1
|
Majeed M, Liao CP, Hobert O. Nervous system-wide analysis of all C. elegans cadherins reveals neuron-specific functions across multiple anatomical scales. SCIENCE ADVANCES 2025; 11:eads2852. [PMID: 39983000 PMCID: PMC11844738 DOI: 10.1126/sciadv.ads2852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/22/2025] [Indexed: 02/23/2025]
Abstract
Differential expression of cell adhesion proteins is a hallmark of cell-type diversity across the animal kingdom. Gene family-wide characterization of their organismal expression and function is, however, lacking. Using genome-engineered reporter alleles, we established an atlas of expression of the entire set of 12 cadherin gene family members in the nematode Caenorhabditis elegans, revealing differential expression across neuronal classes, a dichotomy between broadly and narrowly expressed cadherins, and several context-dependent temporal transitions in expression across development. Engineered mutant null alleles of cadherins were analyzed for defects in morphology, behavior, neuronal soma positions, neurite neighborhood topology and fasciculation, and localization of synapses in many parts of the nervous system. This analysis revealed a restricted pattern of neuronal differentiation defects at discrete subsets of anatomical scales, including a novel role of cadherins in experience-dependent electrical synapse formation. In total, our analysis results in previously little explored perspectives on cadherin deployment and function.
Collapse
Affiliation(s)
| | - Chien-Po Liao
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| |
Collapse
|
2
|
Zhang L, Zhu Y, Ren Y, Xu L, Liu X, Qi X, Jiao T, Sun G, Han H, Zhang J, Sun F, Yang Y, Zhao S. Genetic characterization of Tibetan pigs adapted to high altitude under natural selection based on a large whole-genome dataset. Sci Rep 2024; 14:17062. [PMID: 39048584 PMCID: PMC11269713 DOI: 10.1038/s41598-024-65559-3] [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: 01/27/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
The Qinghai-Tibet Plateau is a valuable genetic resource pool, and the high-altitude adaptation of Tibetan pigs is a classic example of the adaptive evolution of domestic animals. Here, we report the presence of Darwinian positive selection signatures in Tibetan pigs (TBPs) using 348 genome-wide datasets (127 whole-genome sequence datasets (WGSs) and 221 whole-genome single-nucleotide polymorphism (SNP) chip datasets). We characterized a high-confidence list of genetic signatures related response to high-altitude adaptation in Tibetan pigs, including 4,598 candidate SNPs and 131 candidate genes. Functional annotation and enrichment analysis revealed that 131 candidate genes are related to multiple systems and organs in Tibetan pigs. Notably, eight of the top ten novel genes, RALB, NBEA, LIFR, CLEC17A, PRIM2, CDH7, GK5 and FAM83B, were highlighted and associated with improved adaptive heart functions in Tibetan pigs high-altitude adaptation. Moreover, genome-wide association analysis revealed that 29 SNPs were involved in 13 candidate genes associated with at least one adaptive trait. In particular, among the top ten candidate genes, CLEC17A is related to a reduction in hemoglobin (HGB) in Tibetan pigs. Overall, our study provides a robust SNP/gene list involving genetic adaptation for Tibetan pig high-altitude adaptation, and it will be a valuable resource for future Tibetan pig studies.
Collapse
Affiliation(s)
- Lingyun Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yanbin Zhu
- Academy of Agriculture and Animal Husbandry Sciences, Institute of Animal Husbandry and Veterinary Medicine, Lhasa, China
| | - Yue Ren
- Academy of Agriculture and Animal Husbandry Sciences, Institute of Animal Husbandry and Veterinary Medicine, Lhasa, China
| | - Linna Xu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xuanbo Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China
| | - Ting Jiao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- College of Grassland Science, Gansu Agricultural University, Lanzhou, China
| | - Guangming Sun
- Academy of Agriculture and Animal Husbandry Sciences, Institute of Animal Husbandry and Veterinary Medicine, Lhasa, China
| | - Haiyu Han
- The Animal Husbandry Station in Changdu, Changdu, China
| | - Jian Zhang
- The Beast Prevention Station in Gongbujiangda County, Linzhi, China
| | - Fengbo Sun
- The Animal Husbandry Station in Tibet Autonomous Region, Lhasa, China
| | - Yanan Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China.
| |
Collapse
|
3
|
Kaizuka T, Takumi T. Alteration of synaptic protein composition during developmental synapse maturation. Eur J Neurosci 2024; 59:2894-2914. [PMID: 38571321 DOI: 10.1111/ejn.16304] [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: 05/01/2023] [Revised: 01/02/2024] [Accepted: 02/07/2024] [Indexed: 04/05/2024]
Abstract
The postsynaptic density (PSD) is a collection of specialized proteins assembled beneath the postsynaptic membrane of dendritic spines. The PSD proteome comprises ~1000 proteins, including neurotransmitter receptors, scaffolding proteins and signalling enzymes. Many of these proteins have essential roles in synaptic function and plasticity. During brain development, changes are observed in synapse density and in the stability and shape of spines, reflecting the underlying molecular maturation of synapses. Synaptic protein composition changes in terms of protein abundance and the assembly of protein complexes, supercomplexes and the physical organization of the PSD. Here, we summarize the developmental alterations of postsynaptic protein composition during synapse maturation. We describe major PSD proteins involved in postsynaptic signalling that regulates synaptic plasticity and discuss the effect of altered expression of these proteins during development. We consider the abnormality of synaptic profiles and synaptic protein composition in the brain in neurodevelopmental disorders such as autism spectrum disorders. We also explain differences in synapse development between rodents and primates in terms of synaptic profiles and protein composition. Finally, we introduce recent findings related to synaptic diversity and nanoarchitecture and discuss their impact on future research. Synaptic protein composition can be considered a major determinant and marker of synapse maturation in normality and disease.
Collapse
Affiliation(s)
- Takeshi Kaizuka
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
4
|
Wolterhoff N, Hiesinger PR. Synaptic promiscuity in brain development. Curr Biol 2024; 34:R102-R116. [PMID: 38320473 PMCID: PMC10849093 DOI: 10.1016/j.cub.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Precise synaptic connectivity is a prerequisite for the function of neural circuits, yet individual neurons, taken out of their developmental context, readily form unspecific synapses. How does the genome encode brain wiring in light of this apparent contradiction? Synaptic specificity is the outcome of a long series of developmental processes and mechanisms before, during and after synapse formation. How much promiscuity is permissible or necessary at the moment of synaptic partner choice depends on the extent to which prior development restricts available partners or subsequent development corrects initially made synapses. Synaptic promiscuity at the moment of choice can thereby play important roles in the development of precise connectivity, but also facilitate developmental flexibility and robustness. In this review, we assess the experimental evidence for the prevalence and roles of promiscuous synapse formation during brain development. Many well-established experimental approaches are based on developmental genetic perturbation and an assessment of synaptic connectivity only in the adult; this can make it difficult to pinpoint when a given defect or mechanism occurred. In many cases, such studies reveal mechanisms that restrict partner availability already prior to synapse formation. Subsequently, at the moment of choice, factors including synaptic competency, interaction dynamics and molecular recognition further restrict synaptic partners. The discussion of the development of synaptic specificity through the lens of synaptic promiscuity suggests an algorithmic process based on neurons capable of promiscuous synapse formation that are continuously prevented from making the wrong choices, with no single mechanism or developmental time point sufficient to explain the outcome.
Collapse
Affiliation(s)
- Neele Wolterhoff
- Division of Neurobiology, Free University Berlin, 14195 Berlin, Germany
| | - P Robin Hiesinger
- Division of Neurobiology, Free University Berlin, 14195 Berlin, Germany.
| |
Collapse
|
5
|
Mesías RE, Zaki Y, Guevara CA, Friedman LG, Hussein A, Therrien K, Magee AR, Tzavaras N, Del Valle P, Baxter MG, Huntley GW, Benson DL. Development and cadherin-mediated control of prefrontal corticostriatal projections in mice. iScience 2023; 26:108002. [PMID: 37854688 PMCID: PMC10579443 DOI: 10.1016/j.isci.2023.108002] [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: 03/21/2023] [Revised: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Action-outcome associations depend on prefrontal cortex (PFC) projections to the dorsal striatum. To assess how these projections form, we measured PFC axon patterning, synapse formation, and functional maturation in the postnatally developing mouse striatum. Using Hotspot analysis, we show that PFC axons form an adult-like pattern of clustered terminations in the first postnatal week that remains largely stable thereafter. PFC-striatal synaptic strength is adult-like by P21, while excitatory synapse density increases until adulthood. We then tested how the targeted deletion of a candidate adhesion/guidance protein, Cadherin-8 (Cdh8), from corticostriatal neurons regulates pathway development. Mutant mice showed diminished PFC axon targeting and reduced spontaneous glutamatergic synaptic activity in the dorsal striatum. They also exhibited impaired behavioral performance in action-outcome learning. The data show that PFC-striatal axons form striatal territories through an early, directed growth model and they highlight essential contributions of Cdh8 to the anatomical and functional features critical for the formation of action-outcome associations.
