learning. Analysis of the role of the nonhuman primate amygdala in these functions has lagged work in the rodent but provides evidence for conservation of basic functions across species. Here we provide quantitative information regarding the morphological characteristics of the main amygdala nuclei in rats and monkeys, including neuron and glial cell numbers, neuronal soma size, and individual nuclei volumes. The volumes of the lateral, basal, and accessory basal nuclei were, respectively, 32, 39, and 39 times larger in monkeys than in rats. In contrast, the central and medial nuclei were only 8 and 4 times larger in monkeys than in rats. The numbers of neurons in the lateral, basal, and accessory basal nuclei were 14, 11, and 16 times greater in monkeys than in rats, whereas the numbers of neurons in the central and medial nuclei were only 2.3 and 1.5 times greater in monkeys than in rats. Neuron density was between 2.4 and 3.7 times lower in monkeys than in rats, whereas glial density was only between 1.1 and 1.7 times lower in monkeys than in rats. We compare our data in rats and monkeys with those previously published in humans and discuss the theoretical and functional implications that derive from our quantitative structural findings.
Collapse
Affiliation(s)
- Loïc J. Chareyron
- Laboratory of Brain and Cognitive Development, Department of Medicine, University of Fribourg, Switzerland
| | - Pamela Banta Lavenex
- Laboratory of Brain and Cognitive Development, Department of Medicine, University of Fribourg, Switzerland
| | - David G. Amaral
- Department of Psychiatry and Behavioral Sciences, Center for Neuroscience, California National Primate Research Center and the M.I.N.D. Institute, UC Davis, Davis, California, USA
| | - Pierre Lavenex
- Laboratory of Brain and Cognitive Development, Department of Medicine, University of Fribourg, Switzerland
| |
Collapse
|
182
|
Lewitus E, Sherwood CC, Hof PR. Cellular signatures in the primary visual cortex of phylogeny and placentation. Brain Struct Funct 2011; 217:531-47. [PMID: 21863312 DOI: 10.1007/s00429-011-0338-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Accepted: 07/20/2011] [Indexed: 12/20/2022]
Abstract
The long-held view that brain size can be used as an index of general functional capacity across mammals is in conflict with increasing evidence for phyletic differences in cellular organization. Furthermore, it is poorly understood how the internal cellular organization of the brain covaries with overall brain size variation. Using design-based stereology, we quantified glial cell and neuronal densities in the primary visual cortex of 71 mammalian species (spanning 11 orders) to test how those cellular densities are influenced by phylogeny, behavior, environment, and anatomy. We further tested cellular densities against mode of placentation to determine whether a relationship may exist. We provide evidence for cellular signatures of phylogenetic divergence from the mammalian trend in primates and carnivores, as well as considerably divergent scaling patterns between the primate suborders, Strepsirrhini and Haplorrhini, that likely originated at the anthropoid stem. Finally, we show that cellular densities in the mammalian cortex relate to the variability of maternal resources to the fetus in a species.
Collapse
Affiliation(s)
- Eric Lewitus
- Department of Anthropology, University College London, London, WC1H 0BW, UK.
| | | | | |
Collapse
|
183
|
Barzilai A. The neuro-glial-vascular interrelations in genomic instability symptoms. Mech Ageing Dev 2011; 132:395-404. [PMID: 21689674 DOI: 10.1016/j.mad.2011.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 05/25/2011] [Accepted: 06/01/2011] [Indexed: 12/14/2022]
Abstract
A hallmark of neurodegenerative diseases is impairment of certain aspects of "brain functionality", which is defined as the total input and output of the brain's neural circuits and networks. A given neurodegenerative disorder is characterized by affected network organization and topology, cell numbers, cellular functionality, and the interactions between neural circuits. Neuroscientists generally view neurodegenerative disorders as diseases of neuronal cells; however, recent advances suggest a role for glial cells and an impaired vascular system in the etiology of certain neurodegenerative diseases. It is now clear that brain pathology is, to a very great extent, pathology of neurons, glia and the vascular system as these determine the degree of neuronal death as well as the outcome and scale of the neurological deficit. This review article is focused on the intricate interrelations among neurons, glia, the vascular system, neuronal cells, and the DNA damage response. Here I describe various aspects of neural and glial cell fate and the vascular system in genomic instability disorders including ataxia telangiectasia (A-T) and Nijmegen breakage syndrome.
