From Wikipedia, the free encyclopedia

Neurexin family
Identifiers
SymbolNRXN1_fam
Membranome 15
neurexin 1
3D ribbon diagram of alpha-neurexin 1
Identifiers
Symbol NRXN1
NCBI gene 9378
HGNC 8008
OMIM 600565
RefSeq NM_001135659.1
UniProt Q9ULB1
Other data
Locus Chr. 2 p16.3
Search for
Structures Swiss-model
Domains InterPro
neurexin 2
Identifiers
Symbol NRXN2
NCBI gene 9379
HGNC 8009
OMIM 600566
RefSeq NM_015080
UniProt P58401
Other data
Locus Chr. 11 q13.1
Search for
Structures Swiss-model
Domains InterPro
neurexin 3
Identifiers
Symbol NRXN3
NCBI gene 9369
HGNC 8010
OMIM 600567
RefSeq NM_001105250
UniProt Q9HDB5
Other data
Locus Chr. 14 q31
Search for
Structures Swiss-model
Domains InterPro
neurexin
Identifiers
Organism Drosophila melanogaster
SymbolNrx-IV
Entrez 39387
RefSeq (mRNA) NM_168491.3
RefSeq (Prot) NP_524034.2
UniProt Q94887
Other data
Chromosome 3L: 12.14 - 12.15 Mb
Search for
Structures Swiss-model
Domains InterPro
neurexin
Identifiers
Organism Mus musculus
SymbolNrxn1
Entrez 18189
RefSeq (mRNA) NM_177284.2
RefSeq (Prot) NP_064648.3
UniProt Q9CS84
Other data
Chromosome 17: 90.03 - 91.09 Mb
Search for
Structures Swiss-model
Domains InterPro

Neurexins (NRXN) are a family of presynaptic cell adhesion proteins that have roles in connecting neurons at the synapse. [1] They are located mostly on the presynaptic membrane and contain a single transmembrane domain. The extracellular domain interacts with proteins in the synaptic cleft, most notably neuroligin, while the intracellular cytoplasmic portion interacts with proteins associated with exocytosis. [2] Neurexin and neuroligin "shake hands," resulting in the connection between the two neurons and the production of a synapse. [3] Neurexins mediate signaling across the synapse, and influence the properties of neural networks by synapse specificity. [4] Neurexins were discovered as receptors for α-latrotoxin, a vertebrate-specific toxin in black widow spider venom that binds to presynaptic receptors and induces massive neurotransmitter release. [5] In humans, alterations in genes encoding neurexins are implicated in autism and other cognitive diseases, such as Tourette syndrome and schizophrenia. [5]

Structure

In mammals, neurexin is encoded by three different genes ( NRXN1, NRXN2, and NRXN3) each controlled by two different promoters, an upstream alpha (α) and a downstream beta (β), resulting in alpha-neurexins 1-3 (α-neurexins 1–3) and beta-neurexins 1-3 (β-neurexins 1–3). [6] In addition, there are alternative splicing at 5 sites in α-neurexin and 2 in β-neurexin; more than 2000 splice variants are possible, suggesting its role in determining synapse specificity. [7]

The encoded proteins are structurally similar to laminin, slit, and agrin, other proteins involved in axon guidance and synaptogenesis. [7] α-Neurexins and β-neurexins have identical intracellular domains but different extracellular domains. The extracellular domain of α-neurexin is composed of three neurexin repeats which each contain LNS (laminin, neurexin, sex-hormone binding globulin) – EGF (epidermal growth factor) – LNS domains. N1α binds to a variety of ligands including neuroligins and GABA receptors, [2] though neurons of every receptor type express neurexins. β-Neurexins are shorter versions of α-neurexins, containing only one LNS domain. [8] β-Neurexins (located presynaptically) act as receptors for neuroligin (located postsynaptically). Additionally, β-Neurexin has also been found to play a role in angiogenesis. [9]

The C terminus of the short intracellular section of both types of neurexins binds to synaptotagmin and to the PDZ (postsynaptic density (PSD)-95/discs large/zona-occludens-1) domains of CASK and Mint. These interactions form connections between intracellular synaptic vesicles and fusion proteins. [10] Thus neurexins play an important role in assembling presynaptic and postsynaptic machinery.

Trans-synapse, the extracellular LNS domains have a functional region, the hyper-variable surface, formed by loops carrying 3 splice inserts. [2] This region surrounds a coordinated Ca2+ ion and is the site of neuroligin binding, [10] resulting in a neurexin-neuroligin Ca2+-dependent complex at the junction of chemical synapses. [11]

Expression and function

Neurexins are diffusely distributed in neurons and become concentrated at presynaptic terminals as neurons mature. They have also been found at pancreatic beta islet cells even though the function at this location has yet to be elucidated. [4] There exists a trans-synaptic dialog between neurexin and neuroligin. [12] This bi-directional trigger aids in the formation of synapses and is a key component to modifying the neuronal network. Over-expression of either of these proteins causes an increase in synapse forming sites, thus providing evidence that neurexin plays a functional role in synaptogenesis. [8] Conversely, the blocking of β-neurexin interactions reduces the number of excitatory and inhibitory synapses. It is not clear how exactly neurexin promotes the formation of synapses. One possibility is that actin is polymerized on the tail end of β-neurexin, which traps and stabilizes accumulating synaptic vesicles. This forms a forward feeding cycle, where small clusters of β-neurexins recruit more β-neurexins and scaffolding proteins to form a large synaptic adhesive contact. [8]

Neurexin Binding Partners

Neurexin-Neureoligin binding

The trans-synaptic dialog between neurexin and neuroligin organizes the apposition of pre- and post-synaptic machinery by recruiting scaffolding proteins and other synaptic elements such as NMDA receptors, CASK, and synaptotagmin, all of which are necessary for a synapse to exist.