Collapse
Affiliation(s)
- Roxana E. Mesías
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yosif Zaki
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher A. Guevara
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lauren G. Friedman
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ayan Hussein
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karen Therrien
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexandra R. Magee
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nikolaos Tzavaras
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pamela Del Valle
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark G. Baxter
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - George W. Huntley
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deanna L. Benson
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
6
|
Kim M, Jun S, Park H, Tanaka-Yamamoto K, Yamamoto Y. Regulation of cerebellar network development by granule cells and their molecules. Front Mol Neurosci 2023; 16:1236015. [PMID: 37520428 PMCID: PMC10375027 DOI: 10.3389/fnmol.2023.1236015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
The well-organized cerebellar structures and neuronal networks are likely crucial for their functions in motor coordination, motor learning, cognition, and emotion. Such cerebellar structures and neuronal networks are formed during developmental periods through orchestrated mechanisms, which include not only cell-autonomous programs but also interactions between the same or different types of neurons. Cerebellar granule cells (GCs) are the most numerous neurons in the brain and are generated through intensive cell division of GC precursors (GCPs) during postnatal developmental periods. While GCs go through their own developmental processes of proliferation, differentiation, migration, and maturation, they also play a crucial role in cerebellar development. One of the best-characterized contributions is the enlargement and foliation of the cerebellum through massive proliferation of GCPs. In addition to this contribution, studies have shown that immature GCs and GCPs regulate multiple factors in the developing cerebellum, such as the development of other types of cerebellar neurons or the establishment of afferent innervations. These studies have often found impairments of cerebellar development in animals lacking expression of certain molecules in GCs, suggesting that the regulations are mediated by molecules that are secreted from or present in GCs. Given the growing recognition of GCs as regulators of cerebellar development, this review will summarize our current understanding of cerebellar development regulated by GCs and molecules in GCs, based on accumulated studies and recent findings, and will discuss their potential further contributions.
Collapse
Affiliation(s)
- Muwoong Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| |
Collapse
|
7
|
Guo Q, Wang Y, Wang Q, Qian Y, Jiang Y, Dong X, Chen H, Chen X, Liu X, Yu S, Zhu J, Shan S, Wu B, Zhou W, Wang H. In the developing cerebral cortex: axonogenesis, synapse formation, and synaptic plasticity are regulated by SATB2 target genes. Pediatr Res 2023; 93:1519-1527. [PMID: 36028553 DOI: 10.1038/s41390-022-02260-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Special AT-rich sequence-binding protein 2 is essential for the development of cerebral cortex and key molecular node for the establishment of proper neural circuitry and function. Mutations in the SATB2 gene lead to SATB2-associated syndrome, which is characterized by abnormal development of skeleton and central nervous systems. METHODS We generated Satb2 knockout mouse model through CRISPR-Cas9 technology and performed RNA-seq and ChIP-seq of embryonic cerebral cortex. We conducted RT-qPCR, western blot, immunofluorescence staining, luciferase reporter assay and behavioral analysis for experimental verification. RESULTS We identified 1363 downstream effector genes of Satb2 and correlation analysis of Satb2-targeted genes and neurological disease genes showed that Satb2 contribute to cognitive and mental disorders from the early developmental stage. We found that Satb2 directly regulate the expression of Ntng1, Cdh13, Kitl, genes important for axon guidance, synaptic formation, neuron migration, and Satb2 directly activates the expression of Mef2c. We also showed that Satb2 heterozygous knockout mice showed impaired spatial learning and memory. CONCLUSIONS Taken together, our study supportsroles of Satb2 in the regulation of axonogenesis and synaptic formation at the early developmental stage and provides new insights into the complicated regulatory mechanism of Satb2 and new evidence to elucidate the pathogen of SATB2-associated syndrome. IMPACT 1363 downstream effector genes of Satb2 were classified into 5 clusters with different temporal expression patterns. We identified Plxnd1, Ntng1, Efnb2, Ephb1, Plxna2, Epha3, Plxna4, Unc5c, and Flrt2 as axon guidance molecules to regulate axonogenesis. 168 targeted genes of Satb2 were found to regulate synaptic formation in the early development of the cerebral cortex. Transcription factor Mef2c is positively regulated by Satb2, and 28 Mef2c-targeted genes can be directly regulated by Satb2. In the Morris water maze test, Satb2+/- mice showed impaired spatial learning and memory, further strengthening that Satb2 can regulate synaptic functions.
Collapse
Affiliation(s)
- Qiufang Guo
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
- Berry Genomics Co, 102206, Beijing, China
| | - Yaqiong Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Qing Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Yanyan Qian
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Yinmo Jiang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Xinran Dong
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Huiyao Chen
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Xiang Chen
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Xiuyun Liu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Sha Yu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Jitao Zhu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Shifang Shan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 200032, Shanghai, China
| | - Bingbing Wu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Wenhao Zhou
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China.
- Division of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Key Laboratory of Neonatal Diseases, Ministry of Health, 201102, Shanghai, China.
| | - Huijun Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China.
| |
Collapse
|
8
|
Kim T, Park H, Tanaka-Yamamoto K, Yamamoto Y. Developmental timing-dependent organization of synaptic connections between mossy fibers and granule cells in the cerebellum. Commun Biol 2023; 6:446. [PMID: 37095324 PMCID: PMC10125988 DOI: 10.1038/s42003-023-04825-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/07/2023] [Indexed: 04/26/2023] Open
Abstract
The long-standing hypothesis that synapses between mossy fibers (MFs) and cerebellar granule cells (GCs) are organized according to the origins of MFs and locations of GC axons, parallel fibers (PFs), is supported by recent findings. However, the mechanisms of such organized synaptic connections remain unknown. Here, using our technique that enabled PF location-dependent labeling of GCs in mice, we confirmed that synaptic connections of GCs with specific MFs originating from the pontine nucleus (PN-MFs) and dorsal column nuclei (DCoN-MFs) were gently but differentially organized according to their PF locations. We then found that overall MF-GC synaptic connectivity was biased in a way that dendrites of GCs having nearby PFs tended to connect with the same MF terminals, implying that the MF origin- and PF location-dependent organization is associated with the overall biased MF-GC synaptic connectivity. Furthermore, the development of PN-MFs preceded that of DCoN-MFs, which matches the developmental sequence of GCs that preferentially connect with each type of these MFs. Thus, our results revealed that overall MF-GC synaptic connectivity is biased in terms of PF locations, and suggested that such connectivity is likely the result of synaptic formation between developmental timing-matched partners.
Collapse
Affiliation(s)
- Taegon Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
9
|
Mesías RE, Zaki Y, Guevara CA, Friedman LG, Hussein A, Therrien K, Magee AR, Tzavaras N, Valle PD, Baxter MG, Huntley GW, Benson DL. Development of prefrontal corticostriatal connectivity in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532475. [PMID: 36993639 PMCID: PMC10054964 DOI: 10.1101/2023.03.14.532475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Rational decision making is grounded in learning to associate actions with outcomes, a process that depends on projections from prefrontal cortex to dorsomedial striatum. Symptoms associated with a variety of human pathological conditions ranging from schizophrenia and autism to Huntington's and Parkinson's disease point toward functional deficits in this projection, but its development is not well understood, making it difficult to investigate how perturbations in development of this circuitry could contribute to pathophysiology. We applied a novel strategy based on Hotspot Analysis to assess the developmental progression of anatomical positioning of prefrontal cortex to striatal projections. Corticostriatal axonal territories established at P7 expand in concert with striatal growth but remain largely unchanged in positioning through adulthood, indicating they are generated by directed, targeted growth and not modified extensively by postnatal experience. Consistent with these findings, corticostriatal synaptogenesis increased steadily from P7 to P56, with no evidence for widescale pruning. As corticostriatal synapse density increased over late postnatal ages, the strength of evoked PFC input onto dorsomedial striatal projection neurons also increased, but spontaneous glutamatergic synaptic activity was stable. Based on its pattern of expression, we asked whether the adhesion protein, Cdh8, influenced this progression. In mice lacking Cdh8 in PFC corticostriatal projection neurons, axon terminal fields in dorsal striatum shifted ventrally. Corticostriatal synaptogenesis was unimpeded, but spontaneous EPSC frequency declined and mice failed to learn to associate an action with an outcome. Collectively these findings show that corticostriatal axons grow to their target zone and are restrained from an early age, do not undergo postnatal synapse pruning as the most dominant models predict, and that a relatively modest shift in terminal arbor positioning and synapse function has an outsized, negative impact on corticostriatal-dependent behavior.