Collapse
Affiliation(s)
- Ari Barzilai
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
184
|
Abstract
Human evolution is characterized by the rapid expansion of brain size and drastic increase in cognitive capabilities. It has long been suggested that these changes were accompanied by modifications of brain metabolism. Indeed, human-specific changes on gene expression or amino acid sequence were reported for a number of metabolic genes, but actual metabolite measurements in humans and apes have remained scarce. Here, we investigate concentrations of more than 100 metabolites in the prefrontal and cerebellar cortex in 49 humans, 11 chimpanzees, and 45 rhesus macaques of different ages using gas chromatography-mass spectrometry (GC-MS). We show that the brain metabolome undergoes substantial changes, both ontogenetically and evolutionarily: 88% of detected metabolites show significant concentration changes with age, whereas 77% of these metabolic changes differ significantly among species. Although overall metabolic divergence reflects phylogenetic relationships among species, we found a fourfold acceleration of metabolic changes in prefrontal cortex compared with cerebellum in the human lineage. These human-specific metabolic changes are paralleled by changes in expression patterns of the corresponding enzymes, and affect pathways involved in synaptic transmission, memory, and learning.
Collapse
|
185
|
Flinn MV, Nepomnaschy PA, Muehlenbein MP, Ponzi D. Evolutionary functions of early social modulation of hypothalamic-pituitary-adrenal axis development in humans. Neurosci Biobehav Rev 2011; 35:1611-29. [PMID: 21251923 DOI: 10.1016/j.neubiorev.2011.01.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 12/17/2010] [Accepted: 01/05/2011] [Indexed: 02/07/2023]
Abstract
The hypothalamic-pituitary-adrenal axis (HPAA) is highly responsive to social challenges. Because stress hormones can have negative developmental and health consequences, this presents an evolutionary paradox: Why would natural selection have favored mechanisms that elevate stress hormone levels in response to psychosocial stimuli? Here we review the hypothesis that large brains, an extended childhood and intensive family care in humans are adaptations resulting from selective forces exerted by the increasingly complex and dynamic social and cultural environment that co-evolved with these traits. Variations in the modulation of stress responses mediated by specific HPAA characteristics (e.g., baseline cortisol levels, and changes in cortisol levels in response to challenges) are viewed as phenotypically plastic, ontogenetic responses to specific environmental signals. From this perspective, we discuss relations between physiological stress responses and life history trajectories, particularly the development of social competencies. We present brief summaries of data on hormones, indicators of morbidity and social environments from our long-term, naturalistic studies in both Guatemala and Dominica. Results indicate that difficult family environments and traumatic social events are associated with temporal elevations of cortisol, suppressed reproductive functioning and elevated morbidity. The long-term effects of traumatic early experiences on cortisol profiles are complex and indicate domain-specific effects, with normal recovery from physical stressors, but some heightened response to negative-affect social challenges. We consider these results to be consistent with the hypothesis that developmental programming of the HPAA and other neuroendocrine systems associated with stress responses may facilitate cognitive targeting of salient social challenges in specific environments.
Collapse
Affiliation(s)
- Mark V Flinn
- Department of Anthropology, University of Missouri, 107 Swallow Hall, Columbia, MO 65211, USA.