The different combinations of neurexin to neuroligin, and alternative splicing of neuroligin and neurexin genes, control binding between neuroligins and neurexins, adding to synapse specificity. [8] Neurexins alone are capable of recruiting neuroligins in postsynaptic cells to a dendritic surface, resulting in clustered neurotransmitter receptors and other postsynaptic proteins and machinery. Their neuroligin partners can induce presynaptic terminals by recruiting neurexins. Synapse formation can therefore be triggered in either direction by these proteins. [10] Neuroligins and neurexins can also regulate formation of glutamatergic (excitatory) synapses, and GABAergic (inhibitory) contacts using a neuroligin link. Regulating these contacts suggests neurexin-neuroligin binding could balance synaptic input, [7] or maintain an optimal ratio of excitatory to inhibitory contacts.

Additional interacting partners

Dystroglycans

Neurexins not only bind to neuroligin. Additional binding partners of neurexin are dystroglycan. [10] Dystroglycan is Ca2+-dependent and binds preferentially to α-neurexins on LNS domains that lack splice inserts. In mice, a deletion of dystroglycan causes long-term potentiation impairment and developmental abnormalities similar to muscular dystrophy; however baseline synaptic transmission is normal.

Neuroexophilins

Representation of Neurexin and binding partners in the synaptic cleft

Neuroexophilins are also known to bind to neurexins and are present at the synaptic cleft but are not membrane bound. [10] [13] Neuroexophilins are Ca2+-independent and bind exclusively to α-neurexins on the second LNS domain. The increased startle responses and impaired motor coordination of neuroexophilin knockout mice indicates that neuroexophilins have a functional role in certain circuits. [10]

Latrophilins

Latrophilins are adhesion G protein-coupled receptors that reside on the postsynaptic membrane. [13] Without latrophillins in mice a loss of excitatory synapses was experienced in pyramidal neurons. [14] Latrophillins while in association with neurexin have been shown to act as postsynaptic recognition molecules for incoming axons. [13]

Cerebellins

Cerebellins are small proteins that are secreted into the synaptic cleft where they associate with other cerebellins to form a hexamer which binds two neurexins. [15] Cerebellins bind to GluD1 and GluD2 on the postsynaptic side while bound to neurexin presynaptically. GluD1 and GluD2 are homologous to ionotropic glutamate receptors, but function as adhesion molecules instead of glutamate receptors. [13] Despite being present throughout the brain, their function is only known within the cerebellum, the structure they are named after. When removed from the cerebellum a decrease of parallel fiber synapses is observed with a loss of half of all these synapses. [16] Outside of the cerebellum the function of Cerebellin is still not clear.

LRRTMs

LRRTM is a postsynaptic protein that binds to neurexin at the same Ca2+ domain that neuroligin does despite having a distinct structure. [4] It has also been found that LRRTM binds AMPA receptors. [13] This is believed to be what causes the loss of excitatory signaling when LRRTM is not present. [17] Much is still not known about LRRTM even though it is the binding partner that binds to neurexin with the highest affinity. [18]

C1q1s

C1Q1's structure is similar to that of cerebellin as it is a small protein that is secreted that associates with multiple copies of itself. [13] C1q1 while in the synaptic cleft binds neurexin on the presynaptic side and BAI3 which is another adhesion G protein-coupled receptor. The deletion of c1q1 causes the loss of climbing fibers and excitatory signaling in general. [19] C1q1s are found broadly throughout the brain including the prefrontal cortex, amygdala, cerebellum, and potentially more. [20]

Species distribution

Members of the neurexin family are found across all animals, including basal metazoans such as porifera (sponges), cnidaria (jellyfish) and ctenophora (comb jellies). Porifera lack synapses so its role in these organisms is unclear.

Homologues of α-neurexin have also been found in several invertebrate species including Drosophila, Caenorhabditis elegans, honeybees and Aplysia. [12] In Drosophila melanogaster, NRXN genes (only one α-neurexin) are critical in the assembly of glutamatergic neuromuscular junctions but are much simpler. [6] Their functional roles in insects are likely similar to those in vertebrates. [21]

Role in synaptic maturation

Neurexin and neuroligin have been found to be active in synapse maturation and adaptation of synaptic strength. Studies in knockout mice show that the trans-synaptic binding team does not increase the number of synaptic sites, but rather increases the strength of the existing synapses. [12] Deletion of the neurexin genes in the mice significantly impaired synaptic function, but did not alter synaptic structure. This is attributed to the impairment of specific voltage gated ion channels. While neuroligin and neurexin are not required for synaptic formation, they are essential components for proper function. [12]

Clinical importance and applications

Recent studies link mutations in genes encoding neurexin and neuroligin to a spectrum of cognitive disorders, such as autism spectrum disorders (ASDs), schizophrenia, and mental retardation. [5] [22] Cognitive diseases remain difficult to understand, as they are characterized by subtle changes in a subgroup of synapses in a circuit rather than impairment of all systems in all circuits. Depending on the circuit, these subtle synapse changes may produce different neurological symptoms, leading to classification of different diseases. Counterarguments to the relationship between cognitive disorders and these mutations exist, prompting further investigation into the underlying mechanisms producing these cognitive disorders.