Collapse
|
10
|
Hasegawa K, Matsui TK, Kondo J, Kuwako KI. N-WASP-Arp2/3 signaling controls multiple steps of dendrite maturation in Purkinje cells in vivo. Development 2022; 149:285127. [PMID: 36469048 DOI: 10.1242/dev.201214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
During neural development, the actin filament network must be precisely regulated to form elaborate neurite structures. N-WASP tightly controls actin polymerization dynamics by activating an actin nucleator Arp2/3. However, the importance of N-WASP-Arp2/3 signaling in the assembly of neurite architecture in vivo has not been clarified. Here, we demonstrate that N-WASP-Arp2/3 signaling plays a crucial role in the maturation of cerebellar Purkinje cell (PC) dendrites in vivo in mice. N-WASP was expressed and activated in developing PCs. Inhibition of Arp2/3 and N-WASP from the beginning of dendrite formation severely disrupted the establishment of a single stem dendrite, which is a characteristic basic structure of PC dendrites. Inhibition of Arp2/3 after stem dendrite formation resulted in hypoplasia of the PC dendritic tree. Cdc42, an upstream activator of N-WASP, is required for N-WASP-Arp2/3 signaling-mediated PC dendrite maturation. In addition, overactivation of N-WASP is also detrimental to dendrite formation in PCs. These findings reveal that proper activation of N-WASP-Arp2/3 signaling is crucial for multiple steps of PC dendrite maturation in vivo.
Collapse
Affiliation(s)
- Koichi Hasegawa
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| | - Takeshi K Matsui
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| | - Junpei Kondo
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| | - Ken-Ichiro Kuwako
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| |
Collapse
|
11
|
Carlsen EØ, Lee Y, Magnus P, Jugessur A, Page CM, Nustad HE, Håberg SE, Lie RT, Magnus MC. An examination of mediation by DNA methylation on birthweight differences induced by assisted reproductive technologies. Clin Epigenetics 2022; 14:151. [PMID: 36443807 PMCID: PMC9703677 DOI: 10.1186/s13148-022-01381-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Children born after assisted reproductive technologies (ART) differ in birthweight from those naturally conceived. It has been hypothesized that this might be explained by epigenetic mechanisms. We examined whether cord blood DNA methylation mediated the birthweight difference between 890 newborns conceived by ART (764 by fresh embryo transfer and 126 frozen thawed embryo transfer) and 983 naturally conceived newborns from the Norwegian Mother, Father, and Child Cohort Study (MoBa). DNA methylation was measured by the Illumina Infinium MethylationEPIC array. We conducted mediation analyses to assess whether differentially methylated CpGs mediated the differences in birthweight observed between: (1) fresh embryo transfer and natural conception and (2) frozen and fresh embryo transfer. RESULTS We observed a difference in birthweight between fresh embryo transfer and naturally conceived offspring of - 120 g. 44% (95% confidence interval [CI] 26% to 81%) of this difference in birthweight between fresh embryo transfer and naturally conceived offspring was explained by differences in methylation levels at four CpGs near LOXL1, CDH20, and DRC1. DNA methylation differences at two CpGs near PTGS1 and RASGRP4 jointly mediated 22% (95% CI 8.1% to 50.3%) of the birthweight differences between fresh and frozen embryo transfer. CONCLUSION Our findings suggest that DNA methylation is an important mechanism in explaining birthweight differences according to the mode of conception. Further research should examine how gene regulation at these loci influences fetal growth.
Collapse
Affiliation(s)
- Ellen Ø. Carlsen
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Community Medicine, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Yunsung Lee
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Per Magnus
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Astanand Jugessur
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway ,grid.7914.b0000 0004 1936 7443Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Christian M. Page
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway ,grid.5510.10000 0004 1936 8921Department of Mathematics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Haakon E. Nustad
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway ,Deepinsight, Oslo, Norway
| | - Siri E. Håberg
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Rolv T. Lie
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway ,grid.7914.b0000 0004 1936 7443Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Maria C. Magnus
- grid.418193.60000 0001 1541 4204Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
12
|
Moreland T, Poulain FE. To Stick or Not to Stick: The Multiple Roles of Cell Adhesion Molecules in Neural Circuit Assembly. Front Neurosci 2022; 16:889155. [PMID: 35573298 PMCID: PMC9096351 DOI: 10.3389/fnins.2022.889155] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 01/02/2023] Open
Abstract
Precise wiring of neural circuits is essential for brain connectivity and function. During development, axons respond to diverse cues present in the extracellular matrix or at the surface of other cells to navigate to specific targets, where they establish precise connections with post-synaptic partners. Cell adhesion molecules (CAMs) represent a large group of structurally diverse proteins well known to mediate adhesion for neural circuit assembly. Through their adhesive properties, CAMs act as major regulators of axon navigation, fasciculation, and synapse formation. While the adhesive functions of CAMs have been known for decades, more recent studies have unraveled essential, non-adhesive functions as well. CAMs notably act as guidance cues and modulate guidance signaling pathways for axon pathfinding, initiate contact-mediated repulsion for spatial organization of axonal arbors, and refine neuronal projections during circuit maturation. In this review, we summarize the classical adhesive functions of CAMs in axonal development and further discuss the increasing number of other non-adhesive functions CAMs play in neural circuit assembly.
Collapse
|
13
|
Hirsch D, Kohl A, Wang Y, Sela-Donenfeld D. Axonal Projection Patterns of the Dorsal Interneuron Populations in the Embryonic Hindbrain. Front Neuroanat 2022; 15:793161. [PMID: 35002640 PMCID: PMC8738170 DOI: 10.3389/fnana.2021.793161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Unraveling the inner workings of neural circuits entails understanding the cellular origin and axonal pathfinding of various neuronal groups during development. In the embryonic hindbrain, different subtypes of dorsal interneurons (dINs) evolve along the dorsal-ventral (DV) axis of rhombomeres and are imperative for the assembly of central brainstem circuits. dINs are divided into two classes, class A and class B, each containing four neuronal subgroups (dA1-4 and dB1-4) that are born in well-defined DV positions. While all interneurons belonging to class A express the transcription factor Olig3 and become excitatory, all class B interneurons express the transcription factor Lbx1 but are diverse in their excitatory or inhibitory fate. Moreover, within every class, each interneuron subtype displays its own specification genes and axonal projection patterns which are required to govern the stage-by-stage assembly of their connectivity toward their target sites. Remarkably, despite the similar genetic landmark of each dINs subgroup along the anterior-posterior (AP) axis of the hindbrain, genetic fate maps of some dA/dB neuronal subtypes uncovered their contribution to different nuclei centers in relation to their rhombomeric origin. Thus, DV and AP positional information has to be orchestrated in each dA/dB subpopulation to form distinct neuronal circuits in the hindbrain. Over the span of several decades, different axonal routes have been well-documented to dynamically emerge and grow throughout the hindbrain DV and AP positions. Yet, the genetic link between these distinct axonal bundles and their neuronal origin is not fully clear. In this study, we reviewed the available data regarding the association between the specification of early-born dorsal interneuron subpopulations in the hindbrain and their axonal circuitry development and fate, as well as the present existing knowledge on molecular effectors underlying the process of axonal growth.
Collapse
Affiliation(s)
- Dana Hirsch
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.,Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
14
|
Takahashi M, Fukabori R, Kawasaki H, Kobayashi K, Kawakami K. The distribution of Cdh20 mRNA demarcates somatotopic subregions and subpopulations of spiny projection neurons in the rat dorsolateral striatum. J Comp Neurol 2021; 529:3655-3675. [PMID: 34240415 DOI: 10.1002/cne.25215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/21/2021] [Accepted: 07/02/2021] [Indexed: 11/07/2022]
Abstract
The dorsolateral striatum (DLS) of rodents is functionally subdivided into somatotopic subregions that represent each body part along both the dorsoventral and anteroposterior (A-P) axes and play crucial roles in sensorimotor functions via corticostriatal pathways. However, little is known about the spatial gene expression patterns and heterogeneity of spiny projection neurons (SPNs) within somatotopic subregions. Here, we show that the cell adhesion molecule gene Cdh20, which encodes a Type II cadherin, is expressed in discrete subregions covering the inner orofacial area and part of the forelimb area in the ventral domain of the DLS (v-DLS) in rats. Cdh20-expressing cells were localized in the v-DLS at the intermediate level of the striatum along the A-P axis and could be classified as direct-pathway SPNs or indirect-pathway SPNs. Unexpectedly, comprehensive analysis revealed that Cdh20 is expressed in SPNs in the rat DLS but not in the mouse DLS or the ferret putamen (Pu). Our observations reveal that Cdh20 expression demarcates somatotopic subregions and subpopulations of SPNs specifically in the rat DLS and suggest divergent regulation of genes differentially expressed in the v-DLS and Pu among mammals.