| | | | | | | |
Collapse
|
186
|
Mashanov VS, Zueva OR, Garcia-Arraras JE. Organization of glial cells in the adult sea cucumber central nervous system. Glia 2010; 58:1581-93. [PMID: 20578040 DOI: 10.1002/glia.21031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The nervous system of echinoderms has long been considered too unique to be directly comparable to the nervous system of other Deuterostomia. Using two novel monoclonal antibodies in combination with epifluorescence, confocal, and electron microscopy, we demonstrate here that the central nervous system of the sea cucumber Holothuria glaberrima possesses a major non-neuronal cell type, which shares striking similarities with the radial glia of chordates. The basic features in common include (a) an elongated shape, (b) long radial processes, (c) short lateral protrusions branching off the main processes and penetrating into the surrounding neuropile, (d) prominent orderly oriented bundles of intermediate filaments, and (e) ability to produce Reissner's substance. Radial glia account for the majority of glia cells in echinoderms and constitutes more than half of the total cell population in the radial nerve cord and about 45% in the circumoral nerve ring. The difference in glia cell number between those regions is significant, suggesting structural specialization within the seemingly simple echinoderm nervous system. Both cell death and proliferation are seen under normal physiological conditions. Although both glia and neurons undergo apoptosis, most of the mitotic cells are identified as radial glia, indicating a key role of this cell type in cell turnover in the nervous system. A hypothesis is proposed that the radial glia could be an ancestral feature of the deuterostome nervous system, and the origin of this cell type might have predated the diversification of the Chordata and Ambulacraria lineages.
Collapse
Affiliation(s)
- Vladimir S Mashanov
- Department of Biology, University of Puerto Rico, Rio Piedras, PR 00936-8377, Puerto Rico.
| | | | | |
Collapse
|
187
|
Semendeferi K, Teffer K, Buxhoeveden DP, Park MS, Bludau S, Amunts K, Travis K, Buckwalter J. Spatial organization of neurons in the frontal pole sets humans apart from great apes. Cereb Cortex 2010; 21:1485-97. [PMID: 21098620 DOI: 10.1093/cercor/bhq191] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Few morphological differences have been identified so far that distinguish the human brain from the brains of our closest relatives, the apes. Comparative analyses of the spatial organization of cortical neurons, including minicolumns, can aid our understanding of the functionally relevant aspects of microcircuitry. We measured horizontal spacing distance and gray-level ratio in layer III of 4 regions of human and ape cortex in all 6 living hominoid species: frontal pole (Brodmann area [BA] 10), and primary motor (BA 4), primary somatosensory (BA 3), and primary visual cortex (BA 17). Our results identified significant differences between humans and apes in the frontal pole (BA 10). Within the human brain, there were also significant differences between the frontal pole and 2 of the 3 regions studied (BA 3 and BA 17). Differences between BA 10 and BA 4 were present but did not reach significance. These findings in combination with earlier findings on BA 44 and BA 45 suggest that human brain evolution was likely characterized by an increase in the number and width of minicolumns and the space available for interconnectivity between neurons in the frontal lobe, especially the prefrontal cortex.
Collapse
Affiliation(s)
- Katerina Semendeferi
- Department of Anthropology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
188
|
Verkhratsky A. Physiology of neuronal–glial networking. Neurochem Int 2010; 57:332-43. [DOI: 10.1016/j.neuint.2010.02.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 01/05/2010] [Accepted: 02/01/2010] [Indexed: 10/19/2022]
|
189
|
|
190
|
Rappold PM, Tieu K. Astrocytes and therapeutics for Parkinson's disease. Neurotherapeutics 2010; 7:413-23. [PMID: 20880505 PMCID: PMC2948546 DOI: 10.1016/j.nurt.2010.07.001] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/15/2010] [Accepted: 07/01/2010] [Indexed: 12/12/2022] Open
Abstract
Astrocytes play direct, active, and critical roles in mediating neuronal survival and function in various neurodegenerative disorders. This role of astrocytes is well illustrated in amyotrophic lateral sclerosis (ALS), in which the removal of glutamate from the extracellular space by astrocytes confers neuroprotection, whereas astrocytic release of soluble toxic molecules promotes neurodegeneration. In recent years, this context-dependent dual role of astrocytes has also been documented in experimental models of Parkinson's disease. The present review addresses these studies and some potential mechanisms by which astrocytes may influence the neurodegenerative processes in Parkinson's disease, and in particular examines how astrocytes confer neuroprotection either through the removal of toxic molecules from the extracellular space or through the release of trophic factors and antioxidant molecules. In contrast, under pathological conditions, astrocytes release proinflammatory cytokines and other toxic molecules that are detrimental to dopaminergic neurons. These emerging roles of astrocytes in the pathogenesis of Parkinson's disease constitute an exciting development with promising novel therapeutic targets.
