1
|
Edwards E, Schenone D, Sivagnanalingam U, Perry S, Mullen CA. GAP JUNCTION FUNCTION IS ESSENTIAL FOR SURVIVAL OF ACUTE LYMPHOBLASTIC LEUKEMIA CELLS. Exp Oncol 2024; 46:110-118. [PMID: 39396173 DOI: 10.15407/exp-oncology.2024.02.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Acute lymphoblastic leukemia has an intimate physical relationship with nonmalignant bone marrow stromal cells. We have recently demonstrated that stromal cells contribute to the survival of leukemia cells and that there is a bidirectional transfer of intracellular material between them. Understanding the mechanisms of stromal support of leukemia may provide insights into new therapies. AIM To test the hypothesis that gap junctions are formed between acute lymphoblastic leukemia cells and nonmalignant stromal cells, and that gap junction function is essential for the survival of leukemia cells. MATERIALS AND METHODS We employed a well-characterized in vitro model of human bone marrow stromal cells and primary human B lymphoblastic leukemia cells and measured leukemia cell survival in coculture using flow cytometry. We measured the effects of gap junction antagonist peptides, carbenoxolone (a drug known to interfere with the gap junction function), and several leukemia chemotherapy drugs including methotrexate upon leukemia cell survival. RESULTS We demonstrated that stromal cells need to be alive and metabolically active to keep leukemia cells alive. Physical contact between stromal and leukemia cells leads to an increase in gap junction proteins in leukemia cells. Gap junction inhibitory peptides impaired leukemia cell survival as did carbenoxolone, a nonpeptide inhibitor of the gap junction function. Stromal cell survival was not affected. We observed a very modest enhancement of methotrexate antileukemia activity by low-dose carbenoxolone but no significant interactions with dexamethasone, vincristine, mercaptopurine, or doxorubicin. CONCLUSION These studies demonstrate that acute lymphoblastic cell survival is impaired by interference with the gap junction function. The development of drugs targeting gap junctions may provide a novel approach to the therapy of acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- E Edwards
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - D Schenone
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - U Sivagnanalingam
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - S Perry
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - C A Mullen
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
2
|
Miles KD, Barker CM, Russell KP, Appel BH, Doll CA. Electrical Synapses Mediate Embryonic Hyperactivity in a Zebrafish Model of Fragile X Syndrome. J Neurosci 2024; 44:e2275232024. [PMID: 38969506 PMCID: PMC11293453 DOI: 10.1523/jneurosci.2275-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
Although hyperactivity is associated with a wide variety of neurodevelopmental disorders, the early embryonic origins of locomotion have hindered investigation of pathogenesis of these debilitating behaviors. The earliest motor output in vertebrate animals is generated by clusters of early-born motor neurons (MNs) that occupy distinct regions of the spinal cord, innervating stereotyped muscle groups. Gap junction electrical synapses drive early spontaneous behavior in zebrafish, prior to the emergence of chemical neurotransmitter networks. We use a genetic model of hyperactivity to gain critical insight into the consequences of errors in motor circuit formation and function, finding that Fragile X syndrome model mutant zebrafish are hyperexcitable from the earliest phases of spontaneous behavior, show altered sensitivity to blockade of electrical gap junctions, and have increased expression of the gap junction protein Connexin 34/35. We further show that this hyperexcitable behavior can be rescued by pharmacological inhibition of electrical synapses. We also use functional imaging to examine MN and interneuron (IN) activity in early embryogenesis, finding genetic disruption of electrical gap junctions uncouples activity between mnx1 + MNs and INs. Taken together, our work highlights the importance of electrical synapses in motor development and suggests that the origins of hyperactivity in neurodevelopmental disorders may be established during the initial formation of locomotive circuits.
Collapse
Affiliation(s)
- Kaleb D Miles
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Chase M Barker
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Kristen P Russell
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Bruce H Appel
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Caleb A Doll
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
3
|
Wallace JL, Pollen AA. Human neuronal maturation comes of age: cellular mechanisms and species differences. Nat Rev Neurosci 2024; 25:7-29. [PMID: 37996703 DOI: 10.1038/s41583-023-00760-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 11/25/2023]
Abstract
The delayed and prolonged postmitotic maturation of human neurons, compared with neurons from other species, may contribute to human-specific cognitive abilities and neurological disorders. Here we review the mechanisms of neuronal maturation, applying lessons from model systems to understand the specific features of protracted human cortical maturation and species differences. We cover cell-intrinsic features of neuronal maturation, including transcriptional, epigenetic and metabolic mechanisms, as well as cell-extrinsic features, including the roles of activity and synapses, the actions of glial cells and the contribution of the extracellular matrix. We discuss evidence for species differences in biochemical reaction rates, the proposed existence of an epigenetic maturation clock and the contributions of both general and modular mechanisms to species-specific maturation timing. Finally, we suggest approaches to measure, improve and accelerate the maturation of human neurons in culture, examine crosstalk and interactions among these different aspects of maturation and propose conceptual models to guide future studies.
Collapse
Affiliation(s)
- Jenelle L Wallace
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Butler J, Dale N. X-linked Charcot Marie Tooth mutations alter CO 2 sensitivity of connexin32 hemichannels. Front Cell Neurosci 2023; 17:1330983. [PMID: 38188670 PMCID: PMC10771293 DOI: 10.3389/fncel.2023.1330983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Connexin32 (Cx32) is expressed in myelinating Schwann cells. It forms both reflexive gap junctions, to facilitate transfer of molecules from the outer to the inner myelin layers and hemichannels at the paranode to permit action potential-evoked release of ATP into the extracellular space. Loss of function mutations in Cx32 cause X-linked Charcot Marie Tooth disease (CMTX), a slowly developing peripheral neuropathy. The mechanistic links between Cx32 mutations and CMTX are not well understood. As Cx32 hemichannels can be opened by increases in PCO2, we have examined whether CMTX mutations alter this CO2 sensitivity. By using Ca2+ imaging, dye loading and genetically encoded ATP sensors to measure ATP release, we have found 5 CMTX mutations that abolish the CO2 sensitivity of Cx32 hemichannels (A88D, 111-116 Del, C179Y, E102G, V139M). Others cause a partial loss (L56F, R220Stop, and R15W). Some CMTX mutations have no apparent effect on CO2 sensitivity (R15Q, L9F, G12S, V13L, V84I, W133R). The mutation R15W alters multiple additional aspects of hemichannel function including Ca2+ and ATP permeability. The mutations that abolish CO2 sensitivity are transdominant and abolish CO2 sensitivity of co-expressed Cx32WT. We have shown that Schwannoma RT4 D6P2T cells can release ATP in response to elevated PCO2 via the opening of Cx32. This is consistent with the hypothesis that the CO2 sensitivity of Cx32 may be important for maintenance of healthy myelin. Our data, showing a transdominant effect of certain CMTX mutations on CO2 sensitivity, may need to be taken into account in any future gene therapies for this condition.
Collapse
Affiliation(s)
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
5
|
León-Fuentes IM, Salgado-Gil MG, Novoa MS, Retamal MA. Connexins in Cancer, the Possible Role of Connexin46 as a Cancer Stem Cell-Determining Protein. Biomolecules 2023; 13:1460. [PMID: 37892142 PMCID: PMC10604234 DOI: 10.3390/biom13101460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/15/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is a widespread and incurable disease caused by genetic mutations, leading to uncontrolled cell proliferation and metastasis. Connexins (Cx) are transmembrane proteins that facilitate intercellular communication via hemichannels and gap junction channels. Among them, Cx46 is found mostly in the eye lens. However, in pathological conditions, Cx46 has been observed in various types of cancers, such as glioblastoma, melanoma, and breast cancer. It has been demonstrated that elevated Cx46 levels in breast cancer contribute to cellular resistance to hypoxia, and it is an enhancer of cancer aggressiveness supporting a pro-tumoral role. Accordingly, Cx46 is associated with an increase in cancer stem cell phenotype. These cells display radio- and chemoresistance, high proliferative abilities, self-renewal, and differentiation capacities. This review aims to consolidate the knowledge of the relationship between Cx46, its role in forming hemichannels and gap junctions, and its connection with cancer and cancer stem cells.
Collapse
Affiliation(s)
| | | | | | - Mauricio A. Retamal
- Programa de Comunicación Celular en Cáncer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, República de Honduras 12740, Las Condes, Santiago 7610496, Chile; (I.M.L.-F.); (M.G.S.-G.); (M.S.N.)
| |
Collapse
|
6
|
Horibe K, Taga G, Fujimoto K. Geodesic theory of long association fibers arrangement in the human fetal cortex. Cereb Cortex 2023; 33:9778-9786. [PMID: 37482884 PMCID: PMC10472492 DOI: 10.1093/cercor/bhad243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
Association fibers connect different areas of the cerebral cortex over long distances and integrate information to achieve higher brain functions, particularly in humans. Prototyped association fibers are developed to the respective tangential direction throughout the cerebral hemispheres along the deepest border of the subplate during the fetal period. However, how guidance to remote areas is achieved is not known. Because the subplate is located below the cortical surface, the tangential direction of the fibers may be biased by the curved surface geometry due to Sylvian fissure and cortical poles. The fiber length can be minimized if the tracts follow the shortest paths (geodesics) of the curved surface. Here, we propose and examine a theory that geodesics guide the tangential direction of long association fibers by analyzing how geodesics are spatially distributed on the fetal human brains. We found that the geodesics were dense on the saddle-shaped surface of the perisylvian region and sparse on the dome-shaped cortical poles. The geodesics corresponded with the arrangement of five typical association fibers, supporting the theory. Thus, the geodesic theory provides directional guidance information for wiring remote areas and suggests that long association fibers emerge from minimizing their tangential length in fetal brains.