Autism

Autism is a neurodevelopmental disorder characterized by qualitative deficits in social behavior and communication, often including restricted, repetitive patterns of behavior. [23] It includes a subset of three disorders: childhood disintegrative disorder (CDD), Asperger syndrome (AS), and pervasive developmental disorder – not otherwise specified (PDD-NOS). A small percentage of ASD patients present with single mutations in genes encoding neuroligin-neurexin cell adhesion molecules. Neurexin is crucial to synaptic function and connectivity, as highlighted in wide spectrum of neurodevelopmental phenotypes in individuals with neurexin deletions. [22] This provides strong evidence that neurexin deletions result in increased risk of ASDs, and indicate synapse dysfunction as the possible site of autism origin. [24] Dr. Steven Clapcote et al.'s α-neurexin II (Nrxn2α) KO mice experiments demonstrate a causal role for the loss of Nrxn2α in the genesis of autism-related behaviors in mice. [25]

Schizophrenia

Schizophrenia is a debilitating neuropsychiatric illness with multiple genes and environmental exposures involved in its genesis. [26] Further research indicates that deletion of the NRXN1 gene increases the risk of schizophrenia. [27] Genomic duplications and deletions on a micro-level – known as copy number variants (CNVs) – often underlie neurodevelopmental syndromes. Genomic-wide scans suggest that individuals with schizophrenia have rare structural variants that deleted or duplicated one or more genes. [26] As these studies only indicate an increased risk, further research is necessary to elucidate the underlying mechanisms of the genesis of cognitive diseases. [28]

Intellectual disability and Tourette syndrome

Similar to schizophrenia, studies have shown that intellectual disability and Tourette syndrome are also associated with NRXN1 deletions. [5] [26] A recent study shows that NRXN genes 1-3 are essential for survival and play a pivotal and overlapping role with each other in neurodevelopment. These genes have been directly disrupted in Tourette syndrome by independent genomic rearrangements. [29] Another study suggests that NLGN4 mutations can be associated with a wide spectrum of neuropsychiatric conditions and that carriers may be affected with milder symptoms. [30]