Collapse
Affiliation(s)
- Phillip M. Rappold
- grid.16416.340000000419369174Department of Neurology in the Center for Translational Neuromedicine, University of Rochester School of Medicine, 575 Elmwood Avenue, Box 645, 14642 Rochester, NY
| | - Kim Tieu
- grid.16416.340000000419369174Department of Neurology in the Center for Translational Neuromedicine, University of Rochester School of Medicine, 575 Elmwood Avenue, Box 645, 14642 Rochester, NY
| |
Collapse
|
191
|
Jiang Y, Matevossian A, Guo Y, Akbarian S. Setdb1-mediated histone H3K9 hypermethylation in neurons worsens the neurological phenotype of Mecp2-deficient mice. Neuropharmacology 2010; 60:1088-97. [PMID: 20869373 DOI: 10.1016/j.neuropharm.2010.09.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 09/03/2010] [Accepted: 09/16/2010] [Indexed: 12/26/2022]
Abstract
Rett syndrome (RTT, OMIM # 312750), a neurodevelopmental disorder of early childhood, is primarily caused by mutations in the gene encoding methyl-CpG-binding protein 2 (MECP2). Various molecular functions have been ascribed to MECP2, including the regulation of histone modifications associated with repressive chromatin remodeling, but the role of these mechanisms for the pathophysiology of RTT remains unclear. Here, we explore whether or not neuronal expression of the histone H3-lysine 9 specific methyl-transferase, Setdb1 (Set domain, bifurcated 1)/Eset/Kmt1e, which is normally present only at low levels in differentiated neurons, rescues the RTT-like phenotype of Mecp2-deficient mice. A myc-tagged Setdb1 cDNA was expressed through the tau locus for ubiquitous expression in CNS neurons, or under control of the calcium/calmodulin-dependent protein kinase II (CK) promoter to selectively target postmitotic neurons in forebrain. However, the CK-Setdb1 transgene lead to an enhanced neurological deficit, and the tauSetdb1 allele further shortened life span of mice with a brain-wide deletion of Mecp2 during prenatal development. In contrast, no neurological deficits or premature death was observed in CK-Setdb1 and tauSetdb1 mice expressing wildtype Mecp2. However, levels of trimethylated H3K9 at pericentromeric repeats were fully maintained in differentiated neurons from symptomatic Mecp2 null mutant mice. Based on these results, we draw two conclusions: First, neuronal chromatin in RTT brain is not affected by a generalized deficit in H3K9 trimethylation. Second, artificial up-regulation of this repressive chromatin mark via Setdb1 gene delivery specifically to neurons is harmful for the Mecp2-deficient brain. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- Yan Jiang
- Brudnick Neuropsychiatric Research Institute, Department of Psychiatry, University of Massachusetts Medical School, 303 Belmont Street, Worcester, MA 01604, USA
| | | | | | | |
Collapse
|
192
|
Verkhratsky A, Parpura V, Rodríguez JJ. Where the thoughts dwell: the physiology of neuronal-glial "diffuse neural net". ACTA ACUST UNITED AC 2010; 66:133-51. [PMID: 20546785 DOI: 10.1016/j.brainresrev.2010.05.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 05/11/2010] [Accepted: 05/17/2010] [Indexed: 02/07/2023]
Abstract
The mechanisms underlying the production of thoughts by exceedingly complex cellular networks that construct the human brain constitute the most challenging problem of natural sciences. Our understanding of the brain function is very much shaped by the neuronal doctrine that assumes that neuronal networks represent the only substrate for cognition. These neuronal networks however are embedded into much larger and probably more complex network formed by neuroglia. The latter, although being electrically silent, employ many different mechanisms for intercellular signalling. It appears that astrocytes can control synaptic networks and in such a capacity they may represent an integral component of the computational power of the brain rather than being just brain "connective tissue". The fundamental question of whether neuroglia is involved in cognition and information processing remains, however, open. Indeed, a remarkable increase in the number of glial cells that distinguishes the human brain can be simply a result of exceedingly high specialisation of the neuronal networks, which delegated all matters of survival and maintenance to the neuroglia. At the same time potential power of analogue processing offered by internally connected glial networks may represent the alternative mechanism involved in cognition.