Collapse
Affiliation(s)
- Kazuya Horibe
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, 560-0043 Osaka, Japan
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, 560-8531 Osaka, Japan
| | - Gentaro Taga
- Department of Physical and Health Education, Graduate School of Education, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Koichi Fujimoto
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, 560-0043 Osaka, Japan
- Program of Mathematical and Life Sciences, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, 739-8526 Hiroshima, Japan
| |
Collapse
|
7
|
Li H, Guo A, Salgado M, Sáez JC, Lau CG. The connexin hemichannel inhibitor D4 produces rapid antidepressant-like effects in mice. J Neuroinflammation 2023; 20:191. [PMID: 37599352 PMCID: PMC10440914 DOI: 10.1186/s12974-023-02873-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023] Open
Abstract
Depression is a common mood disorder characterized by a range of clinical symptoms, including prolonged low mood and diminished interest. Although many clinical and animal studies have provided significant insights into the pathophysiology of depression, current treatment strategies are not sufficient to manage this disorder. It has been suggested that connexin (Cx)-based hemichannels are candidates for depression intervention by modifying the state of neuroinflammation. In this study, we investigated the antidepressant-like effect of a recently discovered selective Cx hemichannel inhibitor, a small organic molecule called D4. We first showed that D4 reduced hemichannel activity following systemic inflammation after LPS injections. Next, we found that D4 treatment prevented LPS-induced inflammatory response and depressive-like behaviors. These behavioral effects were accompanied by reduced astrocytic activation and hemichannel activity in depressive-like mice induced by repeated low-dose LPS challenges. D4 treatment also reverses depressive-like symptoms in mice subjected to chronic restraint stress (CRS). To test whether D4 broadly affected neural activity, we measured c-Fos expression in depression-related brain regions and found a reduction in c-Fos+ cells in different brain regions. D4 significantly normalized CRS-induced hypoactivation in several brain regions, including the hippocampus, entorhinal cortex, and lateral septum. Together, these results indicate that blocking Cx hemichannels using D4 can normalize neuronal activity and reduce depressive-like symptoms in mice by reducing neuroinflammation. Our work provides evidence of the antidepressant-like effect of D4 and supports glial Cx hemichannels as potential therapeutic targets for depression.
Collapse
Affiliation(s)
- Huanhuan Li
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Anni Guo
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Magdiel Salgado
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, 2381850, Valparaíso, Chile
| | - Juan C Sáez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, 2381850, Valparaíso, Chile
| | - Chunyue Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
8
|
Cao JW, Liu LY, Yu YC. Gap junctions regulate the development of neural circuits in the neocortex. Curr Opin Neurobiol 2023; 81:102735. [PMID: 37263136 DOI: 10.1016/j.conb.2023.102735] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/12/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023]
Abstract
Gap junctions between cells are ubiquitously expressed in the developing brain. They are involved in major steps of neocortical development, including neurogenesis, cell migration, synaptogenesis, and neural circuit formation, and have been implicated in cortical column formation. Dysfunctional gap junctions can contribute to or even cause a variety of brain diseases. Although the role of gap junctions in neocortical development is better known, a comprehensive understanding of their functions is far from complete. Here we explore several critical open questions surrounding gap junctions and their involvement in neural circuit development. Addressing them will greatly impact our understanding of the fundamental mechanisms of neocortical structure and function as well as the etiology of brain disease.
Collapse
Affiliation(s)
- Jun-Wei Cao
- School of Basic Medical Sciences, Xiangnan University, Chenzhou, Hunan 423000, China
| | - Lin-Yun Liu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai 200032, China
| | - Yong-Chun Yu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai 200032, China.
| |
Collapse
|
9
|
Suárez R, Bluett T, McCullough MH, Avitan L, Black DA, Paolino A, Fenlon LR, Goodhill GJ, Richards LJ. Cortical activity emerges in region-specific patterns during early brain development. Proc Natl Acad Sci U S A 2023; 120:e2208654120. [PMID: 37216522 PMCID: PMC10235933 DOI: 10.1073/pnas.2208654120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
The development of precise neural circuits in the brain requires spontaneous patterns of neural activity prior to functional maturation. In the rodent cerebral cortex, patchwork and wave patterns of activity develop in somatosensory and visual regions, respectively, and are present at birth. However, whether such activity patterns occur in noneutherian mammals, as well as when and how they arise during development, remain open questions relevant for understanding brain formation in health and disease. Since the onset of patterned cortical activity is challenging to study prenatally in eutherians, here we offer an approach in a minimally invasive manner using marsupial dunnarts, whose cortex forms postnatally. We discovered similar patchwork and travelling waves in the dunnart somatosensory and visual cortices at stage 27 (equivalent to newborn mice) and examined earlier stages of development to determine the onset of these patterns and how they first emerge. We observed that these patterns of activity emerge in a region-specific and sequential manner, becoming evident as early as stage 24 in somatosensory and stage 25 in visual cortices (equivalent to embryonic day 16 and 17, respectively, in mice), as cortical layers establish and thalamic axons innervate the cortex. In addition to sculpting synaptic connections of existing circuits, evolutionarily conserved patterns of neural activity could therefore help regulate other early events in cortical development.
Collapse
Affiliation(s)
- Rodrigo Suárez
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Tobias Bluett
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
| | - Michael H. McCullough
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
| | - Lilach Avitan
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
| | - Dylan A. Black
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Annalisa Paolino
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Laura R. Fenlon
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| | - Geoffrey J. Goodhill
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Mathematics and Physics, BrisbaneQLD4072, Australia
| | - Linda J. Richards
- The University of Queensland, Queensland Brain Institute, BrisbaneQLD4072, Australia
- The University of Queensland, School of Biomedical Sciences, BrisbaneQLD4072, Australia
| |
Collapse
|
10
|
Gellért L, Luhmann HJ, Kilb W. Axonal connections between S1 barrel, M1, and S2 cortex in the newborn mouse. Front Neuroanat 2023; 17:1105998. [PMID: 36760662 PMCID: PMC9905141 DOI: 10.3389/fnana.2023.1105998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
The development of functionally interconnected networks between primary (S1), secondary somatosensory (S2), and motor (M1) cortical areas requires coherent neuronal activity via corticocortical projections. However, the anatomical substrate of functional connections between S1 and M1 or S2 during early development remains elusive. In the present study, we used ex vivo carbocyanine dye (DiI) tracing in paraformaldehyde-fixed newborn mouse brain to investigate axonal projections of neurons in different layers of S1 barrel field (S1Bf), M1, and S2 toward the subplate (SP), a hub layer for sensory information transfer in the immature cortex. In addition, we performed extracellular recordings in neocortical slices to unravel the functional connectivity between these areas. Our experiments demonstrate that already at P0 neurons from the cortical plate (CP), layer 5/6 (L5/6), and the SP of both M1 and S2 send projections through the SP of S1Bf. Reciprocally, neurons from CP to SP of S1Bf send projections through the SP of M1 and S2. Electrophysiological recordings with multi-electrode arrays in cortical slices revealed weak, but functional synaptic connections between SP and L5/6 within and between S1 and M1. An even lower functional connectivity was observed between S1 and S2. In summary, our findings demonstrate that functional connections between SP and upper cortical layers are not confined to the same cortical area, but corticocortical connection between adjacent cortical areas exist already at the day of birth. Hereby, SP can integrate early cortical activity of M1, S1, and S2 and shape the development of sensorimotor integration at an early stage.
Collapse
|
11
|
Mukherjee D, Kanold PO. Changing subplate circuits: Early activity dependent circuit plasticity. Front Cell Neurosci 2023; 16:1067365. [PMID: 36713777 PMCID: PMC9874351 DOI: 10.3389/fncel.2022.1067365] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Early neural activity in the developing sensory system comprises spontaneous bursts of patterned activity, which is fundamental for sculpting and refinement of immature cortical connections. The crude early connections that are initially refined by spontaneous activity, are further elaborated by sensory-driven activity from the periphery such that orderly and mature connections are established for the proper functioning of the cortices. Subplate neurons (SPNs) are one of the first-born mature neurons that are transiently present during early development, the period of heightened activity-dependent plasticity. SPNs are well integrated within the developing sensory cortices. Their structural and functional properties such as relative mature intrinsic membrane properties, heightened connectivity via chemical and electrical synapses, robust activation by neuromodulatory inputs-place them in an ideal position to serve as crucial elements in monitoring and regulating spontaneous endogenous network activity. Moreover, SPNs are the earliest substrates to receive early sensory-driven activity from the periphery and are involved in its modulation, amplification, and transmission before the maturation of the direct adult-like thalamocortical connectivity. Consequently, SPNs are vulnerable to sensory manipulations in the periphery. A broad range of early sensory deprivations alters SPN circuit organization and functions that might be associated with long term neurodevelopmental and psychiatric disorders. Here we provide a comprehensive overview of SPN function in activity-dependent development during early life and integrate recent findings on the impact of early sensory deprivation on SPNs that could eventually lead to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Didhiti Mukherjee
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Patrick O. Kanold
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States,Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, United States,*Correspondence: Patrick O. Kanold ✉
| |
Collapse
|
12
|
Petanjek Z, Banovac I, Sedmak D, Hladnik A. Dendritic Spines: Synaptogenesis and Synaptic Pruning for the Developmental Organization of Brain Circuits. ADVANCES IN NEUROBIOLOGY 2023; 34:143-221. [PMID: 37962796 DOI: 10.1007/978-3-031-36159-3_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synaptic overproduction and elimination is a regular developmental event in the mammalian brain. In the cerebral cortex, synaptic overproduction is almost exclusively correlated with glutamatergic synapses located on dendritic spines. Therefore, analysis of changes in spine density on different parts of the dendritic tree in identified classes of principal neurons could provide insight into developmental reorganization of specific microcircuits.The activity-dependent stabilization and selective elimination of the initially overproduced synapses is a major mechanism for generating diversity of neural connections beyond their genetic determination. The largest number of overproduced synapses was found in the monkey and human cerebral cortex. The highest (exceeding adult values by two- to threefold) and most protracted overproduction (up to third decade of life) was described for associative layer IIIC pyramidal neurons in the human dorsolateral prefrontal cortex.Therefore, the highest proportion and extraordinarily extended phase of synaptic spine overproduction is a hallmark of neural circuitry in human higher-order associative areas. This indicates that microcircuits processing the most complex human cognitive functions have the highest level of developmental plasticity. This finding is the backbone for understanding the effect of environmental impact on the development of the most complex, human-specific cognitive and emotional capacities, and on the late onset of human-specific neuropsychiatric disorders, such as autism and schizophrenia.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
13
|
Baracaldo-Santamaría D, Corrales-Hernández MG, Ortiz-Vergara MC, Cormane-Alfaro V, Luque-Bernal RM, Calderon-Ospina CA, Cediel-Becerra JF. Connexins and Pannexins: Important Players in Neurodevelopment, Neurological Diseases, and Potential Therapeutics. Biomedicines 2022; 10:2237. [PMID: 36140338 PMCID: PMC9496069 DOI: 10.3390/biomedicines10092237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Cell-to-cell communication is essential for proper embryonic development and its dysfunction may lead to disease. Recent research has drawn attention to a new group of molecules called connexins (Cxs) and pannexins (Panxs). Cxs have been described for more than forty years as pivotal regulators of embryogenesis; however, the exact mechanism by which they provide this regulation has not been clearly elucidated. Consequently, Cxs and Panxs have been linked to congenital neurodegenerative diseases such as Charcot-Marie-Tooth disease and, more recently, chronic hemichannel opening has been associated with adult neurodegenerative diseases (e.g., Alzheimer's disease). Cell-to-cell communication via gap junctions formed by hexameric assemblies of Cxs, known as connexons, is believed to be a crucial component in developmental regulation. As for Panxs, despite being topologically similar to Cxs, they predominantly seem to form channels connecting the cytoplasm to the extracellular space and, despite recent research into Panx1 (Pannexin 1) expression in different regions of the brain during the embryonic phase, it has been studied to a lesser degree. When it comes to the nervous system, Cxs and Panxs play an important role in early stages of neuronal development with a wide span of action ranging from cellular migration during early stages to neuronal differentiation and system circuitry formation. In this review, we describe the most recent available evidence regarding the molecular and structural aspects of Cx and Panx channels, their role in neurodevelopment, congenital and adult neurological diseases, and finally propose how pharmacological modulation of these channels could modify the pathogenesis of some diseases.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - María Gabriela Corrales-Hernández
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Maria Camila Ortiz-Vergara
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Valeria Cormane-Alfaro
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Ricardo-Miguel Luque-Bernal
- Anatomy and Embriology Units, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos-Alberto Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Juan-Fernando Cediel-Becerra
- Histology and Embryology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
14
|
Connexins Signatures of the Neurovascular Unit and Their Physio-Pathological Functions. Int J Mol Sci 2022; 23:ijms23179510. [PMID: 36076908 PMCID: PMC9455936 DOI: 10.3390/ijms23179510] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Central nervous system (CNS) homeostasis is closely linked to the delicate balance of the microenvironment in which different cellular components of the neurovascular unit (NVU) coexist. Intercellular communication plays a pivotal role in exchanges of signaling molecules and mediators essential for survival functions, as well as in the removal of disturbing elements that can lead to related pathologies. The specific signatures of connexins (Cxs), proteins which form either gap junctions (GJs) or hemichannels (HCs), represent the biological substrate of the pathophysiological balance. Connexin 43 (Cx43) is undoubtedly one of the most important factors in glia–neuro–vascular crosstalk. Herein, Cxs signatures of every NVU component are highlighted and their critical influence on functional processes in healthy and pathological conditions of nervous microenvironment is reviewed.