Collapse
Affiliation(s)
- Masanori Takahashi
- Graduate School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.,Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Ryoji Fukabori
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | | |
Collapse
|
15
|
Hayano Y, Ishino Y, Hyun JH, Orozco CG, Steinecke A, Potts E, Oisi Y, Thomas CI, Guerrero-Given D, Kim E, Kwon HB, Kamasawa N, Taniguchi H. IgSF11 homophilic adhesion proteins promote layer-specific synaptic assembly of the cortical interneuron subtype. SCIENCE ADVANCES 2021; 7:eabf1600. [PMID: 34261648 PMCID: PMC8279514 DOI: 10.1126/sciadv.abf1600] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 05/28/2021] [Indexed: 05/16/2023]
Abstract
The most prominent structural hallmark of the mammalian neocortical circuitry is the layer-based organization of specific cell types and synaptic inputs. Accordingly, cortical inhibitory interneurons (INs), which shape local network activity, exhibit subtype-specific laminar specificity of synaptic outputs. However, the underlying molecular mechanisms remain unknown. Here, we demonstrate that Immunoglobulin Superfamily member 11 (IgSF11) homophilic adhesion proteins are preferentially expressed in one of the most distinctive IN subtypes, namely, chandelier cells (ChCs) that specifically innervate axon initial segments of pyramidal neurons (PNs), and their synaptic laminar target. Loss-of-function experiments in either ChCs or postsynaptic cells revealed that IgSF11 is required for ChC synaptic development in the target layer. While overexpression of IgSF11 in ChCs enlarges ChC presynaptic boutons, expressing IgSF11 in nontarget layers induces ectopic ChC synapses. These findings provide evidence that synapse-promoting adhesion proteins, highly localized to synaptic partners, determine the layer-specific synaptic connectivity of the cortical IN subtype.
Collapse
Affiliation(s)
- Yasufumi Hayano
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Yugo Ishino
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Jung Ho Hyun
- Cellular Basis of Neural Circuit Plasticity, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Carlos G Orozco
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - André Steinecke
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Elizabeth Potts
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Yasuhiro Oisi
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Connon I Thomas
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Debbie Guerrero-Given
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Hyung-Bae Kwon
- Cellular Basis of Neural Circuit Plasticity, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Max Planck Institute for Neurobiology, Martinsried, Munich 82152, Germany
| | - Naomi Kamasawa
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Hiroki Taniguchi
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA.
| |
Collapse
|
16
|
Polanco J, Reyes-Vigil F, Weisberg SD, Dhimitruka I, Brusés JL. Differential Spatiotemporal Expression of Type I and Type II Cadherins Associated With the Segmentation of the Central Nervous System and Formation of Brain Nuclei in the Developing Mouse. Front Mol Neurosci 2021; 14:633719. [PMID: 33833667 PMCID: PMC8021962 DOI: 10.3389/fnmol.2021.633719] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/10/2021] [Indexed: 11/20/2022] Open
Abstract
Type I and type II classical cadherins comprise a family of cell adhesion molecules that regulate cell sorting and tissue separation by forming specific homo and heterophilic bonds. Factors that affect cadherin-mediated cell-cell adhesion include cadherin binding affinity and expression level. This study examines the expression pattern of type I cadherins (Cdh1, Cdh2, Cdh3, and Cdh4), type II cadherins (Cdh6, Cdh7, Cdh8, Cdh9, Cdh10, Cdh11, Cdh12, Cdh18, Cdh20, and Cdh24), and the atypical cadherin 13 (Cdh13) during distinct morphogenetic events in the developing mouse central nervous system from embryonic day 11.5 to postnatal day 56. Cadherin mRNA expression levels obtained from in situ hybridization experiments carried out at the Allen Institute for Brain Science (https://alleninstitute.org/) were retrieved from the Allen Developing Mouse Brain Atlas. Cdh2 is the most abundantly expressed type I cadherin throughout development, while Cdh1, Cdh3, and Cdh4 are expressed at low levels. Type II cadherins show a dynamic pattern of expression that varies between neuroanatomical structures and developmental ages. Atypical Cdh13 expression pattern correlates with Cdh2 in abundancy and localization. Analyses of cadherin-mediated relative adhesion estimated from their expression level and binding affinity show substantial differences in adhesive properties between regions of the neural tube associated with the segmentation along the anterior–posterior axis. Differences in relative adhesion were also observed between brain nuclei in the developing subpallium (basal ganglia), suggesting that differential cell adhesion contributes to the segregation of neuronal pools. In the adult cerebral cortex, type II cadherins Cdh6, Cdh8, Cdh10, and Cdh12 are abundant in intermediate layers, while Cdh11 shows a gradated expression from the deeper layer 6 to the superficial layer 1, and Cdh9, Cdh18, and Cdh24 are more abundant in the deeper layers. Person’s correlation analyses of cadherins mRNA expression patterns between areas and layers of the cerebral cortex and the nuclei of the subpallium show significant correlations between certain cortical areas and the basal ganglia. The study shows that differential cadherin expression and cadherin-mediated adhesion are associated with a wide range of morphogenetic events in the developing central nervous system including the organization of neurons into layers, the segregation of neurons into nuclei, and the formation of neuronal circuits.
Collapse
Affiliation(s)
- Julie Polanco
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY, United States
| | - Fredy Reyes-Vigil
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY, United States
| | - Sarah D Weisberg
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY, United States
| | - Ilirian Dhimitruka
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY, United States
| | - Juan L Brusés
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY, United States
| |
Collapse
|
17
|
Chowdhury D, Watters K, Biederer T. Synaptic recognition molecules in development and disease. Curr Top Dev Biol 2021; 142:319-370. [PMID: 33706921 DOI: 10.1016/bs.ctdb.2020.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Synaptic connectivity patterns underlie brain functions. How recognition molecules control where and when neurons form synapses with each other, therefore, is a fundamental question of cellular neuroscience. This chapter delineates adhesion and signaling complexes as well as secreted factors that contribute to synaptic partner recognition in the vertebrate brain. The sections follow a developmental perspective and discuss how recognition molecules (1) guide initial synaptic wiring, (2) provide for the rejection of incorrect partner choices, (3) contribute to synapse specification, and (4) support the removal of inappropriate synapses once formed. These processes involve a rich repertoire of molecular players and key protein families are described, notably the Cadherin and immunoglobulin superfamilies, Semaphorins/Plexins, Leucine-rich repeat containing proteins, and Neurexins and their binding partners. Molecular themes that diversify these recognition systems are defined and highlighted throughout the text, including the neuron-type specific expression and combinatorial action of recognition factors, alternative splicing, and post-translational modifications. Methodological innovations advancing the field such as proteomic approaches and single cell expression studies are additionally described. Further, the chapter highlights the importance of choosing an appropriate brain region to analyze synaptic recognition factors and the advantages offered by laminated structures like the hippocampus or retina. In a concluding section, the profound disease relevance of aberrant synaptic recognition for neurodevelopmental and psychiatric disorders is discussed. Based on the current progress, an outlook is presented on research goals that can further advance insights into how recognition molecules provide for the astounding precision and diversity of synaptic connections.
Collapse
Affiliation(s)
| | - Katherine Watters
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States; Neuroscience Graduate Program, Tufts University School of Medicine, Boston, MA, United States
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
18
|
Heckman EL, Doe CQ. Establishment and Maintenance of Neural Circuit Architecture. J Neurosci 2021; 41:1119-1129. [PMID: 33568445 PMCID: PMC7888231 DOI: 10.1523/jneurosci.1143-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/29/2020] [Accepted: 12/09/2020] [Indexed: 02/03/2023] Open
Abstract
The ability to sense the world, process information, and navigate the environment depends on the assembly and continuous function of neural circuits in the brain. Within the past two decades, new technologies have rapidly advanced our understanding of how neural circuits are wired during development and how they are stably maintained, often for years. Electron microscopy reconstructions of model organism connectomes have provided a map of the stereotyped (and variable) connections in the brain; advanced light microscopy techniques have enabled direct observation of the cellular dynamics that underlie circuit construction and maintenance; transcriptomic and proteomic surveys of both developing and mature neurons have provided insights into the molecular and genetic programs governing circuit establishment and maintenance; and advanced genetic techniques have allowed for high-throughput discovery of wiring regulators. These tools have empowered scientists to rapidly generate and test hypotheses about how circuits establish and maintain connectivity. Thus, the set of principles governing circuit formation and maintenance have been expanded. These principles are discussed in this review.