Collapse
|
193
|
Gandrakota R, Chakravarthy VS, Pradhan RK. A Model of Indispensability of a Large Glial Layer in Cerebrovascular Circulation. Neural Comput 2010; 22:949-68. [DOI: 10.1162/neco.2009.01-09-945] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
We formulate the problem of oxygen delivery to neural tissue as a problem of association. Input to a pool of neurons in one brain area must be matched in space and time with metabolic inputs from the vascular network via the glial network. We thus have a model in which neural, glial, and vascular layers are connected bidirectionally, in that order. Connections between neuro-glial and glial-vascular stages are trained by an unsupervised learning mechanism such that input to the neural layer is sustained by the precisely patterned delivery of metabolic inputs from the vascular layer via the glial layer. Simulations show that the capacity of such a system to sustain patterns is weak when the glial layer is absent. Capacity is higher when a glial layer is present and increases with the layer size. The proposed formulation of neurovascular interactions raises many intriguing questions about the role of glial cells in cerebral circulation.
Collapse
Affiliation(s)
- Rohit Gandrakota
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai, 600036, India
| | - V. S. Chakravarthy
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai, 600036, India
| | - Ranjan K. Pradhan
- Biotechnology and Bioengineering Center, Medical College of Wisconsin, Wauwatosa, WI 53226, U.S.A
| |
Collapse
|
194
|
Montgomery SH, Capellini I, Barton RA, Mundy NI. Reconstructing the ups and downs of primate brain evolution: implications for adaptive hypotheses and Homo floresiensis. BMC Biol 2010; 8:9. [PMID: 20105283 PMCID: PMC2825212 DOI: 10.1186/1741-7007-8-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 01/27/2010] [Indexed: 11/28/2022] Open
Abstract
Background Brain size is a key adaptive trait. It is often assumed that increasing brain size was a general evolutionary trend in primates, yet recent fossil discoveries have documented brain size decreases in some lineages, raising the question of how general a trend there was for brains to increase in mass over evolutionary time. We present the first systematic phylogenetic analysis designed to answer this question. Results We performed ancestral state reconstructions of three traits (absolute brain mass, absolute body mass, relative brain mass) using 37 extant and 23 extinct primate species and three approaches to ancestral state reconstruction: parsimony, maximum likelihood and Bayesian Markov-chain Monte Carlo. Both absolute and relative brain mass generally increased over evolutionary time, but body mass did not. Nevertheless both absolute and relative brain mass decreased along several branches. Applying these results to the contentious case of Homo floresiensis, we find a number of scenarios under which the proposed evolution of Homo floresiensis' small brain appears to be consistent with patterns observed along other lineages, dependent on body mass and phylogenetic position. Conclusions Our results confirm that brain expansion began early in primate evolution and show that increases occurred in all major clades. Only in terms of an increase in absolute mass does the human lineage appear particularly striking, with both the rate of proportional change in mass and relative brain size having episodes of greater expansion elsewhere on the primate phylogeny. However, decreases in brain mass also occurred along branches in all major clades, and we conclude that, while selection has acted to enlarge primate brains, in some lineages this trend has been reversed. Further analyses of the phylogenetic position of Homo floresiensis and better body mass estimates are required to confirm the plausibility of the evolution of its small brain mass. We find that for our dataset the Bayesian analysis for ancestral state reconstruction is least affected by inclusion of fossil data suggesting that this approach might be preferable for future studies on other taxa with a poor fossil record.
Collapse
|
195
|
Filming the glial dreams: real-time imaging of cannabinoid receptor trafficking in astrocytes. ASN Neuro 2009; 1:AN20090049. [PMID: 19909235 PMCID: PMC2826102 DOI: 10.1042/an20090049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
How does the brain process incoming information and produce thoughts? These questions represent, to all likelihood, the most challenging matters ever faced by natural sciences, matters which may never be fully comprehended. The evolution of the nervous system that, in about billion of years, brought into existence the human brain progressed through an ever-increasing complexity of neural networks. This evolution began from the diffuse nervous system, in which primordial neurons were able to sense the environmental inputs and convey them to effector organs and to the neighbouring neurons. At the next evolutionary stage the conglomerates of neuronal cell bodies, the ganglia, appeared, thus forming the primitive centralized nervous system. The developments which ensued went through a continuous increase in complexity of neuronal conglomerates, which eventually formed the central nervous system, which attained maximal perfection in mammals. In this issue of ASN NEURO, Osborne et al. have described details of real-time imaging of cannabinoid receptor trafficking in astrocytes, a technique that will help to elucidate the role of these receptors in the ever-increasing complex neural networks.