Collapse
|
15
|
Burboa PC, Puebla M, Gaete PS, Durán WN, Lillo MA. Connexin and Pannexin Large-Pore Channels in Microcirculation and Neurovascular Coupling Function. Int J Mol Sci 2022; 23:ijms23137303. [PMID: 35806312 PMCID: PMC9266979 DOI: 10.3390/ijms23137303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 01/27/2023] Open
Abstract
Microcirculation homeostasis depends on several channels permeable to ions and/or small molecules that facilitate the regulation of the vasomotor tone, hyperpermeability, the blood–brain barrier, and the neurovascular coupling function. Connexin (Cxs) and Pannexin (Panxs) large-pore channel proteins are implicated in several aspects of vascular physiology. The permeation of ions (i.e., Ca2+) and key metabolites (ATP, prostaglandins, D-serine, etc.) through Cxs (i.e., gap junction channels or hemichannels) and Panxs proteins plays a vital role in intercellular communication and maintaining vascular homeostasis. Therefore, dysregulation or genetic pathologies associated with these channels promote deleterious tissue consequences. This review provides an overview of current knowledge concerning the physiological role of these large-pore molecule channels in microcirculation (arterioles, capillaries, venules) and in the neurovascular coupling function.
Collapse
Affiliation(s)
- Pía C. Burboa
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Departamento de Morfología y Función, Facultad de Salud y Ciencias Sociales, Sede Santiago Centro, Universidad de las Américas, Avenue República 71, Santiago 8370040, Chile;
| | - Mariela Puebla
- Departamento de Morfología y Función, Facultad de Salud y Ciencias Sociales, Sede Santiago Centro, Universidad de las Américas, Avenue República 71, Santiago 8370040, Chile;
| | - Pablo S. Gaete
- Department of Physiology and Membrane Biology, University of California at Davis, Davis, CA 95616, USA;
| | - Walter N. Durán
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Rutgers School of Graduate Studies, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Mauricio A. Lillo
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Correspondence:
| |
Collapse
|
16
|
Luhmann HJ, Kanold PO, Molnár Z, Vanhatalo S. Early brain activity: Translations between bedside and laboratory. Prog Neurobiol 2022; 213:102268. [PMID: 35364141 PMCID: PMC9923767 DOI: 10.1016/j.pneurobio.2022.102268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/01/2022] [Accepted: 03/25/2022] [Indexed: 01/29/2023]
Abstract
Neural activity is both a driver of brain development and a readout of developmental processes. Changes in neuronal activity are therefore both the cause and consequence of neurodevelopmental compromises. Here, we review the assessment of neuronal activities in both preclinical models and clinical situations. We focus on issues that require urgent translational research, the challenges and bottlenecks preventing translation of biomedical research into new clinical diagnostics or treatments, and possibilities to overcome these barriers. The key questions are (i) what can be measured in clinical settings versus animal experiments, (ii) how do measurements relate to particular stages of development, and (iii) how can we balance practical and ethical realities with methodological compromises in measurements and treatments.
Collapse
Affiliation(s)
- Heiko J. Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz, Germany.,Correspondence:, , ,
| | - Patrick O. Kanold
- Department of Biomedical Engineering and Kavli Neuroscience Discovery Institute, Johns Hopkins University, School of Medicine, 720 Rutland Avenue / Miller 379, Baltimore, MD 21205, USA.,Correspondence:, , ,
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford OX1 3PT, UK.
| | - Sampsa Vanhatalo
- BABA Center, Departments of Physiology and Clinical Neurophysiology, Children's Hospital, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
17
|
Kelley KW, Pașca SP. Human brain organogenesis: Toward a cellular understanding of development and disease. Cell 2021; 185:42-61. [PMID: 34774127 DOI: 10.1016/j.cell.2021.10.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023]
Abstract
The construction of the human nervous system is a distinctly complex although highly regulated process. Human tissue inaccessibility has impeded a molecular understanding of the developmental specializations from which our unique cognitive capacities arise. A confluence of recent technological advances in genomics and stem cell-based tissue modeling is laying the foundation for a new understanding of human neural development and dysfunction in neuropsychiatric disease. Here, we review recent progress on uncovering the cellular and molecular principles of human brain organogenesis in vivo as well as using organoids and assembloids in vitro to model features of human evolution and disease.
Collapse
Affiliation(s)
- Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA.
| |
Collapse
|
18
|
CO 2-Sensitive Connexin Hemichannels in Neurons and Glia: Three Different Modes of Signalling? Int J Mol Sci 2021; 22:ijms22147254. [PMID: 34298872 PMCID: PMC8304244 DOI: 10.3390/ijms22147254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 01/28/2023] Open
Abstract
Connexins can assemble into either gap junctions (between two cells) or hemichannels (from one cell to the extracellular space) and mediate cell-to-cell signalling. A subset of connexins (Cx26, Cx30, Cx32) are directly sensitive to CO2 and fluctuations in the level within a physiological range affect their open probability, and thus, change cell conductance. These connexins are primarily found on astrocytes or oligodendrocytes, where increased CO2 leads to ATP release, which acts on P2X and P2Y receptors of neighbouring neurons and changes excitability. CO2-sensitive hemichannels are also found on developing cortical neurons, where they play a role in producing spontaneous neuronal activity. It is plausible that the transient opening of hemichannels allows cation influx, leading to depolarisation. Recently, we have shown that dopaminergic neurons in the substantia nigra and GABAergic neurons in the VTA also express Cx26 hemichannels. An increase in the level of CO2 results in hemichannel opening, increasing whole-cell conductance, and decreasing neuronal excitability. We found that the expression of Cx26 in the dopaminergic neurons in the substantia nigra at P7-10 is transferred to glial cells by P17-21, displaying a shift from being inhibitory (to neuronal activity) in young mice, to potentially excitatory (via ATP release). Thus, Cx26 hemichannels could have three modes of signalling (release of ATP, excitatory flickering open and shut and inhibitory shunting) depending on where they are expressed (neurons or glia) and the stage of development.
Collapse
|
19
|
Wallois F, Routier L, Heberlé C, Mahmoudzadeh M, Bourel-Ponchel E, Moghimi S. Back to basics: the neuronal substrates and mechanisms that underlie the electroencephalogram in premature neonates. Neurophysiol Clin 2020; 51:5-33. [PMID: 33162287 DOI: 10.1016/j.neucli.2020.10.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Electroencephalography is the only clinically available technique that can address the premature neonate normal and pathological functional development week after week. The changes in the electroencephalogram (EEG) result from gradual structural and functional modifications that arise during the last trimester of pregnancy. Here, we review the structural changes over time that underlie the establishment of functional immature neural networks, the impact of certain anatomical specificities (fontanelles, connectivity, etc.) on the EEG, limitations in EEG interpretation, and the utility of high-resolution EEG (HR-EEG) in premature newborns (a promising technique with a high degree of spatiotemporal resolution). In particular, we classify EEG features according to whether they are manifestations of endogenous generators (i.e. theta activities that coalesce with a slow wave or delta brushes) or come from a broader network. Furthermore, we review publications on EEG in premature animals because the data provide a better understanding of what is happening in premature newborns. We then discuss the results and limitations of functional connectivity analyses in premature newborns. Lastly, we report on the magnetoelectroencephalographic studies of brain activity in the fetus. A better understanding of complex interactions at various structural and functional levels during normal neurodevelopment (as assessed using electroencephalography as a benchmark method) might lead to better clinical care and monitoring for premature neonates.