See also

References

  1. ^ Li X, Zhang J, Cao Z, Wu J, Shi Y (September 2006). "Solution structure of GOPC PDZ domain and its interaction with the C-terminal motif of neuroligin". Protein Science. 15 (9): 2149–2158. doi: 10.1110/ps.062087506. PMC  2242614. PMID  16882988.
  2. ^ a b c Chen F, Venugopal V, Murray B, Rudenko G (June 2011). "The structure of neurexin 1α reveals features promoting a role as synaptic organizer". Structure. 19 (6): 779–789. doi: 10.1016/j.str.2011.03.012. PMC  3134934. PMID  21620716.
  3. ^ Scheiffele P, Fan J, Choih J, Fetter R, Serafini T (June 2000). "Neuroligin expressed in nonneuronal cells triggers presynaptic development in contacting axons". Cell. 101 (6): 657–669. doi: 10.1016/S0092-8674(00)80877-6. PMID  10892652. S2CID  16095623.
  4. ^ a b c Reissner C, Runkel F, Missler M (2013). "Neurexins". Genome Biology. 14 (9): 213. doi: 10.1186/gb-2013-14-9-213. PMC  4056431. PMID  24083347.
  5. ^ a b c d Südhof TC (October 2008). "Neuroligins and neurexins link synaptic function to cognitive disease". Nature. 455 (7215): 903–911. Bibcode: 2008Natur.455..903S. doi: 10.1038/nature07456. PMC  2673233. PMID  18923512.
  6. ^ a b Baudouin S, Scheiffele P (May 2010). "SnapShot: Neuroligin-neurexin complexes". Cell. 141 (5): 908, 908.e1. doi: 10.1016/j.cell.2010.05.024. PMID  20510934.
  7. ^ a b c Binder MD (2009). Encyclopedia of Neuroscience: Neurexins. Springer Berlin Heidelberg. p. 2607. ISBN  978-3-540-29678-2.
  8. ^ a b c d Dean C, Dresbach T (January 2006). "Neuroligins and neurexins: linking cell adhesion, synapse formation and cognitive function". Trends in Neurosciences. 29 (1): 21–29. doi: 10.1016/j.tins.2005.11.003. PMID  16337696. S2CID  11664697.
  9. ^ Bottos A, Destro E, Rissone A, Graziano S, Cordara G, Assenzio B, et al. (December 2009). "The synaptic proteins neurexins and neuroligins are widely expressed in the vascular system and contribute to its functions". Proceedings of the National Academy of Sciences of the United States of America. 106 (49): 20782–20787. Bibcode: 2009PNAS..10620782B. doi: 10.1073/pnas.0809510106. PMC  2791601. PMID  19926856.
  10. ^ a b c d e f Craig AM, Kang Y (February 2007). "Neurexin-neuroligin signaling in synapse development". Current Opinion in Neurobiology. 17 (1): 43–52. doi: 10.1016/j.conb.2007.01.011. PMC  2820508. PMID  17275284.
  11. ^ Reissner C, Klose M, Fairless R, Missler M (September 2008). "Mutational analysis of the neurexin/neuroligin complex reveals essential and regulatory components". Proceedings of the National Academy of Sciences of the United States of America. 105 (39): 15124–15129. Bibcode: 2008PNAS..10515124R. doi: 10.1073/pnas.0801639105. PMC  2551626. PMID  18812509.
  12. ^ a b c d Knight D, Xie W, Boulianne GL (December 2011). "Neurexins and neuroligins: recent insights from invertebrates". Molecular Neurobiology. 44 (3): 426–440. doi: 10.1007/s12035-011-8213-1. PMC  3229692. PMID  22037798.
  13. ^ a b c d e f Südhof TC (November 2017). "Synaptic Neurexin Complexes: A Molecular Code for the Logic of Neural Circuits". Cell. 171 (4): 745–769. doi: 10.1016/j.cell.2017.10.024. PMC  5694349. PMID  29100073.
  14. ^ Anderson GR, Maxeiner S, Sando R, Tsetsenis T, Malenka RC, Südhof TC (November 2017). "Postsynaptic adhesion GPCR latrophilin-2 mediates target recognition in entorhinal-hippocampal synapse assembly". The Journal of Cell Biology. 216 (11): 3831–3846. doi: 10.1083/jcb.201703042. PMC  5674891. PMID  28972101.
  15. ^ Südhof TC (August 2023). "Cerebellin-neurexin complexes instructing synapse properties". Current Opinion in Neurobiology. 81: 102727. doi: 10.1016/j.conb.2023.102727. PMID  37209532.
  16. ^ Hirai H, Pang Z, Bao D, Miyazaki T, Li L, Miura E, et al. (November 2005). "Cbln1 is essential for synaptic integrity and plasticity in the cerebellum". Nature Neuroscience. 8 (11): 1534–1541. doi: 10.1038/nn1576. PMID  16234806. S2CID  41611184.
  17. ^ Um JW, Choi TY, Kang H, Cho YS, Choii G, Uvarov P, et al. (February 2016). "LRRTM3 Regulates Excitatory Synapse Development through Alternative Splicing and Neurexin Binding". Cell Reports. 14 (4): 808–822. doi: 10.1016/j.celrep.2015.12.081. PMID  26776509.
  18. ^ Lisé MF, El-Husseini A (August 2006). "The neuroligin and neurexin families: from structure to function at the synapse". Cellular and Molecular Life Sciences. 63 (16): 1833–1849. doi: 10.1007/s00018-006-6061-3. PMID  16794786. S2CID  1720692.
  19. ^ Sigoillot SM, Iyer K, Binda F, González-Calvo I, Talleur M, Vodjdani G, et al. (February 2015). "The Secreted Protein C1QL1 and Its Receptor BAI3 Control the Synaptic Connectivity of Excitatory Inputs Converging on Cerebellar Purkinje Cells". Cell Reports. 10 (5): 820–832. doi: 10.1016/j.celrep.2015.01.034. PMID  25660030. S2CID  5215066.
  20. ^ Iijima T, Miura E, Watanabe M, Yuzaki M (May 2010). "Distinct expression of C1q-like family mRNAs in mouse brain and biochemical characterization of their encoded proteins". The European Journal of Neuroscience. 31 (9): 1606–1615. doi: 10.1111/j.1460-9568.2010.07202.x. PMID  20525073. S2CID  7273855.
  21. ^ Biswas S, Russell RJ, Jackson CJ, Vidovic M, Ganeshina O, Oakeshott JG, Claudianos C (2008). "Bridging the synaptic gap: neuroligins and neurexin I in Apis mellifera". PLOS ONE. 3 (10): e3542. Bibcode: 2008PLoSO...3.3542B. doi: 10.1371/journal.pone.0003542. PMC  2570956. PMID  18974885.
  22. ^ a b Cuttler K, Hassan M, Carr J, Cloete R, Bardien S (October 2021). "Emerging evidence implicating a role for neurexins in neurodegenerative and neuropsychiatric disorders". Open Biology. 11 (10): 210091. doi: 10.1098/rsob.210091. PMC  8492176. PMID  34610269.
  23. ^ Lord C, Cook EH, Leventhal BL, Amaral DG (November 2000). "Autism spectrum disorders". Neuron. 28 (2): 355–363. doi: 10.1016/S0896-6273(00)00115-X. PMID  11144346. S2CID  7100507.
  24. ^ Pizzarelli R, Cherubini E (2011). "Alterations of GABAergic signaling in autism spectrum disorders". Neural Plasticity. 2011: 297153. doi: 10.1155/2011/297153. PMC  3134996. PMID  21766041.
  25. ^ Dachtler J, Glasper J, Cohen RN, Ivorra JL, Swiffen DJ, Jackson AJ, et al. (November 2014). "Deletion of α-neurexin II results in autism-related behaviors in mice". Translational Psychiatry. 4 (11): e484. doi: 10.1038/tp.2014.123. PMC  4259993. PMID  25423136.
  26. ^ a b c Walsh T, McClellan JM, McCarthy SE, Addington AM, Pierce SB, Cooper GM, et al. (April 2008). "Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia". Science. 320 (5875): 539–543. Bibcode: 2008Sci...320..539W. doi: 10.1126/science.1155174. PMID  18369103. S2CID  14385126.
  27. ^ Kirov G, Rujescu D, Ingason A, Collier DA, O'Donovan MC, Owen MJ (September 2009). "Neurexin 1 (NRXN1) deletions in schizophrenia". Schizophrenia Bulletin. 35 (5): 851–854. doi: 10.1093/schbul/sbp079. PMC  2728827. PMID  19675094.
  28. ^ Kirov G, Gumus D, Chen W, Norton N, Georgieva L, Sari M, et al. (February 2008). "Comparative genome hybridization suggests a role for NRXN1 and APBA2 in schizophrenia". Human Molecular Genetics. 17 (3): 458–465. doi: 10.1093/hmg/ddm323. hdl: 11858/00-001M-0000-0010-8041-A. PMID  17989066.
  29. ^ Boschero AC, Negreiros de Paiva CE (1977). "Transplantation of islets of Langerhans in diabetic rats". Acta Physiologica Latino Americana. 27 (1): 1–6. PMID  356520.
  30. ^ Kuttig P (September 1970). "[Social service for paraplegics and childhood paralytics]". Deutsche Schwesternzeitung (in German). 23 (9): 450–452. PMID  5202006.