Collapse
Affiliation(s)
- Emily L Heckman
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, Oregon 97403
| | - Chris Q Doe
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, Oregon 97403
| |
Collapse
|
19
|
Puñal VM, Ahmed M, Thornton-Kolbe EM, Clowney EJ. Untangling the wires: development of sparse, distributed connectivity in the mushroom body calyx. Cell Tissue Res 2021; 383:91-112. [PMID: 33404837 PMCID: PMC9835099 DOI: 10.1007/s00441-020-03386-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/07/2020] [Indexed: 01/16/2023]
Abstract
Appropriate perception and representation of sensory stimuli pose an everyday challenge to the brain. In order to represent the wide and unpredictable array of environmental stimuli, principle neurons of associative learning regions receive sparse, combinatorial sensory inputs. Despite the broad role of such networks in sensory neural circuits, the developmental mechanisms underlying their emergence are not well understood. As mammalian sensory coding regions are numerically complex and lack the accessibility of simpler invertebrate systems, we chose to focus this review on the numerically simpler, yet functionally similar, Drosophila mushroom body calyx. We bring together current knowledge about the cellular and molecular mechanisms orchestrating calyx development, in addition to drawing insights from literature regarding construction of sparse wiring in the mammalian cerebellum. From this, we formulate hypotheses to guide our future understanding of the development of this critical perceptual center.
Collapse
Affiliation(s)
- Vanessa M. Puñal
- Department of Molecular, Cellular & Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA,Department of Molecular & Integrative Physiology, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria Ahmed
- Department of Molecular, Cellular & Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Emma M. Thornton-Kolbe
- Department of Molecular, Cellular & Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA,Neuroscience Graduate Program, The University of Michigan, Ann Arbor, MI 48109, USA
| | - E. Josephine Clowney
- Department of Molecular, Cellular & Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
20
|
Ikuta R, Myoenzono K, Wasano J, Hamaguchi-Hamada K, Hamada S, Kurumata-Shigeto M. N-cadherin localization in taste buds of mouse circumvallate papillae. J Comp Neurol 2020; 529:2227-2242. [PMID: 33319419 DOI: 10.1002/cne.25090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 01/03/2023]
Abstract
Taste buds, the receptor organs for taste, contain 50-100 taste bud cells. Although these cells undergo continuous turnover, the structural and functional integrity of taste buds is maintained. The molecular mechanisms by which synaptic connectivity between taste buds and afferent fibers is formed and maintained remain ambiguous. In the present study, we examined the localization of N-cadherin in the taste buds of the mouse circumvallate papillae because N-cadherin, one of the classical cadherins, is important for the formation and maintenance of synapses. At the light microscopic level, N-cadherin was predominantly detected in type II cells and nerve fibers in the connective tissues in and around the vallate papillae. At the ultrastructural level, N-cadherin immunoreactivity appears along the cell membrane and in the intracellular vesicles of type II cells. N-cadherin immunoreactivity also is evident in the membranes of afferent terminals at the contact sites to N-cadherin-positive type II cells. At channel type synapses between type II cells and nerve fibers, N-cadherin is present surrounding, but not within, the presumed neurotransmitter release zone, identified by large mitochondria apposed to the taste cells. The present results suggest that N-cadherin is important for the formation or maintenance of type II cell afferent synapses in taste buds.
Collapse
Affiliation(s)
- Rio Ikuta
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | - Kanae Myoenzono
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan.,Humanome Lab., Inc., Tokyo, Japan
| | - Jun Wasano
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | | | - Shun Hamada
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | - Mami Kurumata-Shigeto
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| |
Collapse
|
21
|
Refinement of Cerebellar Network Organization by Extracellular Signaling During Development. Neuroscience 2020; 462:44-55. [PMID: 32502568 DOI: 10.1016/j.neuroscience.2020.05.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022]
Abstract
The cerebellum forms regular neural network structures consisting of a few major types of neurons, such as Purkinje cells, granule cells, and molecular layer interneurons, and receives two major inputs from climbing fibers and mossy fibers. Its regular structures consist of three well-defined layers, with each type of neuron designated to a specific location and forming specific synaptic connections. During the first few weeks of postnatal development in rodents, the cerebellum goes through dynamic changes via proliferation, migration, differentiation, synaptogenesis, and maturation, to create such a network structure. The development of this organized network structure presumably relies on the communication between developing elements in the network, including not only individual neurons, but also their dendrites, axons, and synapses. Therefore, it is reasonable that extracellular signaling via synaptic transmission, secreted molecules, and cell adhesion molecules, plays important roles in cerebellar network development. Although it is not yet clear as to how overall cerebellar development is orchestrated, there is indeed accumulating lines of evidence that extracellular signaling acts toward the development of individual elements in the cerebellar networks. In this article, we introduce what we have learned from many studies regarding the extracellular signaling required for cerebellar network development, including our recent study suggesting the importance of unbiased synaptic inputs from parallel fibers.
Collapse
|
22
|
Sanes JR, Zipursky SL. Synaptic Specificity, Recognition Molecules, and Assembly of Neural Circuits. Cell 2020; 181:536-556. [DOI: 10.1016/j.cell.2020.04.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 01/02/2023]
|
23
|
Kuwako KI, Okano H. The LKB1-SIK Pathway Controls Dendrite Self-Avoidance in Purkinje Cells. Cell Rep 2019; 24:2808-2818.e4. [PMID: 30208308 DOI: 10.1016/j.celrep.2018.08.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/12/2018] [Accepted: 08/08/2018] [Indexed: 02/08/2023] Open
Abstract
Strictly controlled dendrite patterning underlies precise neural connection. Dendrite self-avoidance is a crucial system preventing self-crossing and clumping of dendrites. Although many cell-surface molecules that regulate self-avoidance have been identified, the signaling pathway that orchestrates it remains poorly understood, particularly in mammals. Here, we demonstrate that the LKB1-SIK kinase pathway plays a pivotal role in the self-avoidance of Purkinje cell (PC) dendrites by ensuring dendritic localization of Robo2, a regulator of self-avoidance. LKB1 is activated in developing PCs, and PC-specific deletion of LKB1 severely disrupts the self-avoidance of PC dendrites without affecting gross morphology. SIK1 and SIK2, downstream kinases of LKB1, mediate LKB1-dependent dendrite self-avoidance. Furthermore, loss of LKB1 leads to significantly decreased Robo2 levels in the dendrite but not in the cell body. Finally, restoration of dendritic Robo2 level via overexpression largely rescues the self-avoidance defect in LKB1-deficient PCs. These findings reveal an LKB1-pathway-mediated developmental program that establishes dendrite self-avoidance.
Collapse
Affiliation(s)
- Ken-Ichiro Kuwako
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
24
|
Kim B. Evolutionarily conserved and divergent functions for cell adhesion molecules in neural circuit assembly. J Comp Neurol 2019; 527:2061-2068. [PMID: 30779135 DOI: 10.1002/cne.24666] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Abstract
The developing nervous system generates remarkably precise synaptic connections between neurons and their postsynaptic target cells. Numerous neural cell adhesion proteins have been identified to mediate cell recognition between synaptic partners in several model organisms. Here, I review the role of protein interactions of cell adhesion molecules in neural circuit assembly and address how these interactions are utilized to form different neural circuitries in different species. The emerging evidence suggests that the extracellular trans-interactions of cell adhesion proteins for neural wiring are evolutionarily conserved across taxa, but they are often used in different steps of circuit assembly. I also highlight how these conserved protein interactions work together as a group to specify neural connectivity.
Collapse
Affiliation(s)
- Byunghyuk Kim
- Department of Life Science, Dongguk University Seoul, Goyang, Republic of Korea
| |
Collapse
|
25
|
Memi F, Killen AC, Barber M, Parnavelas JG, Andrews WD. Cadherin 8 regulates proliferation of cortical interneuron progenitors. Brain Struct Funct 2018; 224:277-292. [PMID: 30315415 PMCID: PMC6373371 DOI: 10.1007/s00429-018-1772-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/05/2018] [Indexed: 01/18/2023]
Abstract
Cortical interneurons are born in the ventral forebrain and migrate tangentially in two streams at the levels of the intermediate zone (IZ) and the pre-plate/marginal zone to the developing cortex where they switch to radial migration before settling in their final positions in the cortical plate. In a previous attempt to identify the molecules that regulate stream specification, we performed transcriptomic analysis of GFP-labelled interneurons taken from the two migratory streams during corticogenesis. A number of cadherins were found to be expressed differentially, with Cadherin-8 (Cdh8) selectively present in the IZ stream. We verified this expression pattern at the mRNA and protein levels on tissue sections and found approximately half of the interneurons of the IZ expressed Cdh8. Furthermore, this cadherin was also detected in the germinal zones of the subpallium, suggesting that it might be involved not only in the migration of interneurons but also in their generation. Quantitative analysis of cortical interneurons in animals lacking the cadherin at E18.5 revealed a significant increase in their numbers. Subsequent functional in vitro experiments showed that blocking Cdh8 function led to increased cell proliferation, with the opposite results observed with over-expression, supporting its role in interneuron generation.