Collapse
Affiliation(s)
- Fabrice Wallois
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France.
| | - Laura Routier
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Claire Heberlé
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Mahdi Mahmoudzadeh
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Emilie Bourel-Ponchel
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Sahar Moghimi
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| |
Collapse
|
20
|
Moghimi S, Shadkam A, Mahmoudzadeh M, Calipe O, Panzani M, Edalati M, Ghorbani M, Routier L, Wallois F. The intimate relationship between coalescent generators in very premature human newborn brains: Quantifying the coupling of nested endogenous oscillations. Hum Brain Mapp 2020; 41:4691-4703. [PMID: 33463873 PMCID: PMC7555093 DOI: 10.1002/hbm.25150] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 06/26/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Temporal theta slow-wave activity (TTA-SW) in premature infants is a specific neurobiomarker of the early neurodevelopment of perisylvian networks observed as early as 24 weeks of gestational age (wGA). It is present at the turning point between non-sensory driven spontaneous networks and cortical network functioning. Despite its clinical importance, the underlying mechanisms responsible for this spontaneous nested activity and its functional role have not yet been determined. The coupling between neural oscillations at different timescales is a key feature of ongoing neural activity, the characteristics of which are determined by the network structure and dynamics. The underlying mechanisms of cross-frequency coupling (CFC) are associated with several putative functions in adults. In order to show that this generic mechanism is already in place early in the course of development, we analyzed electroencephalography recordings from sleeping preterm newborns (24-27 wGA). Employing cross-frequency phase-amplitude coupling analyses, we found that TTAs were orchestrated by the SWs defined by a precise temporal relationship. Notably, TTAs were synchronized to the SW trough, and were suppressed during the SW peak. Spontaneous endogenous TTA-SWs constitute one of the very early signatures of the developing temporal neural networks with key functions, such as language and communication. The presence of a fine-tuned relationship between the slow activity and the TTA in premature neonates emphasizes the complexity and relative maturity of the intimate mechanisms that shape the CFC, the disruption of which can have severe neurodevelopmental consequences.
Collapse
Affiliation(s)
- Sahar Moghimi
- Electrical Engineering DepartmentFerdowsi University of MashhadIran
- Rayan Center for Neuroscience and BehaviorFerdowsi University of MashhadMashhadIran
- Inserm UMR1105, Groupe de Recherches sur l'Analyse Multimodale de la Fonction CérébraleCentre Universitaire de Recherches en SanteAmiens CedexFrance
| | - Azadeh Shadkam
- Electrical Engineering DepartmentFerdowsi University of MashhadIran
| | - Mahdi Mahmoudzadeh
- Inserm UMR1105, Groupe de Recherches sur l'Analyse Multimodale de la Fonction CérébraleCentre Universitaire de Recherches en SanteAmiens CedexFrance
- Inserm UMR1105, EFSN PédiatriquesCentre Hospitalier Universitaire Amiens sudAmiens CedexFrance
| | - Olivia Calipe
- Inserm UMR1105, Groupe de Recherches sur l'Analyse Multimodale de la Fonction CérébraleCentre Universitaire de Recherches en SanteAmiens CedexFrance
| | - Marine Panzani
- Inserm UMR1105, Groupe de Recherches sur l'Analyse Multimodale de la Fonction CérébraleCentre Universitaire de Recherches en SanteAmiens CedexFrance
| | - Mohammadreza Edalati
- Electrical Engineering DepartmentFerdowsi University of MashhadIran
- Inserm UMR1105, Groupe de Recherches sur l'Analyse Multimodale de la Fonction CérébraleCentre Universitaire de Recherches en SanteAmiens CedexFrance
| | - Maryam Ghorbani
- Electrical Engineering DepartmentFerdowsi University of MashhadIran
- Rayan Center for Neuroscience and BehaviorFerdowsi University of MashhadMashhadIran
| | - Laura Routier
- Inserm UMR1105, Groupe de Recherches sur l'Analyse Multimodale de la Fonction CérébraleCentre Universitaire de Recherches en SanteAmiens CedexFrance
- Inserm UMR1105, EFSN PédiatriquesCentre Hospitalier Universitaire Amiens sudAmiens CedexFrance
| | - Fabrice Wallois
- Inserm UMR1105, Groupe de Recherches sur l'Analyse Multimodale de la Fonction CérébraleCentre Universitaire de Recherches en SanteAmiens CedexFrance
- Inserm UMR1105, EFSN PédiatriquesCentre Hospitalier Universitaire Amiens sudAmiens CedexFrance
| |
Collapse
|
21
|
Molnár Z, Luhmann HJ, Kanold PO. Transient cortical circuits match spontaneous and sensory-driven activity during development. Science 2020; 370:370/6514/eabb2153. [PMID: 33060328 DOI: 10.1126/science.abb2153] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
At the earliest developmental stages, spontaneous activity synchronizes local and large-scale cortical networks. These networks form the functional template for the establishment of global thalamocortical networks and cortical architecture. The earliest connections are established autonomously. However, activity from the sensory periphery reshapes these circuits as soon as afferents reach the cortex. The early-generated, largely transient neurons of the subplate play a key role in integrating spontaneous and sensory-driven activity. Early pathological conditions-such as hypoxia, inflammation, or exposure to pharmacological compounds-alter spontaneous activity patterns, which subsequently induce disturbances in cortical network activity. This cortical dysfunction may lead to local and global miswiring and, at later stages, can be associated with neurological and psychiatric conditions.
Collapse
Affiliation(s)
- Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford OX1 3PT, UK.
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz 55128, Germany.
| | - Patrick O Kanold
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, 720 Rutland Avenue, MRB 379, Baltimore, MD 21205, USA. .,Johns Hopkins University Kavli Neuroscience Discovery Institute, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Luhmann HJ, Fukuda A. Can we understand human brain development from experimental studies in rodents? Pediatr Int 2020; 62:1139-1144. [PMID: 32531857 DOI: 10.1111/ped.14339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/05/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
Animal models are needed to gain an understanding of the genetic, molecular, cellular, and network mechanisms of human brain development. In rodents, a large spectrum of in vitro and in vivo approaches allows detailed analyses and specific experimental manipulations for studying the sequence of developmental steps in corticogenesis. Neurogenesis, neuronal migration, cellular differentiation, programmed cell death, synaptogenesis, and myelination are surprisingly similar in the rodent cortex and the human cortex. Spontaneous EEG activity in the pre- and early postnatal human cortex resembles the activity patterns recorded with intracortical multi-electrode arrays in newborn rodents. This early activity is generated by thalamic activation of a subplate-driven local network coupled via gap junctions, which controls the development of cortical columns and the spatio-temporal pattern of apoptosis. Disturbances of this activity may induce disturbances in cortical structure and function leading to neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Atsuo Fukuda
- Department of Physiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
23
|
Developmental GABA polarity switch and neuronal plasticity in Bioengineered Neuronal Organoids. Nat Commun 2020; 11:3791. [PMID: 32728089 PMCID: PMC7391775 DOI: 10.1038/s41467-020-17521-w] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are promising tools for disease modeling and drug development. For proper neuronal network formation excitatory and inhibitory neurons as well as glia need to co-develop. Here, we report the directed self-organization of human induced pluripotent stem cells in a collagen hydrogel towards a highly interconnected neuronal network at a macroscale tissue format. Bioengineered Neuronal Organoids (BENOs) comprise interconnected excitatory and inhibitory neurons with supportive astrocytes and oligodendrocytes. Giant depolarizing potential (GDP)-like events observed in early BENO cultures mimic early network activity of the fetal brain. The observed GABA polarity switch and reduced GDPs in >40 day BENO indicate progressive neuronal network maturation. BENOs demonstrate expedited complex network burst development after two months and evidence for long-term potentiation. The similarity of structural and functional properties to the fetal brain may allow for the application of BENOs in studies of neuronal plasticity and modeling of disease. Brain organoids are important tools to study early development and disease but little is known of their network activity and plasticity. Here the authors generate iPSC-derived neuronal organoids that display early network formation and maturation with evidence for a GABA polarity switch and long-term potentiation.
Collapse
|
24
|
Kostović I. The enigmatic fetal subplate compartment forms an early tangential cortical nexus and provides the framework for construction of cortical connectivity. Prog Neurobiol 2020; 194:101883. [PMID: 32659318 DOI: 10.1016/j.pneurobio.2020.101883] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/05/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022]
Abstract
The most prominent transient compartment of the primate fetal cortex is the deep, cell-sparse, synapse-containing subplate compartment (SPC). The developmental role of the SPC and its extraordinary size in humans remain enigmatic. This paper evaluates evidence on the development and connectivity of the SPC and discusses its role in the pathogenesis of neurodevelopmental disorders. A synthesis of data shows that the subplate becomes a prominent compartment by its expansion from the deep cortical plate (CP), appearing well-delineated on MR scans and forming a tangential nexus across the hemisphere, consisting of an extracellular matrix, randomly distributed postmigratory neurons, multiple branches of thalamic and long corticocortical axons. The SPC generates early spontaneous non-synaptic and synaptic activity and mediates cortical response upon thalamic stimulation. The subplate nexus provides large-scale interareal connectivity possibly underlying fMR resting-state activity, before corticocortical pathways are established. In late fetal phase, when synapses appear within the CP, transient the SPC coexists with permanent circuitry. The histogenetic role of the SPC is to provide interactive milieu and capacity for guidance, sorting, "waiting" and target selection of thalamocortical and corticocortical pathways. The new evolutionary role of the SPC and its remnant white matter neurons is linked to the increasing number of associative pathways in the human neocortex. These roles attributed to the SPC are regulated using a spatiotemporal gene expression during critical periods, when pathogenic factors may disturb vulnerable circuitry of the SPC, causing neurodevelopmental cognitive circuitry disorders.
Collapse
Affiliation(s)
- Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, Salata 12, 10000 Zagreb, Croatia.
| |
Collapse
|
25
|
Khakipoor S, Crouch EE, Mayer S. Human organoids to model the developing human neocortex in health and disease. Brain Res 2020; 1742:146803. [PMID: 32240655 DOI: 10.1016/j.brainres.2020.146803] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/28/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
Rodent models have catalyzed major discoveries in the neocortex, a brain region unique to mammals. However, since the neocortex has expanded considerably in primates, employing rodent models has limitations. Human fetal brain tissue is a scarce resource with limitations for experimental manipulations. In order to create an experimentally tractable representation of human brain development, a number of labs have recently created in vitro models of the developing human brain. These models, generated using human embryonic stem cells or induced pluripotent stem cells, are called "organoids". Organoids have successfully and rapidly uncovered new mechanisms of human brain development in health and disease. In the future, we envision that this strategy will enable faster and more efficient translation of basic neuroscience findings to therapeutic applications. In this review, we discuss the generation of the first human cerebral organoids, progress since their debut, and challenges to be overcome in the future.