Collapse
|
196
|
Sherwood CC, Raghanti MA, Stimpson CD, Spocter MA, Uddin M, Boddy AM, Wildman DE, Bonar CJ, Lewandowski AH, Phillips KA, Erwin JM, Hof PR. Inhibitory interneurons of the human prefrontal cortex display conserved evolution of the phenotype and related genes. Proc Biol Sci 2009; 277:1011-20. [PMID: 19955152 DOI: 10.1098/rspb.2009.1831] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inhibitory interneurons participate in local processing circuits, playing a central role in executive cognitive functions of the prefrontal cortex. Although humans differ from other primates in a number of cognitive domains, it is not currently known whether the interneuron system has changed in the course of primate evolution leading to our species. In this study, we examined the distribution of different interneuron subtypes in the prefrontal cortex of anthropoid primates as revealed by immunohistochemistry against the calcium-binding proteins calbindin, calretinin and parvalbumin. In addition, we tested whether genes involved in the specification, differentiation and migration of interneurons show evidence of positive selection in the evolution of humans. Our findings demonstrate that cellular distributions of interneuron subtypes in human prefrontal cortex are similar to other anthropoid primates and can be explained by general scaling rules. Furthermore, genes underlying interneuron development are highly conserved at the amino acid level in primate evolution. Taken together, these results suggest that the prefrontal cortex in humans retains a similar inhibitory circuitry to that in closely related primates, even though it performs functional operations that are unique to our species. Thus, it is likely that other significant modifications to the connectivity and molecular biology of the prefrontal cortex were overlaid on this conserved interneuron architecture in the course of human evolution.
Collapse
Affiliation(s)
- Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20052, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Koerber JT, Klimczak R, Jang JH, Dalkara D, Flannery JG, Schaffer DV. Molecular evolution of adeno-associated virus for enhanced glial gene delivery. Mol Ther 2009; 17:2088-95. [PMID: 19672246 PMCID: PMC2788045 DOI: 10.1038/mt.2009.184] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 07/15/2009] [Indexed: 01/15/2023] Open
Abstract
The natural tropism of most viral vectors, including adeno-associated viral (AAV) vectors, leads to predominant transduction of neurons and epithelia within the central nervous system (CNS) and retina. Despite the clinical relevance of glia for homeostasis in neural tissue, and as causal contributors in genetic disorders such as Alzheimer's and amyotrophic lateral sclerosis, efforts to develop more efficient gene delivery vectors for glia have met with limited success. Recently, viral vector engineering involving high-throughput random diversification and selection has enabled the rapid creation of AAV vectors with valuable new gene delivery properties. We have engineered novel AAV variants capable of efficient glia transduction by employing directed evolution with a panel of four distinct AAV libraries, including a new semi-random peptide replacement strategy. These variants transduced both human and rat astrocytes in vitro up to 15-fold higher than their parent serotypes, and injection into the rat striatum yielded astrocyte transduction levels up to 16% of the total transduced cell population, despite the human astrocyte selection platform. Furthermore, one variant exhibited a substantial shift in tropism toward Müller glia within the retina, further highlighting the general utility of these variants for efficient glia transduction in multiple species within the CNS and retina.