External links

From Wikipedia, the free encyclopedia

Neurexin family
Identifiers
SymbolNRXN1_fam
Membranome 15
neurexin 1
3D ribbon diagram of alpha-neurexin 1
Identifiers
Symbol NRXN1
NCBI gene 9378
HGNC 8008
OMIM 600565
RefSeq NM_001135659.1
UniProt Q9ULB1
Other data
Locus Chr. 2 p16.3
Search for
Structures Swiss-model
Domains InterPro
neurexin 2
Identifiers
Symbol NRXN2
NCBI gene 9379
HGNC 8009
OMIM 600566
RefSeq NM_015080
UniProt P58401
Other data
Locus Chr. 11 q13.1
Search for
Structures Swiss-model
Domains InterPro
neurexin 3
Identifiers
Symbol NRXN3
NCBI gene 9369
HGNC 8010
OMIM 600567
RefSeq NM_001105250
UniProt Q9HDB5
Other data
Locus Chr. 14 q31
Search for
Structures Swiss-model
Domains InterPro
neurexin
Identifiers
Organism Drosophila melanogaster
SymbolNrx-IV
Entrez 39387
RefSeq (mRNA) NM_168491.3
RefSeq (Prot) NP_524034.2
UniProt Q94887
Other data
Chromosome 3L: 12.14 - 12.15 Mb
Search for
Structures Swiss-model
Domains InterPro
neurexin
Identifiers
Organism Mus musculus
SymbolNrxn1
Entrez 18189
RefSeq (mRNA) NM_177284.2
RefSeq (Prot) NP_064648.3
UniProt Q9CS84
Other data
Chromosome 17: 90.03 - 91.09 Mb
Search for
Structures Swiss-model
Domains InterPro

Neurexins (NRXN) are a family of presynaptic cell adhesion proteins that have roles in connecting neurons at the synapse. [1] They are located mostly on the presynaptic membrane and contain a single transmembrane domain. The extracellular domain interacts with proteins in the synaptic cleft, most notably neuroligin, while the intracellular cytoplasmic portion interacts with proteins associated with exocytosis. [2] Neurexin and neuroligin "shake hands," resulting in the connection between the two neurons and the production of a synapse. [3] Neurexins mediate signaling across the synapse, and influence the properties of neural networks by synapse specificity. [4] Neurexins were discovered as receptors for α-latrotoxin, a vertebrate-specific toxin in black widow spider venom that binds to presynaptic receptors and induces massive neurotransmitter release. [5] In humans, alterations in genes encoding neurexins are implicated in autism and other cognitive diseases, such as Tourette syndrome and schizophrenia. [5]

Structure

In mammals, neurexin is encoded by three different genes ( NRXN1, NRXN2, and NRXN3) each controlled by two different promoters, an upstream alpha (α) and a downstream beta (β), resulting in alpha-neurexins 1-3 (α-neurexins 1–3) and beta-neurexins 1-3 (β-neurexins 1–3). [6] In addition, there are alternative splicing at 5 sites in α-neurexin and 2 in β-neurexin; more than 2000 splice variants are possible, suggesting its role in determining synapse specificity. [7]

The encoded proteins are structurally similar to laminin, slit, and agrin, other proteins involved in axon guidance and synaptogenesis. [7] α-Neurexins and β-neurexins have identical intracellular domains but different extracellular domains. The extracellular domain of α-neurexin is composed of three neurexin repeats which each contain LNS (laminin, neurexin, sex-hormone binding globulin) – EGF (epidermal growth factor) – LNS domains. N1α binds to a variety of ligands including neuroligins and GABA receptors, [2] though neurons of every receptor type express neurexins. β-Neurexins are shorter versions of α-neurexins, containing only one LNS domain. [8] β-Neurexins (located presynaptically) act as receptors for neuroligin (located postsynaptically). Additionally, β-Neurexin has also been found to play a role in angiogenesis. [9]

The C terminus of the short intracellular section of both types of neurexins binds to synaptotagmin and to the PDZ (postsynaptic density (PSD)-95/discs large/zona-occludens-1) domains of CASK and Mint. These interactions form connections between intracellular synaptic vesicles and fusion proteins. [10] Thus neurexins play an important role in assembling presynaptic and postsynaptic machinery.

Trans-synapse, the extracellular LNS domains have a functional region, the hyper-variable surface, formed by loops carrying 3 splice inserts. [2] This region surrounds a coordinated Ca2+ ion and is the site of neuroligin binding, [10] resulting in a neurexin-neuroligin Ca2+-dependent complex at the junction of chemical synapses. [11]

Expression and function

Neurexins are diffusely distributed in neurons and become concentrated at presynaptic terminals as neurons mature. They have also been found at pancreatic beta islet cells even though the function at this location has yet to be elucidated. [4] There exists a trans-synaptic dialog between neurexin and neuroligin. [12] This bi-directional trigger aids in the formation of synapses and is a key component to modifying the neuronal network. Over-expression of either of these proteins causes an increase in synapse forming sites, thus providing evidence that neurexin plays a functional role in synaptogenesis. [8] Conversely, the blocking of β-neurexin interactions reduces the number of excitatory and inhibitory synapses. It is not clear how exactly neurexin promotes the formation of synapses. One possibility is that actin is polymerized on the tail end of β-neurexin, which traps and stabilizes accumulating synaptic vesicles. This forms a forward feeding cycle, where small clusters of β-neurexins recruit more β-neurexins and scaffolding proteins to form a large synaptic adhesive contact. [8]

Neurexin Binding Partners

Neurexin-Neureoligin binding

The trans-synaptic dialog between neurexin and neuroligin organizes the apposition of pre- and post-synaptic machinery by recruiting scaffolding proteins and other synaptic elements such as NMDA receptors, CASK, and synaptotagmin, all of which are necessary for a synapse to exist.