Collapse
Affiliation(s)
- Shokoufeh Khakipoor
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Elizabeth E Crouch
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Simone Mayer
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
26
|
Ohtaka-Maruyama C. Subplate Neurons as an Organizer of Mammalian Neocortical Development. Front Neuroanat 2020; 14:8. [PMID: 32265668 PMCID: PMC7103628 DOI: 10.3389/fnana.2020.00008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 02/20/2020] [Indexed: 12/30/2022] Open
Abstract
Subplate neurons (SpNs) are one of the earliest born and matured neurons in the developing cerebral cortex and play an important role in the early development of the neocortex. It has been known that SpNs have an essential role in thalamocortical axon (TCA) pathfinding and the establishment of the first neural circuit from the thalamus towards cortical layer IV. In addition to this function, it has recently been revealed in mouse corticogenesis that SpNs play an important role in the regulation of radial neuronal migration during the mid-embryonic stage. Moreover, accumulating studies throw light on the possible roles of SpNs in adult brain functions and also their involvement in psychiatric or other neurological disorders. As SpNs are unique to mammals, they may have contributed to the evolution of the mammalian neocortex by efficiently organizing cortical formation during the limited embryonic period of corticogenesis. By increasing our knowledge of the functions of SpNs, we will clarify how SpNs act as an organizer of mammalian neocortical formation.
Collapse
Affiliation(s)
- Chiaki Ohtaka-Maruyama
- Neural Network Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
27
|
Kocovic DM, Limaye PV, Colburn LCH, Singh MB, Milosevic MM, Tadic J, Petronijevic M, Vrzic-Petronijevic S, Andjus PR, Antic SD. Cadmium versus Lanthanum Effects on Spontaneous Electrical Activity and Expression of Connexin Isoforms Cx26, Cx36, and Cx45 in the Human Fetal Cortex. Cereb Cortex 2020; 30:1244-1259. [PMID: 31408166 PMCID: PMC7132928 DOI: 10.1093/cercor/bhz163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/25/2019] [Accepted: 06/25/2019] [Indexed: 12/29/2022] Open
Abstract
Electrical activity is important for brain development. In brain slices, human subplate neurons exhibit spontaneous electrical activity that is highly sensitive to lanthanum. Based on the results of pharmacological experiments in human fetal tissue, we hypothesized that hemichannel-forming connexin (Cx) isoforms 26, 36, and 45 would be expressed on neurons in the subplate (SP) zone. RNA sequencing of dissected human cortical mantles at ages of 17-23 gestational weeks revealed that Cx45 has the highest expression, followed by Cx36 and Cx26. The levels of Cx and pannexin expression between male and female fetal cortices were not significantly different. Immunohistochemical analysis detected Cx45- and Cx26-expressing neurons in the upper segment of the SP zone. Cx45 was present on the cell bodies of human SP neurons, while Cx26 was found on both cell bodies and dendrites. Cx45, Cx36, and Cx26 were strongly expressed in the cortical plate, where newborn migrating neurons line up to form cortical layers. New information about the expression of 3 "neuronal" Cx isoforms in each cortical layer/zone (e.g., SP, cortical plate) and pharmacological data with cadmium and lanthanum may improve our understanding of the cellular mechanisms underlying neuronal development in human fetuses and potential vulnerabilities.
Collapse
Affiliation(s)
- Dusica M Kocovic
- Faculty of Biology, University of Belgrade, Belgrade 11000, Serbia
| | - Pallavi V Limaye
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Lauren C H Colburn
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Mandakini B Singh
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Milena M Milosevic
- Faculty of Biology, University of Belgrade, Belgrade 11000, Serbia
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Jasmina Tadic
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | | | | | - Pavle R Andjus
- Faculty of Biology, University of Belgrade, Belgrade 11000, Serbia
| | - Srdjan D Antic
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
28
|
Ghazizadeh Z, Kiviniemi T, Olafsson S, Plotnick D, Beerens ME, Zhang K, Gillon L, Steinbaugh MJ, Barrera V, Sui SH, Werdich AA, Kapur S, Eranti A, Gunn J, Jalkanen J, Airaksinen J, Kleber AG, Hollmén M, MacRae CA. Metastable Atrial State Underlies the Primary Genetic Substrate for MYL4 Mutation-Associated Atrial Fibrillation. Circulation 2019; 141:301-312. [PMID: 31735076 DOI: 10.1161/circulationaha.119.044268] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common clinical arrhythmia and is associated with heart failure, stroke, and increased mortality. The myocardial substrate for AF is poorly understood because of limited access to primary human tissue and mechanistic questions around existing in vitro or in vivo models. METHODS Using an MYH6:mCherry knock-in reporter line, we developed a protocol to generate and highly purify human pluripotent stem cell-derived cardiomyocytes displaying physiological and molecular characteristics of atrial cells. We modeled human MYL4 mutants, one of the few definitive genetic causes of AF. To explore non-cell-autonomous components of AF substrate, we also created a zebrafish Myl4 knockout model, which exhibited molecular, cellular, and physiologic abnormalities that parallel those in humans bearing the cognate mutations. RESULTS There was evidence of increased retinoic acid signaling in both human embryonic stem cells and zebrafish mutant models, as well as abnormal expression and localization of cytoskeletal proteins, and loss of intracellular nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide + hydrogen. To identify potentially druggable proximate mechanisms, we performed a chemical suppressor screen integrating multiple human cellular and zebrafish in vivo endpoints. This screen identified Cx43 (connexin 43) hemichannel blockade as a robust suppressor of the abnormal phenotypes in both models of MYL4 (myosin light chain 4)-related atrial cardiomyopathy. Immunofluorescence and coimmunoprecipitation studies revealed an interaction between MYL4 and Cx43 with altered localization of Cx43 hemichannels to the lateral membrane in MYL4 mutants, as well as in atrial biopsies from unselected forms of human AF. The membrane fraction from MYL4-/- human embryonic stem cell derived atrial cells demonstrated increased phospho-Cx43, which was further accentuated by retinoic acid treatment and by the presence of risk alleles at the Pitx2 locus. PKC (protein kinase C) was induced by retinoic acid, and PKC inhibition also rescued the abnormal phenotypes in the atrial cardiomyopathy models. CONCLUSIONS These data establish a mechanistic link between the transcriptional, metabolic and electrical pathways previously implicated in AF substrate and suggest novel avenues for the prevention or therapy of this common arrhythmia.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Tuomas Kiviniemi
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Sigurast Olafsson
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David Plotnick
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Manu E Beerens
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kun Zhang
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Leah Gillon
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Victor Barrera
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Shannan Ho Sui
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Andreas A Werdich
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sunil Kapur
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Antti Eranti
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Jarmo Gunn
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Juho Jalkanen
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Juhani Airaksinen
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Andre G Kleber
- Department of Pathology, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, MA (A.G.K.)
| | - Maija Hollmén
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Calum A MacRae
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Genetics and Network Medicine Divisions (C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Harvard Stem Cell Institute, Boston, MA (C.A.M.)
| |
Collapse
|
29
|
Singh MB, White JA, McKimm EJ, Milosevic MM, Antic SD. Mechanisms of Spontaneous Electrical Activity in the Developing Cerebral Cortex-Mouse Subplate Zone. Cereb Cortex 2019; 29:3363-3379. [PMID: 30169554 PMCID: PMC7963116 DOI: 10.1093/cercor/bhy205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/28/2018] [Accepted: 08/05/2018] [Indexed: 12/15/2022] Open
Abstract
Subplate (SP) neurons exhibit spontaneous plateau depolarizations mediated by connexin hemichannels. Postnatal (P1-P6) mice show identical voltage pattern and drug-sensitivity as observed in slices from human fetal cortex; indicating that the mouse is a useful model for studying the cellular physiology of the developing neocortex. In mouse SP neurons, spontaneous plateau depolarizations were insensitive to blockers of: synaptic transmission (glutamatergic, GABAergic, or glycinergic), pannexins (probenecid), or calcium channels (mibefradil, verapamil, diltiazem); while highly sensitive to blockers of gap junctions (octanol), hemichannels (La3+, lindane, Gd3+), or glial metabolism (DLFC). Application of La3+ (100 μM) does not exert its effect on electrical activity by blocking calcium channels. Intracellular application of Gd3+ determined that Gd3+-sensitive pores (putative connexin hemichannels) reside on the membrane of SP neurons. Immunostaining of cortical sections (P1-P6) detected connexins 26, and 45 in neurons, but not connexins 32 and 36. Vimentin-positive glial cells were detected in the SP zone suggesting a potential physiological interaction between SP neurons and radial glia. SP spontaneous activity was reduced by blocking glial metabolism with DFLC or by blocking purinergic receptors by PPADS. Connexin hemichannels and ATP release from vimentin-positive glial cells may underlie spontaneous plateau depolarizations in the developing mammalian cortex.
Collapse
Affiliation(s)
- Mandakini B Singh
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT, USA
| | - Jesse A White
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT, USA
| | - Eric J McKimm
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT, USA
| | - Milena M Milosevic
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT, USA
| | - Srdjan D Antic
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT, USA
| |
Collapse
|
30
|
Multimodal Single-Cell Analysis Reveals Physiological Maturation in the Developing Human Neocortex. Neuron 2019; 102:143-158.e7. [PMID: 30770253 DOI: 10.1016/j.neuron.2019.01.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/20/2018] [Accepted: 01/14/2019] [Indexed: 12/21/2022]
Abstract
In the developing human neocortex, progenitor cells generate diverse cell types prenatally. Progenitor cells and newborn neurons respond to signaling cues, including neurotransmitters. While single-cell RNA sequencing has revealed cellular diversity, physiological heterogeneity has yet to be mapped onto these developing and diverse cell types. By combining measurements of intracellular Ca2+ elevations in response to neurotransmitter receptor agonists and RNA sequencing of the same single cells, we show that Ca2+ responses are cell-type-specific and change dynamically with lineage progression. Physiological response properties predict molecular cell identity and additionally reveal diversity not captured by single-cell transcriptomics. We find that the serotonin receptor HTR2A selectively activates radial glia cells in the developing human, but not mouse, neocortex, and inhibiting HTR2A receptors in human radial glia disrupts the radial glial scaffold. We show highly specific neurotransmitter signaling during neurogenesis in the developing human neocortex and highlight evolutionarily divergent mechanisms of physiological signaling.