Collapse
Affiliation(s)
- Fani Memi
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Abigail C Killen
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Melissa Barber
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - William D Andrews
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
26
|
Duan X, Krishnaswamy A, Laboulaye MA, Liu J, Peng YR, Yamagata M, Toma K, Sanes JR. Cadherin Combinations Recruit Dendrites of Distinct Retinal Neurons to a Shared Interneuronal Scaffold. Neuron 2018; 99:1145-1154.e6. [PMID: 30197236 PMCID: PMC6284407 DOI: 10.1016/j.neuron.2018.08.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
Distinct neuronal types connect in complex ways to generate functional neural circuits. The molecular diversity required to specify this connectivity could be supplied by multigene families of synaptic recognition molecules, but most studies to date have assessed just one or a few members at a time. Here, we analyze roles of cadherins (Cdhs) in formation of retinal circuits comprising eight neuronal types that inform the brain about motion in four directions. We show that at least 15 classical Cdhs are expressed by neurons in these circuits and at least 6 (Cdh6-10 and 18) act individually or in combinations to promote specific connectivity among the cells. They act in part by directing the processes of output neurons and excitatory interneurons to a cellular scaffold formed by inhibitory interneurons. Because Cdhs are expressed combinatorially by many central neurons, similar interactions could be involved in patterning circuits throughout the brain.
Collapse
Affiliation(s)
- Xin Duan
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun Krishnaswamy
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Mallory A Laboulaye
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jinyue Liu
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yi-Rong Peng
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Masahito Yamagata
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Kenichi Toma
- Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
27
|
Abstract
The cadherin superfamily comprises a large, diverse collection of cell surface receptors that are expressed in the nervous system throughout development and have been shown to be essential for the proper assembly of the vertebrate nervous system. As our knowledge of each family member has grown, it has become increasingly clear that the functions of various cadherin subfamilies are intertwined: they can be present in the same protein complexes, impinge on the same developmental processes, and influence the same signaling pathways. This interconnectedness may illustrate a central way in which core developmental events are controlled to bring about the robust and precise assembly of neural circuitry.
Collapse
Affiliation(s)
- James D Jontes
- Department of Neuroscience, Ohio State University, Ohio 43210
| |
Collapse
|
28
|
Yamagata M, Duan X, Sanes JR. Cadherins Interact With Synaptic Organizers to Promote Synaptic Differentiation. Front Mol Neurosci 2018; 11:142. [PMID: 29760652 PMCID: PMC5936767 DOI: 10.3389/fnmol.2018.00142] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 01/17/2023] Open
Abstract
Classical cadherins, a set of ~20 related recognition and signaling molecules, have been implicated in many aspects of neural development, including the formation and remodeling of synapses. Mechanisms underlying some of these steps have been studied by expressing N-cadherin (cdh2), a Type 1 cadherin, in heterologous cells, but analysis is complicated because widely used lines express cdh2 endogenously. We used CRISPR-mediated gene editing to generate a Human embryonic kidney (HEK)293 variant lacking Cdh2, then compared the behavior of rodent cortical and hippocampal neurons co-cultured with parental, cdh2 mutant and cdh2-rescued 293 lines. The comparison demonstrated that Cdh2 promotes neurite branching and that it is required for three synaptic organizers, neurologin1 (NLGL1), leucine-rich repeat transmembrane protein 2 (LRRtm2), and Cell Adhesion Molecule 1 (Cadm1/SynCAM) to stimulate presynaptic differentiation, assayed by clustering of synaptic vesicles at sites of neurite-293 cell contact. Similarly, Cdh2 is required for a presynaptic organizing molecule, Neurexin1β, to promote postsynaptic differentiation in dendrites. We also show that another Type I cadherin, Cdh4, and a Type II cadherin, Cdh6, can substitute for Cdh2 in these assays. Finally, we provide evidence that the effects of cadherins require homophilic interactions between neurites and the heterologous cells. Together, these results indicate that classical cadherins act together with synaptic organizers to promote synaptic differentiation, perhaps in part by strengthening the intracellular adhesion required for the organizers to act efficiently. We propose that cadherins promote high affinity contacts between appropriate partners, which then enable synaptic differentiation.
Collapse
Affiliation(s)
- Masahito Yamagata
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
| | - Xin Duan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
| |
Collapse
|
29
|
Prolyl carboxypeptidase in Agouti-related Peptide neurons modulates food intake and body weight. Mol Metab 2018; 10:28-38. [PMID: 29459251 PMCID: PMC5985234 DOI: 10.1016/j.molmet.2018.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/26/2018] [Accepted: 02/04/2018] [Indexed: 12/15/2022] Open
Abstract
Objective Prolyl carboxypeptidase (PRCP) plays a role in the regulation of energy metabolism by inactivating hypothalamic α-melanocyte stimulating hormone (α-MSH) levels. Although detected in the arcuate nucleus, limited PRCP expression has been observed in the arcuate POMC neurons, and its site of action in regulating metabolism is still ill-defined. Methods We performed immunostaining to assess the localization of PRCP in arcuate Neuropeptide Y/Agouti-related Peptide (NPY/AgRP) neurons. Hypothalamic explants were then used to assess the intracellular localization of PRCP and its release at the synaptic levels. Finally, we generated a mouse model to assess the role of PRCP in NPY/AgRP neurons of the arcuate nucleus in the regulation of metabolism. Results Here we show that PRCP is expressed in NPY/AgRP-expressing neurons of the arcuate nucleus. In hypothalamic explants, stimulation by ghrelin increased PRCP concentration in the medium and decreased PRCP content in synaptic extract, suggesting that PRCP is released at the synaptic level. In support of this, hypothalamic explants from mice with selective deletion of PRCP in AgRP neurons (PrcpAgRPKO) showed reduced ghrelin-induced PRCP concentration in the medium compared to controls mice. Furthermore, male PrcpAgRPKO mice had decreased body weight and fat mass compared to controls. However, this phenotype was sex-specific as female PrcpAgRPKO mice show metabolic differences only when challenged by high fat diet feeding. The improved metabolism of PrcpAgRPKO mice was associated with reduced food intake and increased energy expenditure, locomotor activity, and hypothalamic α-MSH levels. Administration of SHU9119, a potent melanocortin receptor antagonist, selectively in the PVN of PrcpAgRPKO male mice increased food intake to a level similar to that of control mice. Conclusions Altogether, our data indicate that PRCP is released at the synaptic levels and that PRCP in AgRP neurons contributes to the modulation of α-MSH degradation and related metabolic control in mice. PRCP is expressed in the arcuate NPY/AgRP neurons. PRCP is released in the synaptic space following ghrelin stimulation. Male mice with PRCP deletion in NPY/AgRP neurons show leaner phenotype with decreased food intake on standard chow diet. Female mice with PRCP deletion in NPY/AgRP neurons show leaner phenotype with decreased food intake only on high fat diet. Selective blockade of PVN melanocortin receptors increases feeding in male mice with PRCP deletion in NPY/AgRP neurons.