The different combinations of neurexin to neuroligin, and alternative splicing of neuroligin and neurexin genes, control binding between neuroligins and neurexins, adding to synapse specificity. [8] Neurexins alone are capable of recruiting neuroligins in postsynaptic cells to a dendritic surface, resulting in clustered neurotransmitter receptors and other postsynaptic proteins and machinery. Their neuroligin partners can induce presynaptic terminals by recruiting neurexins. Synapse formation can therefore be triggered in either direction by these proteins. [10] Neuroligins and neurexins can also regulate formation of glutamatergic (excitatory) synapses, and GABAergic (inhibitory) contacts using a neuroligin link. Regulating these contacts suggests neurexin-neuroligin binding could balance synaptic input, [7] or maintain an optimal ratio of excitatory to inhibitory contacts.

Additional interacting partners

Dystroglycans

Neurexins not only bind to neuroligin. Additional binding partners of neurexin are dystroglycan. [10] Dystroglycan is Ca2+-dependent and binds preferentially to α-neurexins on LNS domains that lack splice inserts. In mice, a deletion of dystroglycan causes long-term potentiation impairment and developmental abnormalities similar to muscular dystrophy; however baseline synaptic transmission is normal.

Neuroexophilins

Representation of Neurexin and binding partners in the synaptic cleft

Neuroexophilins are also known to bind to neurexins and are present at the synaptic cleft but are not membrane bound. [10] [13] Neuroexophilins are Ca2+-independent and bind exclusively to α-neurexins on the second LNS domain. The increased startle responses and impaired motor coordination of neuroexophilin knockout mice indicates that neuroexophilins have a functional role in certain circuits. [10]

Latrophilins

Latrophilins are adhesion G protein-coupled receptors that reside on the postsynaptic membrane. [13] Without latrophillins in mice a loss of excitatory synapses was experienced in pyramidal neurons. [14] Latrophillins while in association with neurexin have been shown to act as postsynaptic recognition molecules for incoming axons. [13]

Cerebellins

Cerebellins are small proteins that are secreted into the synaptic cleft where they associate with other cerebellins to form a hexamer which binds two neurexins. [15] Cerebellins bind to GluD1 and GluD2 on the postsynaptic side while bound to neurexin presynaptically. GluD1 and GluD2 are homologous to ionotropic glutamate receptors, but function as adhesion molecules instead of glutamate receptors. [13] Despite being present throughout the brain, their function is only known within the cerebellum, the structure they are named after. When removed from the cerebellum a decrease of parallel fiber synapses is observed with a loss of half of all these synapses. [16] Outside of the cerebellum the function of Cerebellin is still not clear.

LRRTMs

LRRTM is a postsynaptic protein that binds to neurexin at the same Ca2+ domain that neuroligin does despite having a distinct structure. [4] It has also been found that LRRTM binds AMPA receptors. [13] This is believed to be what causes the loss of excitatory signaling when LRRTM is not present. [17] Much is still not known about LRRTM even though it is the binding partner that binds to neurexin with the highest affinity. [18]

C1q1s

C1Q1's structure is similar to that of cerebellin as it is a small protein that is secreted that associates with multiple copies of itself. [13] C1q1 while in the synaptic cleft binds neurexin on the presynaptic side and BAI3 which is another adhesion G protein-coupled receptor. The deletion of c1q1 causes the loss of climbing fibers and excitatory signaling in general. [19] C1q1s are found broadly throughout the brain including the prefrontal cortex, amygdala, cerebellum, and potentially more. [20]

Species distribution

Members of the neurexin family are found across all animals, including basal metazoans such as porifera (sponges), cnidaria (jellyfish) and ctenophora (comb jellies). Porifera lack synapses so its role in these organisms is unclear.

Homologues of α-neurexin have also been found in several invertebrate species including Drosophila, Caenorhabditis elegans, honeybees and Aplysia. [12] In Drosophila melanogaster, NRXN genes (only one α-neurexin) are critical in the assembly of glutamatergic neuromuscular junctions but are much simpler. [6] Their functional roles in insects are likely similar to those in vertebrates. [21]

Role in synaptic maturation

Neurexin and neuroligin have been found to be active in synapse maturation and adaptation of synaptic strength. Studies in knockout mice show that the trans-synaptic binding team does not increase the number of synaptic sites, but rather increases the strength of the existing synapses. [12] Deletion of the neurexin genes in the mice significantly impaired synaptic function, but did not alter synaptic structure. This is attributed to the impairment of specific voltage gated ion channels. While neuroligin and neurexin are not required for synaptic formation, they are essential components for proper function. [12]

Clinical importance and applications

Recent studies link mutations in genes encoding neurexin and neuroligin to a spectrum of cognitive disorders, such as autism spectrum disorders (ASDs), schizophrenia, and mental retardation. [5] [22] Cognitive diseases remain difficult to understand, as they are characterized by subtle changes in a subgroup of synapses in a circuit rather than impairment of all systems in all circuits. Depending on the circuit, these subtle synapse changes may produce different neurological symptoms, leading to classification of different diseases. Counterarguments to the relationship between cognitive disorders and these mutations exist, prompting further investigation into the underlying mechanisms producing these cognitive disorders.