Collapse
|
31
|
Kostović I, Išasegi IŽ, Krsnik Ž. Sublaminar organization of the human subplate: developmental changes in the distribution of neurons, glia, growing axons and extracellular matrix. J Anat 2018; 235:481-506. [PMID: 30549027 DOI: 10.1111/joa.12920] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2018] [Indexed: 12/21/2022] Open
Abstract
The objective of this paper was to collect normative data essential for analyzing the subplate (SP) role in pathogenesis of developmental disorders, characterized by abnormal circuitry, such as hypoxic-ischemic lesions, autism and schizophrenia. The main cytological features of the SP, such as low cell density, early differentiation of neurons and glia, plexiform arrangement of axons and dendrites, presence of synapses and a large amount of extracellular matrix (ECM) distinguish this compartment from the cell-dense cortical plate (CP; towards pia) and large fiber bundles of external axonal strata of fetal white matter (towards ventricle). For SP delineation from these adjacent layers based on combined cytological criteria, we analyzed the sublaminar distribution of different microstructural elements and the associated maturational gradients throughout development, using immunocytochemical and histological techniques on postmortem brain material (Zagreb Neuroembryological Collection). The analysis revealed that the SP compartment of the lateral neocortex shows changes in laminar organization throughout fetal development: the monolayer in the early fetal period (presubplate) undergoes dramatic bilaminar transformation between 13 and 15 postconceptional weeks (PCW), followed by subtle sublamination in three 'floors' (deep, intermediate, superficial) of midgestation (15-21 PCW). During the stationary phase (22-28 PCW), SP persists as a trilaminar compartment, gradually losing its sublaminar organization towards the end of gestation and remains as a single layer of SP remnant in the newborn brain. Based on these sublaminar transformations, we have documented developmental changes in the distribution, maturational gradients and expression of molecular markers in SP synapses, transitional forms of astroglia, neurons and ECM, which occur concomitantly with the ingrowth of thalamo-cortical, basal forebrain and cortico-cortical axons in a deep to superficial fashion. The deep SP is the zone of ingrowing axons - 'entrance (ingrowth) zone'. The process of axonal ingrowth begins with thalamo-cortical fibers and basal forebrain afferents, indicating an oblique geometry. During the later fetal period, deep SP receives long cortico-cortical axons exhibiting a tangential geometry. Intermediate SP ('proper') is the navigation and 'nexus' sublamina consisting of a plexiform arrangement of cellular elements providing guidance and substrate for axonal growth, and also containing transient connectivity of dendrites and axons in a tangential plane without radial boundaries immersed in an ECM-rich continuum. Superficial SP is the axonal accumulation ('waiting compartment') and target selection zone, indicating a dense distribution of synaptic markers, accumulation of thalamo-cortical axons (around 20 PCW), overlapping with dendrites from layer VI neurons. In the late preterm brain period, superficial SP contains a chondroitin sulfate non-immunoreactive band. The developmental dynamics for the distribution of neuronal, glial and ECM markers comply with sequential ingrowth of afferents in three levels of SP: ECM and synaptic markers shift from deep to superficial SP, with transient forms of glia following this arrangement, and calretinin neurons are concentrated in the SP during the formation phase. These results indicate developmental and morphogenetic roles in the SP cellular (transient glia, neurons and synapses) and ECM framework, enabling the spatial accommodation, navigation and establishment of numerous connections of cortical pathways in the expanded human brain. The original findings of early developmental dynamics of transitional subtypes of astroglia, calretinin neurons, ECM and synaptic markers presented in the SP are interesting in the light of recent concepts concerning its functional and morphogenetic role and an increasing interest in SP as a prospective substrate of abnormalities in cortical circuitry, leading to a cognitive deficit in different neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Iris Žunić Išasegi
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| |
Collapse
|
32
|
Luhmann HJ, Kirischuk S, Kilb W. The Superior Function of the Subplate in Early Neocortical Development. Front Neuroanat 2018; 12:97. [PMID: 30487739 PMCID: PMC6246655 DOI: 10.3389/fnana.2018.00097] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022] Open
Abstract
During early development the structure and function of the cerebral cortex is critically organized by subplate neurons (SPNs), a mostly transient population of glutamatergic and GABAergic neurons located below the cortical plate. At the molecular and morphological level SPNs represent a rather diverse population of cells expressing a variety of genetic markers and revealing different axonal-dendritic morphologies. Electrophysiologically SPNs are characterized by their rather mature intrinsic membrane properties and firing patterns. They are connected via electrical and chemical synapses to local and remote neurons, e.g., thalamic relay neurons forming the first thalamocortical input to the cerebral cortex. Therefore SPNs are robustly activated at pre- and perinatal stages by the sensory periphery. Although SPNs play pivotal roles in early neocortical activity, development and plasticity, they mostly disappear by programmed cell death during further maturation. On the one hand, SPNs may be selectively vulnerable to hypoxia-ischemia contributing to brain damage, on the other hand there is some evidence that enhanced survival rates or alterations in SPN distribution may contribute to the etiology of neurological or psychiatric disorders. This review aims to give a comprehensive and up-to-date overview on the many functions of SPNs during early physiological and pathophysiological development of the cerebral cortex.
Collapse
Affiliation(s)
- Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
33
|
Ortiz-Escribano N, Szymanska KJ, Bol M, Vandenberghe L, Decrock E, Van Poucke M, Peelman L, Van den Abbeel E, Van Soom A, Leybaert L. Blocking connexin channels improves embryo development of vitrified bovine blastocysts. Biol Reprod 2018; 96:288-301. [PMID: 28203704 DOI: 10.1095/biolreprod.116.144121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/01/2016] [Accepted: 01/03/2017] [Indexed: 01/13/2023] Open
Abstract
Connexins (Cxs) are required for normal embryo development and implantation. They form gap junctions (GJs) connecting the cytoplasm of adjacent cells and hemichannels (HCs), which are normally closed but open in response to stress conditions. Excessive HC opening is detrimental for cell function and may lead to cell death. We found that hatching of in vitro-produced bovine embryos, matured in serum-containing conditions, was significantly improved when vitrification/warming was done in the presence of Gap26 that targets GJA1 (Cx43) and GJA4 (Cx37). Further work showed that HCs from blastocysts produced after oocyte maturation in the presence of serum were open shortly after vitrification/warming, and this was prevented by Gap26. Gap26, applied for the exposure times used, inhibited Cx43 and Cx37 HCs while it did not have an effect on GJs. Interestingly, Gap26 had no effect on blastocyst degeneration or cell death. We conclude that blocking HCs protects embryos during vitrification and warming by a functional effect not linked to cell death.
Collapse
Affiliation(s)
| | | | - Melissa Bol
- Physiology group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - Lynn Vandenberghe
- Reproduction, Obstetrics and Herd Health, Ghent University, Merelbeke, Belgium
| | - Elke Decrock
- Physiology group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - Mario Van Poucke
- Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, Belgium
| | - Luc Peelman
- Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, Belgium
| | | | - Ann Van Soom
- Reproduction, Obstetrics and Herd Health, Ghent University, Merelbeke, Belgium
| | - Luc Leybaert
- Physiology group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
34
|
Mäkinen MEL, Ylä-Outinen L, Narkilahti S. GABA and Gap Junctions in the Development of Synchronized Activity in Human Pluripotent Stem Cell-Derived Neural Networks. Front Cell Neurosci 2018; 12:56. [PMID: 29559893 PMCID: PMC5845705 DOI: 10.3389/fncel.2018.00056] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/16/2018] [Indexed: 01/03/2023] Open
Abstract
The electrical activity of the brain arises from single neurons communicating with each other. However, how single neurons interact during early development to give rise to neural network activity remains poorly understood. We studied the emergence of synchronous neural activity in human pluripotent stem cell (hPSC)-derived neural networks simultaneously on a single-neuron level and network level. The contribution of gamma-aminobutyric acid (GABA) and gap junctions to the development of synchronous activity in hPSC-derived neural networks was studied with GABA agonist and antagonist and by blocking gap junctional communication, respectively. We characterized the dynamics of the network-wide synchrony in hPSC-derived neural networks with high spatial resolution (calcium imaging) and temporal resolution microelectrode array (MEA). We found that the emergence of synchrony correlates with a decrease in very strong GABA excitation. However, the synchronous network was found to consist of a heterogeneous mixture of synchronously active cells with variable responses to GABA, GABA agonists and gap junction blockers. Furthermore, we show how single-cell distributions give rise to the network effect of GABA, GABA agonists and gap junction blockers. Finally, based on our observations, we suggest that the earliest form of synchronous neuronal activity depends on gap junctions and a decrease in GABA induced depolarization but not on GABAA mediated signaling.
Collapse
Affiliation(s)
- Meeri Eeva-Liisa Mäkinen
- NeuroGroup Laboratory, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Laura Ylä-Outinen
- NeuroGroup Laboratory, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup Laboratory, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| |
Collapse
|
35
|
Pierozan P, Biasibetti-Brendler H, Schmitz F, Ferreira F, Netto CA, Wyse ATS. Synergistic Toxicity of the Neurometabolites Quinolinic Acid and Homocysteine in Cortical Neurons and Astrocytes: Implications in Alzheimer's Disease. Neurotox Res 2017; 34:147-163. [PMID: 29124681 DOI: 10.1007/s12640-017-9834-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/22/2017] [Accepted: 10/18/2017] [Indexed: 12/19/2022]
Abstract
The brain of patients affected by Alzheimer's disease (AD) develops progressive neurodegeneration linked to the formation of proteins aggregates. However, their single actions cannot explain the extent of brain damage observed in this disorder, and the characterization of co-adjuvant involved in the early toxic processes evoked in AD is essential. In this line, quinolinic acid (QUIN) and homocysteine (Hcy) appear to be involved in the AD neuropathogenesis. Herein, we investigate the effects of QUIN and Hcy on early toxic events in cortical neurons and astrocytes. Exposure of primary cortical cultures to these neurometabolites for 24 h induced concentration-dependent neurotoxicity. In addition, QUIN (25 μM) and Hcy (30 μM) triggered ROS production, lipid peroxidation, diminished of Na+,K+-ATPase activity, and morphologic alterations, culminating in reduced neuronal viability by necrotic cell death. In astrocytes, QUIN (100 μM) and Hcy (30 μM) induced caspase-3-dependent apoptosis and morphologic alterations through oxidative status imbalance. To establish specific mechanisms, we preincubated cell cultures with different protective agents. The combined toxicity of QUIN and Hcy was attenuated by melatonin and Trolox in neurons and by NMDA antagonists and glutathione in astrocytes. Cellular death and morphologic alterations were prevented when co-culture was treated with metabolites, suggesting the activation of protector mechanisms dependent on soluble factors and astrocyte and neuron communication through gap junctions. These findings suggest that early damaging events involved in AD can be magnified by synergistic toxicity of the QUIN and Hcy. Therefore, this study opens new possibilities to elucidate the molecular mechanisms of neuron-astrocyte interactions and their role in neuroprotection against QUIN and Hcy.