Collapse
Affiliation(s)
- James T Koerber
- Department of Chemical Engineering, Helen Wills Neuroscience Institute, The University of California, Berkeley, California 94720-1462, USA
| | | | | | | | | | | |
Collapse
|
198
|
Phylogenomic analyses reveal convergent patterns of adaptive evolution in elephant and human ancestries. Proc Natl Acad Sci U S A 2009; 106:20824-9. [PMID: 19926857 DOI: 10.1073/pnas.0911239106] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Specific sets of brain-expressed genes, such as aerobic energy metabolism genes, evolved adaptively in the ancestry of humans and may have evolved adaptively in the ancestry of other large-brained mammals. The recent addition of genomes from two afrotherians (elephant and tenrec) to the expanding set of publically available sequenced mammalian genomes provided an opportunity to test this hypothesis. Elephants resemble humans by having large brains and long life spans; tenrecs, in contrast, have small brains and short life spans. Thus, we investigated whether the phylogenomic patterns of adaptive evolution are more similar between elephant and human than between either elephant and tenrec lineages or human and mouse lineages, and whether aerobic energy metabolism genes are especially well represented in the elephant and human patterns. Our analyses encompassed approximately 6,000 genes in each of these lineages with each gene yielding extensive coding sequence matches in interordinal comparisons. Each gene's nonsynonymous and synonymous nucleotide substitution rates and dN/dS ratios were determined. Then, from gene ontology information on genes with the higher dN/dS ratios, we identified the more prevalent sets of genes that belong to specific functional categories and that evolved adaptively. Elephant and human lineages showed much slower nucleotide substitution rates than tenrec and mouse lineages but more adaptively evolved genes. In correlation with absolute brain size and brain oxygen consumption being largest in elephants and next largest in humans, adaptively evolved aerobic energy metabolism genes were most evident in the elephant lineage and next most evident in the human lineage.
Collapse
|
199
|
Abstract
Astrocytes are the main neural cell type responsible for the maintenance of brain homeostasis. They form highly organized anatomical domains that are interconnected into extensive networks. These features, along with the expression of a wide array of receptors, transporters, and ion channels, ideally position them to sense and dynamically modulate neuronal activity. Astrocytes cooperate with neurons on several levels, including neurotransmitter trafficking and recycling, ion homeostasis, energy metabolism, and defense against oxidative stress. The critical dependence of neurons upon their constant support confers astrocytes with intrinsic neuroprotective properties which are discussed here. Conversely, pathogenic stimuli may disturb astrocytic function, thus compromising neuronal functionality and viability. Using neuroinflammation, Alzheimer's disease, and hepatic encephalopathy as examples, we discuss how astrocytic defense mechanisms may be overwhelmed in pathological conditions, contributing to disease progression.
Collapse
Affiliation(s)
- Mireille Bélanger
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
200
|
Herculano-Houzel S. The human brain in numbers: a linearly scaled-up primate brain. Front Hum Neurosci 2009; 3:31. [PMID: 19915731 PMCID: PMC2776484 DOI: 10.3389/neuro.09.031.2009] [Citation(s) in RCA: 667] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 09/29/2009] [Indexed: 11/13/2022] Open
Abstract
The human brain has often been viewed as outstanding among mammalian brains: the most cognitively able, the largest-than-expected from body size, endowed with an overdeveloped cerebral cortex that represents over 80% of brain mass, and purportedly containing 100 billion neurons and 10x more glial cells. Such uniqueness was seemingly necessary to justify the superior cognitive abilities of humans over larger-brained mammals such as elephants and whales. However, our recent studies using a novel method to determine the cellular composition of the brain of humans and other primates as well as of rodents and insectivores show that, since different cellular scaling rules apply to the brains within these orders, brain size can no longer be considered a proxy for the number of neurons in the brain. These studies also showed that the human brain is not exceptional in its cellular composition, as it was found to contain as many neuronal and non-neuronal cells as would be expected of a primate brain of its size. Additionally, the so-called overdeveloped human cerebral cortex holds only 19% of all brain neurons, a fraction that is similar to that found in other mammals. In what regards absolute numbers of neurons, however, the human brain does have two advantages compared to other mammalian brains: compared to rodents, and probably to whales and elephants as well, it is built according to the very economical, space-saving scaling rules that apply to other primates; and, among economically built primate brains, it is the largest, hence containing the most neurons. These findings argue in favor of a view of cognitive abilities that is centered on absolute numbers of neurons, rather than on body size or encephalization, and call for a re-examination of several concepts related to the exceptionality of the human brain.
Collapse
Affiliation(s)
- Suzana Herculano-Houzel
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brasil.
| |
Collapse
|