Autism

Autism is a neurodevelopmental disorder characterized by qualitative deficits in social behavior and communication, often including restricted, repetitive patterns of behavior. [23] It includes a subset of three disorders: childhood disintegrative disorder (CDD), Asperger syndrome (AS), and pervasive developmental disorder – not otherwise specified (PDD-NOS). A small percentage of ASD patients present with single mutations in genes encoding neuroligin-neurexin cell adhesion molecules. Neurexin is crucial to synaptic function and connectivity, as highlighted in wide spectrum of neurodevelopmental phenotypes in individuals with neurexin deletions. [22] This provides strong evidence that neurexin deletions result in increased risk of ASDs, and indicate synapse dysfunction as the possible site of autism origin. [24] Dr. Steven Clapcote et al.'s α-neurexin II (Nrxn2α) KO mice experiments demonstrate a causal role for the loss of Nrxn2α in the genesis of autism-related behaviors in mice. [25]

Schizophrenia

Schizophrenia is a debilitating neuropsychiatric illness with multiple genes and environmental exposures involved in its genesis. [26] Further research indicates that deletion of the NRXN1 gene increases the risk of schizophrenia. [27] Genomic duplications and deletions on a micro-level – known as copy number variants (CNVs) – often underlie neurodevelopmental syndromes. Genomic-wide scans suggest that individuals with schizophrenia have rare structural variants that deleted or duplicated one or more genes. [26] As these studies only indicate an increased risk, further research is necessary to elucidate the underlying mechanisms of the genesis of cognitive diseases. [28]

Intellectual disability and Tourette syndrome

Similar to schizophrenia, studies have shown that intellectual disability and Tourette syndrome are also associated with NRXN1 deletions. [5] [26] A recent study shows that NRXN genes 1-3 are essential for survival and play a pivotal and overlapping role with each other in neurodevelopment. These genes have been directly disrupted in Tourette syndrome by independent genomic rearrangements. [29] Another study suggests that NLGN4 mutations can be associated with a wide spectrum of neuropsychiatric conditions and that carriers may be affected with milder symptoms. [30]