Collapse
Affiliation(s)
- Paula Pierozan
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil.
| | - Helena Biasibetti-Brendler
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felipe Schmitz
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Ferreira
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Laboratório de Isquemia Cerebral e Psicobiologia dos Transtornos Mentais, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| | - Angela T S Wyse
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| |
Collapse
|
36
|
Rich MT, Torregrossa MM. Molecular and synaptic mechanisms regulating drug-associated memories: Towards a bidirectional treatment strategy. Brain Res Bull 2017; 141:58-71. [PMID: 28916448 DOI: 10.1016/j.brainresbull.2017.09.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/21/2017] [Accepted: 09/05/2017] [Indexed: 12/11/2022]
Abstract
The successful treatment of substance use disorders is dependent on the establishment of a long-term abstinent state. Relapse can be suppressed by interfering with memories of drug use that are evoked by re-exposure to drug-associated contexts and cues. Two strategies for accomplishing this goal are either to prevent drug-memory reconsolidation or to induce the formation of a competing, extinction memory. However, clinical attempts to prolong abstinence by behavioral modification of drug-related memories have had limited success. One approach to improve behavioral treatment strategies is to identify the molecular mechanisms that regulate these memory processes and then use pharmacological tools as supplements to improve efficacy. Still, due to the involvement of several overlapping signaling cascades in both reconsolidation and extinction, it is difficult to specifically modify one of the two processes. For example, attempting to elicit extinction may instead initiate reconsolidation, resulting in the unintentional strengthening of drug-related memories. A better approach is to identify diverging components of the two processes, whereby a single medication would simultaneously weaken reconsolidation and enhance extinction. This review will provide an overview of the neural substrates that are involved in the regulation of drug-associated memories, and will discuss emerging approaches to pharmacologically weaken these memories, including recent efforts to precisely and bidirectionally target reconsolidation and extinction. Ultimately, pharmacologically-enhanced memory-based approaches have the potential to produce more informed relapse-prevention therapies.
Collapse
Affiliation(s)
- Matthew T Rich
- Department of Psychiatry, University of Pittsburgh, 3811 O'Hara St., Pittsburgh, PA 15213, United States; Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, 4400 Fifth Ave, Pittsburgh, PA, 15213, United States.
| | - Mary M Torregrossa
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, United States.
| |
Collapse
|
37
|
Majoul IV, Ernesti JS, Butkevich EV, Duden R. Drebrins and Connexins: A Biomedical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:225-247. [DOI: 10.1007/978-4-431-56550-5_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
38
|
Kaminska A, Delattre V, Laschet J, Dubois J, Labidurie M, Duval A, Manresa A, Magny JF, Hovhannisyan S, Mokhtari M, Ouss L, Boissel A, Hertz-Pannier L, Sintsov M, Minlebaev M, Khazipov R, Chiron C. Cortical Auditory-Evoked Responses in Preterm Neonates: Revisited by Spectral and Temporal Analyses. Cereb Cortex 2017; 28:3429-3444. [DOI: 10.1093/cercor/bhx206] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Indexed: 11/12/2022] Open
Affiliation(s)
- A Kaminska
- INSERM U1129, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- CEA, 91191 Gif sur Yvette, France
- Department of Clinical Neurophysiology, AP-HP, Necker-Enfants Malades Hospital, Paris, France
| | - V Delattre
- INSERM U1129, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- CEA, 91191 Gif sur Yvette, France
- Neurospin, UNIACT, CEA, Gif sur Yvette, France
| | - J Laschet
- INSERM U1129, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- CEA, 91191 Gif sur Yvette, France
| | - J Dubois
- INSERM U992, CEA/DRF/I2BM/Neurospin/UNICOG, Gif-sur-Yvette, France
- Paris Saclay University, Paris-Sud University, Gif-sur-Yvette, France
| | - M Labidurie
- INSERM U1129, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- CEA, 91191 Gif sur Yvette, France
| | - A Duval
- INSERM U1129, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- CEA, 91191 Gif sur Yvette, France
- Neurospin, UNIACT, CEA, Gif sur Yvette, France
| | - A Manresa
- Laboratory of Psychology and Neurosciences (LPN) (EA 47000), Rouen University, Rouen, France
| | - J -F Magny
- Neonatal Intensive Care Unit, AP-HP, Necker-Enfants Malades Hospital, Paris, France
| | - S Hovhannisyan
- Neonatal Intensive Care Unit, AP-HP, Necker-Enfants Malades Hospital, Paris, France
| | - M Mokhtari
- Neonatal Intensive Care Unit, AP-HP, Bicetre Hospital, Kremlin-Bicetre, France
| | - L Ouss
- Department of Pediatric Neurology, AP-HP, Necker-Enfants Malades Hospital, Paris, France
| | - A Boissel
- Laboratory of Psychology and Neurosciences (LPN) (EA 47000), Rouen University, Rouen, France
| | - L Hertz-Pannier
- INSERM U1129, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- CEA, 91191 Gif sur Yvette, France
- Neurospin, UNIACT, CEA, Gif sur Yvette, France
| | - M Sintsov
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - M Minlebaev
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
- INSERM U901/ INMED, Aix-Marseille University, Marseille, France
| | - R Khazipov
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
- INSERM U901/ INMED, Aix-Marseille University, Marseille, France
| | - C Chiron
- INSERM U1129, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- CEA, 91191 Gif sur Yvette, France
| |
Collapse
|
39
|
Meyer G, González-Gómez M. The Subpial Granular Layer and Transient Versus Persisting Cajal-Retzius Neurons of the Fetal Human Cortex. Cereb Cortex 2017; 28:2043-2058. [DOI: 10.1093/cercor/bhx110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/18/2017] [Indexed: 12/20/2022] Open
Affiliation(s)
- Gundela Meyer
- Units of Anatomy (MGG) and Histology (GM), Department of Basic Medical Science, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Miriam González-Gómez
- Units of Anatomy (MGG) and Histology (GM), Department of Basic Medical Science, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| |
Collapse
|
40
|
Neonatal CX26 removal impairs neocortical development and leads to elevated anxiety. Proc Natl Acad Sci U S A 2017; 114:3228-3233. [PMID: 28265099 DOI: 10.1073/pnas.1613237114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Electrical coupling between excitatory neurons in the neocortex is developmentally regulated. It is initially prominent but eliminated at later developmental stages when chemical synapses emerge. However, it remains largely unclear whether early electrical coupling networks broadly contribute to neocortical circuit formation and animal behavior. Here, we report that neonatal electrical coupling between neocortical excitatory neurons is critical for proper neuronal development, synapse formation, and animal behavior. Conditional deletion of Connexin 26 (CX26) in the superficial layer excitatory neurons of the mouse neocortex around birth significantly reduces spontaneous firing activity and the frequency and size of spontaneous network oscillations at postnatal day 5-6. Moreover, CX26-conditional knockout (CX26-cKO) neurons tend to have simpler dendritic trees and lower spine density compared with wild-type neurons. Importantly, early, but not late, postnatal deletion of CX26, decreases the frequency of miniature excitatory postsynaptic currents (mEPSCs) in both young and adult mice, whereas miniature inhibitory postsynaptic currents (mIPSCs) were unaffected. Furthermore, CX26-cKO mice exhibit increased anxiety-related behavior. These results suggest that electrical coupling between excitatory neurons at early postnatal stages is a critical step for neocortical development and function.
Collapse
|
41
|
Evolution of Osteocrin as an activity-regulated factor in the primate brain. Nature 2016; 539:242-247. [PMID: 27830782 DOI: 10.1038/nature20111] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 09/19/2016] [Indexed: 11/08/2022]
Abstract
Sensory stimuli drive the maturation and function of the mammalian nervous system in part through the activation of gene expression networks that regulate synapse development and plasticity. These networks have primarily been studied in mice, and it is not known whether there are species- or clade-specific activity-regulated genes that control features of brain development and function. Here we use transcriptional profiling of human fetal brain cultures to identify an activity-dependent secreted factor, Osteocrin (OSTN), that is induced by membrane depolarization of human but not mouse neurons. We find that OSTN has been repurposed in primates through the evolutionary acquisition of DNA regulatory elements that bind the activity-regulated transcription factor MEF2. In addition, we demonstrate that OSTN is expressed in primate neocortex and restricts activity-dependent dendritic growth in human neurons. These findings suggest that, in response to sensory input, OSTN regulates features of neuronal structure and function that are unique to primates.
Collapse
|
42
|
Calderón JF, Retamal MA. Regulation of Connexins Expression Levels by MicroRNAs, an Update. Front Physiol 2016; 7:558. [PMID: 27932990 PMCID: PMC5122916 DOI: 10.3389/fphys.2016.00558] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/04/2016] [Indexed: 12/13/2022] Open
Abstract
Control of cell-cell coordination and communication is regulated by several factors, including paracrine and autocrine release of biomolecules, and direct exchange of soluble factors between cells through gap junction channels. Additionally, hemichannels also participate in cell-cell coordination through the release of signaling molecules, such as ATP and glutamate. A family of transmembrane proteins named connexins forms both gap junction channels and hemichannels. Because of their importance in cell and tissue coordination, connexins are controlled both by post-translational and post-transcriptional modifications. In recent years, non-coding RNAs have garnered research interest due to their ability to exert post-transcriptional regulation of gene expression. One of the most recent, well-documented control mechanisms of protein synthesis is found through the action of small, single-stranded RNA, called micro RNAs (miRNAs or miRs). Put simply, miRNAs are negative regulators of the expression of a myriad proteins involved in many physiological and pathological processes. This mini review will briefly summarize what is currently known about the action of miRNAs over Cxs expression/function in different organs under some relevant physiological and pathological conditions.