Collapse
|
30
|
Cadherin-10 Maintains Excitatory/Inhibitory Ratio through Interactions with Synaptic Proteins. J Neurosci 2017; 37:11127-11139. [PMID: 29030434 DOI: 10.1523/jneurosci.1153-17.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/12/2017] [Accepted: 10/02/2017] [Indexed: 11/21/2022] Open
Abstract
Appropriate excitatory/inhibitory (E/I) balance is essential for normal cortical function and is altered in some psychiatric disorders, including autism spectrum disorders (ASDs). Cell-autonomous molecular mechanisms that control the balance of excitatory and inhibitory synapse function remain poorly understood; no proteins that regulate excitatory and inhibitory synapse strength in a coordinated reciprocal manner have been identified. Using super-resolution imaging, electrophysiology, and molecular manipulations, we show that cadherin-10, encoded by CDH10 within the ASD risk locus 5p14.1, maintains both excitatory and inhibitory synaptic scaffold structure in cultured cortical neurons from rats of both sexes. Cadherin-10 localizes to both excitatory and inhibitory synapses in neocortex, where it is organized into nanoscale puncta that influence the size of their associated PSDs. Knockdown of cadherin-10 reduces excitatory but increases inhibitory synapse size and strength, altering the E/I ratio in cortical neurons. Furthermore, cadherin-10 exhibits differential participation in complexes with PSD-95 and gephyrin, which may underlie its role in maintaining the E/I ratio. Our data provide a new mechanism whereby a protein encoded by a common ASD risk factor controls E/I ratios by regulating excitatory and inhibitory synapses in opposing directions.SIGNIFICANCE STATEMENT The correct balance between excitatory/inhibitory (E/I) is crucial for normal brain function and is altered in psychiatric disorders such as autism. However, the molecular mechanisms that underlie this balance remain elusive. To address this, we studied cadherin-10, an adhesion protein that is genetically linked to autism and understudied at the cellular level. Using a combination of advanced microscopy techniques and electrophysiology, we show that cadherin-10 forms nanoscale puncta at excitatory and inhibitory synapses, maintains excitatory and inhibitory synaptic structure, and is essential for maintaining the correct balance between excitation and inhibition in neuronal dendrites. These findings reveal a new mechanism by which E/I balance is controlled in neurons and may bear relevance to synaptic dysfunction in autism.
Collapse
|
31
|
Basu R, Duan X, Taylor MR, Martin EA, Muralidhar S, Wang Y, Gangi-Wellman L, Das SC, Yamagata M, West PJ, Sanes JR, Williams ME. Heterophilic Type II Cadherins Are Required for High-Magnitude Synaptic Potentiation in the Hippocampus. Neuron 2017; 96:160-176.e8. [PMID: 28957665 DOI: 10.1016/j.neuron.2017.09.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 08/03/2017] [Accepted: 09/11/2017] [Indexed: 11/26/2022]
Abstract
Hippocampal CA3 neurons form synapses with CA1 neurons in two layers, stratum oriens (SO) and stratum radiatum (SR). Each layer develops unique synaptic properties but molecular mechanisms that mediate these differences are unknown. Here, we show that SO synapses normally have significantly more mushroom spines and higher-magnitude long-term potentiation (LTP) than SR synapses. Further, we discovered that these differences require the Type II classic cadherins, cadherins-6, -9, and -10. Though cadherins typically function via trans-cellular homophilic interactions, our results suggest presynaptic cadherin-9 binds postsynaptic cadherins-6 and -10 to regulate mushroom spine density and high-magnitude LTP in the SO layer. Loss of these cadherins has no effect on the lower-magnitude LTP typically observed in the SR layer, demonstrating that cadherins-6, -9, and -10 are gatekeepers for high-magnitude LTP. Thus, Type II cadherins may uniquely contribute to the specificity and strength of synaptic changes associated with learning and memory.
Collapse
Affiliation(s)
- Raunak Basu
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Xin Duan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA 94117, USA
| | - Matthew R Taylor
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - E Anne Martin
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Shruti Muralidhar
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Yueqi Wang
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Luke Gangi-Wellman
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Sujan C Das
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Masahito Yamagata
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Peter J West
- Department of Pharmacology and Toxicology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Megan E Williams
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
32
|
Kratochwil CF, Maheshwari U, Rijli FM. The Long Journey of Pontine Nuclei Neurons: From Rhombic Lip to Cortico-Ponto-Cerebellar Circuitry. Front Neural Circuits 2017; 11:33. [PMID: 28567005 PMCID: PMC5434118 DOI: 10.3389/fncir.2017.00033] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/28/2017] [Indexed: 01/26/2023] Open
Abstract
The pontine nuclei (PN) are the largest of the precerebellar nuclei, neuronal assemblies in the hindbrain providing principal input to the cerebellum. The PN are predominantly innervated by the cerebral cortex and project as mossy fibers to the cerebellar hemispheres. Here, we comprehensively review the development of the PN from specification to migration, nucleogenesis and circuit formation. PN neurons originate at the posterior rhombic lip and migrate tangentially crossing several rhombomere derived territories to reach their final position in ventral part of the pons. The developing PN provide a classical example of tangential neuronal migration and a study system for understanding its molecular underpinnings. We anticipate that understanding the mechanisms of PN migration and assembly will also permit a deeper understanding of the molecular and cellular basis of cortico-cerebellar circuit formation and function.
Collapse
Affiliation(s)
- Claudius F Kratochwil
- Chair in Zoology and Evolutionary Biology, Department of Biology, University of KonstanzKonstanz, Germany.,Zukunftskolleg, University of KonstanzKonstanz, Germany
| | - Upasana Maheshwari
- Friedrich Miescher Institute for Biomedical ResearchBasel, Switzerland.,University of BaselBasel, Switzerland
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical ResearchBasel, Switzerland.,University of BaselBasel, Switzerland
| |
Collapse
|
33
|
Analysis of gene expression in the nervous system identifies key genes and novel candidates for health and disease. Neurogenetics 2017; 18:81-95. [PMID: 28190221 PMCID: PMC5359387 DOI: 10.1007/s10048-017-0509-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/20/2017] [Indexed: 01/09/2023]
Abstract
The incidence of neurodegenerative diseases in the developed world has risen over the last century, concomitant with an increase in average human lifespan. A major challenge is therefore to identify genes that control neuronal health and viability with a view to enhancing neuronal health during ageing and reducing the burden of neurodegeneration. Analysis of gene expression data has recently been used to infer gene functions for a range of tissues from co-expression networks. We have now applied this approach to transcriptomic datasets from the mammalian nervous system available in the public domain. We have defined the genes critical for influencing neuronal health and disease in different neurological cell types and brain regions. The functional contribution of genes in each co-expression cluster was validated using human disease and knockout mouse phenotypes, pathways and gene ontology term annotation. Additionally a number of poorly annotated genes were implicated by this approach in nervous system function. Exploiting gene expression data available in the public domain allowed us to validate key nervous system genes and, importantly, to identify additional genes with minimal functional annotation but with the same expression pattern. These genes are thus novel candidates for a role in neurological health and disease and could now be further investigated to confirm their function and regulation during ageing and neurodegeneration.
Collapse
|
34
|
Byun H, Kwon S, Ahn HJ, Liu H, Forrest D, Demb JB, Kim IJ. Molecular features distinguish ten neuronal types in the mouse superficial superior colliculus. J Comp Neurol 2016; 524:2300-21. [PMID: 26713509 DOI: 10.1002/cne.23952] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/12/2015] [Accepted: 12/17/2015] [Indexed: 01/24/2023]
Abstract
The superior colliculus (SC) is a midbrain center involved in controlling head and eye movements in response to inputs from multiple sensory modalities. Visual inputs arise from both the retina and visual cortex and converge onto the superficial layer of the SC (sSC). Neurons in the sSC send information to deeper layers of the SC and to thalamic nuclei that modulate visually guided behaviors. Presently, our understanding of sSC neurons is impeded by a lack of molecular markers that define specific cell types. To better understand the identity and organization of sSC neurons, we took a systematic approach to investigate gene expression within four molecular families: transcription factors, cell adhesion molecules, neuropeptides, and calcium binding proteins. Our analysis revealed 12 molecules with distinct expression patterns in mouse sSC: cadherin 7, contactin 3, netrin G2, cadherin 6, protocadherin 20, retinoid-related orphan receptor β, brain-specific homeobox/POU domain protein 3b, Ets variant gene 1, substance P, somatostatin, vasoactive intestinal polypeptide, and parvalbumin. Double labeling experiments, by either in situ hybridization or immunostaining, demonstrated that the 12 molecular markers collectively define 10 different sSC neuronal types. The characteristic positions of these cell types divide the sSC into four distinct layers. The 12 markers identified here will serve as valuable tools to examine molecular mechanisms that regulate development of sSC neuronal types. These markers could also be used to examine the connections between specific cell types that form retinocollicular, corticocollicular, or colliculothalamic pathways. J. Comp. Neurol. 524:2300-2321, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Haewon Byun
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, 06511
| | - Soohyun Kwon
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, 06511
| | - Hee-Jeong Ahn
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, 06511
| | - Hong Liu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Jonathan B Demb
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, 06511.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, 06511
| | - In-Jung Kim
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, 06511.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, 06511
| |
Collapse
|
35
|
Abstract
Neurons are highly polarized specialized cells. Neuronal integrity and functional roles are critically dependent on dendritic architecture and synaptic structure, function and plasticity. The cadherins are glycosylated transmembrane proteins that form cell adhesion complexes in various tissues. They are associated with a group of cytosolic proteins, the catenins. While the functional roles of the complex have been extensively investigates in non-neuronal cells, it is becoming increasingly clear that components of the complex have critical roles in regulating dendritic and synaptic architecture, function and plasticity in neurons. Consistent with these functional roles, aberrations in components of the complex have been implicated in a variety of neurodevelopmental disorders. In this review, we discuss the roles of the classical cadherins and catenins in various aspects of dendrite and synapse architecture and function and their relevance to human neurological disorders. Cadherins are glycosylated transmembrane proteins that were initially identified as Ca(2+)-dependent cell adhesion molecules. They are present on plasma membrane of a variety of cell types from primitive metazoans to humans. In the past several years, it has become clear that in addition to providing mechanical adhesion between cells, cadherins play integral roles in tissue morphogenesis and homeostasis. The cadherin family is composed of more than 100 members and classified into several subfamilies, including classical cadherins and protocadherins. Several of these cadherin family members have been implicated in various aspects of neuronal development and function. (1-3) The classical cadherins are associated with a group of cytosolic proteins, collectively called the catenins. While the functional roles of the cadherin-catenin cell adhesion complex have been extensively investigated in epithelial cells, it is now clear that components of the complex are well expressed in central neurons at different stages during development. (4,5) Recent exciting studies have shed some light on the functional roles of cadherins and catenins in central neurons. In this review, we will provide a brief overview of the cadherin superfamily, describe cadherin family members expressed in central neurons, cadherin-catenin complexes in central neurons and then focus on role of the cadherin-catenin complex in dendrite morphogenesis and synapse morphogenesis, function and plasticity. The final section is dedicated to discussion of the emerging list of neural disorders linked to cadherins and catenins. While the roles of cadherins and catenins have been examined in several different types of neurons, the focus of this review is their role in mammalian central neurons, particularly those of the cortex and hippocampus. Accompanying this review is a series of excellent reviews targeting the roles of cadherins and protocadherins in other aspects of neural development.