See also

References

  1. ^ Li X, Zhang J, Cao Z, Wu J, Shi Y (September 2006). "Solution structure of GOPC PDZ domain and its interaction with the C-terminal motif of neuroligin". Protein Science. 15 (9): 2149–2158. doi: 10.1110/ps.062087506. PMC  2242614. PMID  16882988.
  2. ^ a b c Chen F, Venugopal V, Murray B, Rudenko G (June 2011). "The structure of neurexin 1α reveals features promoting a role as synaptic organizer". Structure. 19 (6): 779–789. doi: 10.1016/j.str.2011.03.012. PMC  3134934. PMID  21620716.
  3. ^ Scheiffele P, Fan J, Choih J, Fetter R, Serafini T (June 2000). "Neuroligin expressed in nonneuronal cells triggers presynaptic development in contacting axons". Cell. 101 (6): 657–669. doi: 10.1016/S0092-8674(00)80877-6. PMID  10892652. S2CID  16095623.
  4. ^ a b c Reissner C, Runkel F, Missler M (2013). "Neurexins". Genome Biology. 14 (9): 213. doi: 10.1186/gb-2013-14-9-213. PMC  4056431. PMID  24083347.
  5. ^ a b c d Südhof TC (October 2008). "Neuroligins and neurexins link synaptic function to cognitive disease". Nature. 455 (7215): 903–911. Bibcode: 2008Natur.455..903S. doi: 10.1038/nature07456. PMC  2673233. PMID  18923512.
  6. ^ a b Baudouin S, Scheiffele P (May 2010). "SnapShot: Neuroligin-neurexin complexes". Cell. 141 (5): 908, 908.e1. doi: 10.1016/j.cell.2010.05.024. PMID  20510934.
  7. ^ a b c Binder MD (2009). Encyclopedia of Neuroscience: Neurexins. Springer Berlin Heidelberg. p. 2607. ISBN  978-3-540-29678-2.
  8. ^ a b c d Dean C, Dresbach T (January 2006). "Neuroligins and neurexins: linking cell adhesion, synapse formation and cognitive function". Trends in Neurosciences. 29 (1): 21–29. doi: 10.1016/j.tins.2005.11.003. PMID  16337696. S2CID  11664697.
  9. ^ Bottos A, Destro E, Rissone A, Graziano S, Cordara G, Assenzio B, et al. (December 2009). "The synaptic proteins neurexins and neuroligins are widely expressed in the vascular system and contribute to its functions". Proceedings of the National Academy of Sciences of the United States of America. 106 (49): 20782–20787. Bibcode: 2009PNAS..10620782B. doi: 10.1073/pnas.0809510106. PMC  2791601. PMID  19926856.
  10. ^ a b c d e f Craig AM, Kang Y (February 2007). "Neurexin-neuroligin signaling in synapse development". Current Opinion in Neurobiology. 17 (1): 43–52. doi: 10.1016/j.conb.2007.01.011. PMC  2820508. PMID  17275284.
  11. ^ Reissner C, Klose M, Fairless R, Missler M (September 2008). "Mutational analysis of the neurexin/neuroligin complex reveals essential and regulatory components". Proceedings of the National Academy of Sciences of the United States of America. 105 (39): 15124–15129. Bibcode: 2008PNAS..10515124R. doi: 10.1073/pnas.0801639105. PMC  2551626. PMID  18812509.
  12. ^ a b c d Knight D, Xie W, Boulianne GL (December 2011). "Neurexins and neuroligins: recent insights from invertebrates". Molecular Neurobiology. 44 (3): 426–440. doi: 10.1007/s12035-011-8213-1. PMC  3229692. PMID  22037798.
  13. ^ a b c d e f Südhof TC (November 2017). "Synaptic Neurexin Complexes: A Molecular Code for the Logic of Neural Circuits". Cell. 171 (4): 745–769. doi: 10.1016/j.cell.2017.10.024. PMC  5694349. PMID  29100073.
  14. ^ Anderson GR, Maxeiner S, Sando R, Tsetsenis T, Malenka RC, Südhof TC (November 2017). "Postsynaptic adhesion GPCR latrophilin-2 mediates target recognition in entorhinal-hippocampal synapse assembly". The Journal of Cell Biology. 216 (11): 3831–3846. doi: 10.1083/jcb.201703042. PMC  5674891. PMID  28972101.
  15. ^ Südhof TC (August 2023). "Cerebellin-neurexin complexes instructing synapse properties". Current Opinion in Neurobiology. 81: 102727. doi: 10.1016/j.conb.2023.102727. PMID  37209532.
  16. ^ Hirai H, Pang Z, Bao D, Miyazaki T, Li L, Miura E, et al. (November 2005). "Cbln1 is essential for synaptic integrity and plasticity in the cerebellum". Nature Neuroscience. 8 (11): 1534–1541. doi: 10.1038/nn1576. PMID  16234806. S2CID  41611184.
  17. ^ Um JW, Choi TY, Kang H, Cho YS, Choii G, Uvarov P, et al. (February 2016). "LRRTM3 Regulates Excitatory Synapse Development through Alternative Splicing and Neurexin Binding". Cell Reports. 14 (4): 808–822. doi: 10.1016/j.celrep.2015.12.081. PMID  26776509.
  18. ^ Lisé MF, El-Husseini A (August 2006). "The neuroligin and neurexin families: from structure to function at the synapse". Cellular and Molecular Life Sciences. 63 (16): 1833–1849. doi: 10.1007/s00018-006-6061-3. PMID  16794786. S2CID  1720692.
  19. ^ Sigoillot SM, Iyer K, Binda F, González-Calvo I, Talleur M, Vodjdani G, et al. (February 2015). "The Secreted Protein C1QL1 and Its Receptor BAI3 Control the Synaptic Connectivity of Excitatory Inputs Converging on Cerebellar Purkinje Cells". Cell Reports. 10 (5): 820–832. doi: 10.1016/j.celrep.2015.01.034. PMID  25660030. S2CID  5215066.
  20. ^ Iijima T, Miura E, Watanabe M, Yuzaki M (May 2010). "Distinct expression of C1q-like family mRNAs in mouse brain and biochemical characterization of their encoded proteins". The European Journal of Neuroscience. 31 (9): 1606–1615. doi: 10.1111/j.1460-9568.2010.07202.x. PMID  20525073. S2CID  7273855.
  21. ^ Biswas S, Russell RJ, Jackson CJ, Vidovic M, Ganeshina O, Oakeshott JG, Claudianos C (2008). "Bridging the synaptic gap: neuroligins and neurexin I in Apis mellifera". PLOS ONE. 3 (10): e3542. Bibcode: 2008PLoSO...3.3542B. doi: 10.1371/journal.pone.0003542. PMC  2570956. PMID  18974885.
  22. ^ a b Cuttler K, Hassan M, Carr J, Cloete R, Bardien S (October 2021). "Emerging evidence implicating a role for neurexins in neurodegenerative and neuropsychiatric disorders". Open Biology. 11 (10): 210091. doi: 10.1098/rsob.210091. PMC  8492176. PMID  34610269.
  23. ^ Lord C, Cook EH, Leventhal BL, Amaral DG (November 2000). "Autism spectrum disorders". Neuron. 28 (2): 355–363. doi: 10.1016/S0896-6273(00)00115-X. PMID  11144346. S2CID  7100507.
  24. ^ Pizzarelli R, Cherubini E (2011). "Alterations of GABAergic signaling in autism spectrum disorders". Neural Plasticity. 2011: 297153. doi: 10.1155/2011/297153. PMC  3134996. PMID  21766041.
  25. ^ Dachtler J, Glasper J, Cohen RN, Ivorra JL, Swiffen DJ, Jackson AJ, et al. (November 2014). "Deletion of α-neurexin II results in autism-related behaviors in mice". Translational Psychiatry. 4 (11): e484. doi: 10.1038/tp.2014.123. PMC  4259993. PMID  25423136.
  26. ^ a b c Walsh T, McClellan JM, McCarthy SE, Addington AM, Pierce SB, Cooper GM, et al. (April 2008). "Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia". Science. 320 (5875): 539–543. Bibcode: 2008Sci...320..539W. doi: 10.1126/science.1155174. PMID  18369103. S2CID  14385126.
  27. ^ Kirov G, Rujescu D, Ingason A, Collier DA, O'Donovan MC, Owen MJ (September 2009). "Neurexin 1 (NRXN1) deletions in schizophrenia". Schizophrenia Bulletin. 35 (5): 851–854. doi: 10.1093/schbul/sbp079. PMC  2728827. PMID  19675094.
  28. ^ Kirov G, Gumus D, Chen W, Norton N, Georgieva L, Sari M, et al. (February 2008). "Comparative genome hybridization suggests a role for NRXN1 and APBA2 in schizophrenia". Human Molecular Genetics. 17 (3): 458–465. doi: 10.1093/hmg/ddm323. hdl: 11858/00-001M-0000-0010-8041-A. PMID  17989066.
  29. ^ Boschero AC, Negreiros de Paiva CE (1977). "Transplantation of islets of Langerhans in diabetic rats". Acta Physiologica Latino Americana. 27 (1): 1–6. PMID  356520.
  30. ^ Kuttig P (September 1970). "[Social service for paraplegics and childhood paralytics]". Deutsche Schwesternzeitung (in German). 23 (9): 450–452. PMID  5202006.

External links


Videos

Youtube | Vimeo | Bing

Websites

Google | Yahoo | Bing

Encyclopedia

Google | Yahoo | Bing

Facebook