Collapse
Affiliation(s)
- Juan F Calderón
- Facultad de Medicina, Center for Genetics and Genomics, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Mauricio A Retamal
- Facultad de Medicina, Centro de Fisiología Celular e Integrativa, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| |
Collapse
|
43
|
Orellana JA. Physiological Functions of Glial Cell Hemichannels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:93-108. [DOI: 10.1007/978-3-319-40764-7_5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Gajardo-Gómez R, Labra VC, Orellana JA. Connexins and Pannexins: New Insights into Microglial Functions and Dysfunctions. Front Mol Neurosci 2016; 9:86. [PMID: 27713688 PMCID: PMC5031785 DOI: 10.3389/fnmol.2016.00086] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/05/2016] [Indexed: 12/11/2022] Open
Abstract
Under physiological conditions, microglia adopt a resting phenotype associated with the production of anti-inflammatory and neurotrophic factors. In response to a wide variety of insults, these cells shift to an activated phenotype that is necessary for the proper restoration of brain homeostasis. However, when the intensity of a threat is relatively high, microglial activation worsens the progression of damage rather than providing protection, with potentially significant consequences for neuronal survival. Coordinated interactions among microglia and other brain cells, including astrocytes and neurons, are critical for the development of timely and optimal inflammatory responses in the brain parenchyma. Tissue synchronization is in part mediated by connexins and pannexins, which are protein families that form different plasma membrane channels to communicate with neighboring cells. Gap junction channels (which are exclusively formed by connexins in vertebrates) connect the cytoplasm of contacting cells to coordinate electrical and metabolic coupling. Hemichannels (HCs) and pannexons (which are formed by connexins and pannexins, respectively) communicate the intra- and extracellular compartments and serve as diffusion pathways for the exchange of ions and small molecules. In this review article, we discuss the available evidence concerning the functional expression and regulation of connexin- and pannexin-based channels in microglia and their contributions to microglial function and dysfunction. Specifically, we focus on the possible implications of these channels in microglia-to-microglia, microglia-to-astrocyte and neuron-to-microglia interactions in the inflamed brain.
Collapse
Affiliation(s)
- Rosario Gajardo-Gómez
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Valeria C Labra
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
45
|
Orellana JA, Retamal MA, Moraga-Amaro R, Stehberg J. Role of Astroglial Hemichannels and Pannexons in Memory and Neurodegenerative Diseases. Front Integr Neurosci 2016; 10:26. [PMID: 27489539 PMCID: PMC4951483 DOI: 10.3389/fnint.2016.00026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/06/2016] [Indexed: 11/13/2022] Open
Abstract
Under physiological conditions, astroglial hemichannels and pannexons allow the release of gliotransmitters from astrocytes. These gliotransmitters are critical in modulating synaptic transmission, plasticity and memory. However, recent evidence suggests that under pathological conditions, they may be central in the development of various neurodegenerative diseases. Here we review current literature on the role of astroglial hemichannels and pannexons in memory, stress and the development of neurodegenerative diseases, and propose that they are not only crucial for normal brain function, including memory, but also a potential target for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Rodrigo Moraga-Amaro
- Laboratorio de Neurobiología, Centro de Investigaciones Biomédicas, Universidad Andres Bello Santiago, Chile
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Centro de Investigaciones Biomédicas, Universidad Andres Bello Santiago, Chile
| |
Collapse
|
46
|
Luhmann HJ, Sinning A, Yang JW, Reyes-Puerta V, Stüttgen MC, Kirischuk S, Kilb W. Spontaneous Neuronal Activity in Developing Neocortical Networks: From Single Cells to Large-Scale Interactions. Front Neural Circuits 2016; 10:40. [PMID: 27252626 PMCID: PMC4877528 DOI: 10.3389/fncir.2016.00040] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/06/2016] [Indexed: 11/13/2022] Open
Abstract
Neuronal activity has been shown to be essential for the proper formation of neuronal circuits, affecting developmental processes like neurogenesis, migration, programmed cell death, cellular differentiation, formation of local and long-range axonal connections, synaptic plasticity or myelination. Accordingly, neocortical areas reveal distinct spontaneous and sensory-driven neuronal activity patterns already at early phases of development. At embryonic stages, when immature neurons start to develop voltage-dependent channels, spontaneous activity is highly synchronized within small neuronal networks and governed by electrical synaptic transmission. Subsequently, spontaneous activity patterns become more complex, involve larger networks and propagate over several neocortical areas. The developmental shift from local to large-scale network activity is accompanied by a gradual shift from electrical to chemical synaptic transmission with an initial excitatory action of chloride-gated channels activated by GABA, glycine and taurine. Transient neuronal populations in the subplate (SP) support temporary circuits that play an important role in tuning early neocortical activity and the formation of mature neuronal networks. Thus, early spontaneous activity patterns control the formation of developing networks in sensory cortices, and disturbances of these activity patterns may lead to long-lasting neuronal deficits.
Collapse
Affiliation(s)
- Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz Mainz, Germany
| | - Anne Sinning
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz Mainz, Germany
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz Mainz, Germany
| | - Vicente Reyes-Puerta
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz Mainz, Germany
| | - Maik C Stüttgen
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz Mainz, Germany
| |
Collapse
|
47
|
Del Rio R, Quintanilla RA, Orellana JA, Retamal MA. Neuron-Glia Crosstalk in the Autonomic Nervous System and Its Possible Role in the Progression of Metabolic Syndrome: A New Hypothesis. Front Physiol 2015; 6:350. [PMID: 26648871 PMCID: PMC4664731 DOI: 10.3389/fphys.2015.00350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/09/2015] [Indexed: 01/26/2023] Open
Abstract
Metabolic syndrome (MS) is characterized by the following physiological alterations: increase in abdominal fat, insulin resistance, high concentration of triglycerides, low levels of HDL, high blood pressure, and a generalized inflammatory state. One of the pathophysiological hallmarks of this syndrome is the presence of neurohumoral activation, which involve autonomic imbalance associated to hyperactivation of the sympathetic nervous system. Indeed, enhanced sympathetic drive has been linked to the development of endothelial dysfunction, hypertension, stroke, myocardial infarct, and obstructive sleep apnea. Glial cells, the most abundant cells in the central nervous system, control synaptic transmission, and regulate neuronal function by releasing bioactive molecules called gliotransmitters. Recently, a new family of plasma membrane channels called hemichannels has been described to allow the release of gliotransmitters and modulate neuronal firing rate. Moreover, a growing amount of evidence indicates that uncontrolled hemichannel opening could impair glial cell functions, affecting synaptic transmission and neuronal survival. Given that glial cell functions are disturbed in various metabolic diseases, we hypothesize that progression of MS may relies on hemichannel-dependent impairment of glial-to-neuron communication by a mechanism related to dysfunction of inflammatory response and mitochondrial metabolism of glial cells. In this manuscript, we discuss how glial cells may contribute to the enhanced sympathetic drive observed in MS, and shed light about the possible role of hemichannels in this process.
Collapse
Affiliation(s)
- Rodrigo Del Rio
- Centro de Investigación Biomédica, Universidad Autónoma de Chile Santiago, Chile ; Dirección de Investigación, Universidad Científica del Sur Lima, Perú
| | | | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina. Clínica Alemana Universidad del Desarrollo Santiago, Chile
| |
Collapse
|
48
|
Rigas P, Adamos DA, Sigalas C, Tsakanikas P, Laskaris NA, Skaliora I. Spontaneous Up states in vitro: a single-metric index of the functional maturation and regional differentiation of the cerebral cortex. Front Neural Circuits 2015; 9:59. [PMID: 26528142 PMCID: PMC4603250 DOI: 10.3389/fncir.2015.00059] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/22/2015] [Indexed: 12/12/2022] Open
Abstract
Understanding the development and differentiation of the neocortex remains a central focus of neuroscience. While previous studies have examined isolated aspects of cellular and synaptic organization, an integrated functional index of the cortical microcircuit is still lacking. Here we aimed to provide such an index, in the form of spontaneously recurring periods of persistent network activity -or Up states- recorded in mouse cortical slices. These coordinated network dynamics emerge through the orchestrated regulation of multiple cellular and synaptic elements and represent the default activity of the cortical microcircuit. To explore whether spontaneous Up states can capture developmental changes in intracortical networks we obtained local field potential recordings throughout the mouse lifespan. Two independent and complementary methodologies revealed that Up state activity is systematically modified by age, with the largest changes occurring during early development and adolescence. To explore possible regional heterogeneities we also compared the development of Up states in two distinct cortical areas and show that primary somatosensory cortex develops at a faster pace than primary motor cortex. Our findings suggest that in vitro Up states can serve as a functional index of cortical development and differentiation and can provide a baseline for comparing experimental and/or genetic mouse models.
Collapse
Affiliation(s)
- Pavlos Rigas
- Neurophysiology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of AthensAthens, Greece
| | - Dimitrios A. Adamos
- Neuroinformatics Group, Aristotle University of ThessalonikiThessaloniki, Greece
- School of Music Studies, Aristotle University of ThessalonikiThessaloniki, Greece
| | - Charalambos Sigalas
- Neurophysiology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of AthensAthens, Greece
| | - Panagiotis Tsakanikas
- Neurophysiology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of AthensAthens, Greece
| | - Nikolaos A. Laskaris
- Neuroinformatics Group, Aristotle University of ThessalonikiThessaloniki, Greece
- AIIA Lab, Department of Informatics, Aristotle University of ThessalonikiThessaloniki, Greece
| | - Irini Skaliora
- Neurophysiology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of AthensAthens, Greece
| |
Collapse
|
49
|
Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC, Green CR, Leybaert L. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci 2015; 72:2823-51. [PMID: 26118660 PMCID: PMC11113968 DOI: 10.1007/s00018-015-1962-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.
Collapse
Affiliation(s)
- Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, 75231 Paris Cedex 05, France
- University Pierre et Marie
Curie, ED, N°158, 75005 Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Colin R. Green
- Department of Ophthalmology, The University of Auckland, Auckland, New Zealand
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| |
Collapse
|
50
|
Davidson J, Green C, Bennet L, Gunn A. Battle of the hemichannels – Connexins and Pannexins in ischemic brain injury. Int J Dev Neurosci 2014; 45:66-74. [DOI: 10.1016/j.ijdevneu.2014.12.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 12/20/2022] Open
Affiliation(s)
- J.O. Davidson
- Department of PhysiologyThe University of AucklandAucklandNew Zealand
| | - C.R. Green
- Department of OphthalmologyThe University of AucklandAucklandNew Zealand
| | - L. Bennet
- Department of PhysiologyThe University of AucklandAucklandNew Zealand
| | - A.J. Gunn
- Department of PhysiologyThe University of AucklandAucklandNew Zealand
| |
Collapse
|