Collapse
Affiliation(s)
- Eunju Seong
- a Developmental Neuroscience; Munroe-Meyer Institute; University of Nebraska Medical Center ; Omaha , NE USA
| | | | | |
Collapse
|
36
|
Lin J, Wang C, Yang C, Fu S, Redies C. Pax3 and Pax7 interact reciprocally and regulate the expression of cadherin-7 through inducing neuron differentiation in the developing chicken spinal cord. J Comp Neurol 2015; 524:940-62. [PMID: 26287727 DOI: 10.1002/cne.23885] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 01/12/2023]
Abstract
Pax3 and Pax7 are closely related transcription factors that are widely expressed in the developing nervous system and somites. In the CNS, both genes are expressed in the dorsal part of the neural tube during development. Pax3 and Pax7 are involved in the sonic hedgehog (Shh) signaling pathway and are inhibited by Shh overexpression. The present study confirms in vivo that Pax3 overexpression represses the expression of Pax7, whereas Pax7 overexpression endogenously enhances and ectopically induces the expression of Pax3 in the developing chicken spinal cord. Overexpression of Pax3 and Pax7 represses the endogenous expression of cadherin-7, a member of the cadherin family of morphogenetic genes, and induces its ectopic expression. The present study also shows that overexpression of Pax3 and Pax7 changes the fate and morphology of cells in the neuroepithelial layer and induces the expression of postmitotic neuronal markers. We show that both Pax3 and Pax7 promote the differentiation of neural progenitor cells into neurons. Furthermore, the downregulation of Pax3 and Pax7 with specific shRNAs results in apoptosis in the developing spinal cord. Collectively, these results suggest that the transcription factors Pax3 and Pax7 play important roles in regulating morphogenesis and cell differentiation in the developing spinal cord.
Collapse
Affiliation(s)
- Juntang Lin
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany.,College of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Congrui Wang
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany.,College of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Ciqing Yang
- College of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Sulei Fu
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany
| | - Christoph Redies
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany
| |
Collapse
|
37
|
Sokoloff G, Plumeau AM, Mukherjee D, Blumberg MS. Twitch-related and rhythmic activation of the developing cerebellar cortex. J Neurophysiol 2015; 114:1746-56. [PMID: 26156383 PMCID: PMC4571769 DOI: 10.1152/jn.00284.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/03/2015] [Indexed: 02/08/2023] Open
Abstract
The cerebellum is a critical sensorimotor structure that exhibits protracted postnatal development in mammals. Many aspects of cerebellar circuit development are activity dependent, but little is known about the nature and sources of the activity. Based on previous findings in 6-day-old rats, we proposed that myoclonic twitches, the spontaneous movements that occur exclusively during active sleep (AS), provide generalized as well as topographically precise activity to the developing cerebellum. Taking advantage of known stages of cerebellar cortical development, we examined the relationship between Purkinje cell activity (including complex and simple spikes), nuchal and hindlimb EMG activity, and behavioral state in unanesthetized 4-, 8-, and 12-day-old rats. AS-dependent increases in complex and simple spike activity peaked at 8 days of age, with 60% of units exhibiting significantly more activity during AS than wakefulness. Also, at all three ages, approximately one-third of complex and simple spikes significantly increased their activity within 100 ms of twitches in one of the two muscles from which we recorded. Finally, we observed rhythmicity of complex and simple spikes that was especially prominent at 8 days of age and was greatly diminished by 12 days of age, likely due to developmental changes in climbing fiber and mossy fiber innervation patterns. All together, these results indicate that the neurophysiological activity of the developing cerebellum can be used to make inferences about changes in its microcircuitry. They also support the hypothesis that sleep-related twitches are a prominent source of discrete climbing and mossy fiber activity that could contribute to the activity-dependent development of this critical sensorimotor structure.
Collapse
Affiliation(s)
- Greta Sokoloff
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa; DeLTA Center, University of Iowa, Iowa City, Iowa;
| | - Alan M Plumeau
- Interdisciplinary Program in Neuroscience, University of Iowa, Iowa City, Iowa; and
| | - Didhiti Mukherjee
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa; DeLTA Center, University of Iowa, Iowa City, Iowa
| | - Mark S Blumberg
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa; DeLTA Center, University of Iowa, Iowa City, Iowa; Department of Biology, University of Iowa, Iowa City, Iowa
| |
Collapse
|
38
|
Matsunaga E, Nambu S, Oka M, Iriki A. Complex and dynamic expression of cadherins in the embryonic marmoset cerebral cortex. Dev Growth Differ 2015; 57:474-483. [PMID: 26081465 PMCID: PMC4744772 DOI: 10.1111/dgd.12228] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 05/05/2015] [Accepted: 05/09/2015] [Indexed: 01/14/2023]
Abstract
Cadherin is a cell adhesion molecule widely expressed in the nervous system. Previously, we analyzed the expression of nine classic cadherins (Cdh4, Cdh6, Cdh7, Cdh8, Cdh9, Cdh10, Cdh11, Cdh12, and Cdh20) and T-cadherin (Cdh13) in the developing postnatal common marmoset (Callithrix jacchus) brain, and found differential expressions between mice and marmosets. In this study, to explore primate-specific cadherin expression at the embryonic stage, we extensively analyzed the expression of these cadherins in the developing embryonic marmoset brain. Each cadherin showed differential spatial and temporal expression and exhibited temporally complicated expression. Furthermore, the expression of some cadherins differed from that in rodent brains, even at the embryonic stage. These results suggest the possibility that the differential expressions of diverse cadherins are involved in primate specific cortical development, from the prenatal to postnatal period.
Collapse
Affiliation(s)
- Eiji Matsunaga
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako, 351-0198, Japan
| | - Sanae Nambu
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako, 351-0198, Japan
| | - Mariko Oka
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako, 351-0198, Japan
| | - Atsushi Iriki
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako, 351-0198, Japan
| |
Collapse
|
39
|
Abstract
During brain development, billions of neurons organize into highly specific circuits. To form specific circuits, neurons must build the appropriate types of synapses with appropriate types of synaptic partners while avoiding incorrect partners in a dense cellular environment. Defining the cellular and molecular rules that govern specific circuit formation has significant scientific and clinical relevance because fine scale connectivity defects are thought to underlie many cognitive and psychiatric disorders. Organizing specific neural circuits is an enormously complicated developmental process that requires the concerted action of many molecules, neural activity, and temporal events. This review focuses on one class of molecules postulated to play an important role in target selection and specific synapse formation: the classic cadherins. Cadherins have a well-established role in epithelial cell adhesion, and although it has long been appreciated that most cadherins are expressed in the brain, their role in synaptic specificity is just beginning to be unraveled. Here, we review past and present studies implicating cadherins as active participants in the formation, function, and dysfunction of specific neural circuits and pose some of the major remaining questions.
Collapse
Affiliation(s)
- Raunak Basu
- a Department of Neurobiology and Anatomy ; University of Utah ; Salt Lake City , UT USA
| | | | | |
Collapse
|