Kalirin

Last updated
KALRN
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases KALRN , ARHGEF24, CHD5, CHDS5, DUET, DUO, HAPIP, TRAD, kalirin, RhoGEF kinase, kalirin RhoGEF kinase
External IDs OMIM: 604605 MGI: 2685385 HomoloGene: 57160 GeneCards: KALRN
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001164268
NM_177357

RefSeq (protein)

NP_001157740
NP_796331

Location (UCSC) Chr 3: 124.03 – 124.73 Mb Chr 16: 33.79 – 34.39 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Kalirin, also known as Huntingtin-associated protein-interacting protein (HAPIP), protein duo (DUO), or serine/threonine-protein kinase with Dbl- and pleckstrin homology domain, is a protein that in humans is encoded by the KALRN gene. [5] [6] Kalirin was first identified in 1997 as a protein interacting with huntingtin-associated protein 1. [5] Is also known to play an important role in nerve growth and axonal development. [7]

Kalirin is a member of the Dbl family of proteins and is a Rho guanine nucleotide exchange factor.

It is named after the multiple-handed Hindu goddess Kali for its ability to interact with numerous other proteins. Kalirin's other name, DUO, comes from the fact that it is 98% identical to rat DUO protein and 80.6% identical to a human protein named TRIO. Unlike TRIO, which is expressed in numerous tissues, Kalirin isoforms are mainly found in the brain.

Clinical significance

Several isoforms of Kalirin are produced through alternative splicing. [8] One of the isoforms, Kalirin-7, was found to be necessary for the remodeling of synapses in mature cortical neurons and is thought to be important in the development of schizophrenia, [9] [10] [11] [12] as demonstrated by adolescent development of schizophrenia-like symptoms in kalirin knockout mice. [13] Alzheimer's disease may also be linked to kalirin-7. [12] [14] [15]

The KALRN gene, has been linked to multiple neurological disorders both through large exome and genome sequencing efforts, as well as post mortem and clinical studies.

Several mutations within KALRN have been linked to neurological disease. In autism spectrum disorder a frameshift mutation was found [16] [17] that is likely to lead to transcript decay, and heterozygosity. Another, found within the second GEF domain, is predicted to be highly deleterious to RhoA-GEF activity and likely affects the function of kalirin9 and 12 isoforms early in neuronal development. [18] A patient harboring a homozygous mutation in kalirin's spectrin repeat domain was found to have severe intellectual disability, [19] and both truncating and missense mutations have been identified in patients with developmental delay. [20] Several intronic variants have been associated with addiction and were found to alter the function of brain regions responsible for reward anticipation. [21] This link to addiction is supported by animal models, where loss of kalirin results in altered cocaine self-administration and synaptic and expression changes in response to cocaine. [22] [23] [24] Perhaps the most compelling genetic links are between kalirin and schizophrenia. Numerous missense mutations in KALRN have been identified in exome sequencing studies of schizophrenia cohorts [25] that are predicted to be deleterious to protein function.

Neuronal studies have provided insight into the mechanisms of some missense mutations, particularly within the GEF domains of KALRN. A mutation found within the Rac-GEF domain was found to induce strong reductions in Rac activation, neuronal branching and spine density. [26] These effects were mirrored by mutations in the RhoA-GEF domain, producing similar neuronal deficits, but by promoting RhoA-GEF activity. [27] In addition to exome sequencing, post-mortem studies have consistently found alterations in kalirin transcript levels within the brain [28] [29] further supporting a role for kalirin in the etiology of schizophrenia.

In addition to neurodevelopmental disorders, kalirin has been found to be underexpressed in the post-mortem Alzheimer's brain. [15] [14] This loss of kalirin expression was recapitulated in animal models of Alzheimer's disease. [30] [31] Moreover, introduction of kalirin7 into culture [32] or animal models [31] of Alzheimer's disease was able to rescue synaptic and behavioral deficits, suggesting an important role for kalirin in regulating synapse loss and cognitive impairment in Alzheimer's disease.

Function

The majority of kalirin's effects are induced through its catalytic GEF domain signaling. By promoting the release of GDP from Rac and RhoA, it acts as an activator of GTPase signaling within the cell. [33] Although able to activate Rac and RhoA, much of its activity in regulating neuronal morphology has been attributed to Rac-PAK pathway activation. [34] kalirin has found been found to exert control over dendritic arborization, [35] axonal growth, [33] [36] dendritic spine formation [37] and synaptic activity [38] [13] and plasticity. [38] [39] [40] [41] These effects are regulated by protein-protein interactions and post-translational modifications within the non-catalytic domains, and have been shown to exert control over kalirin subcellular targeting and activation. [38] [37] [42]

Kalirin has been found to play a critical role in synaptic activity and plasticity. Loss of KALRN results in decreased nMDAr and AMPAr-mediated mEPSC, [13] and kalirin7 knockout animals show strong deficits in NMDAr mediated long term potentiation [13] [40] and long term depression. [39] This may be linked to the ability to regulate RAC-PAK signaling and actin dynamics, which in turn can regulate the size and density of dendritic spines. [13] Within dendritic spines, kalirin interacts with multiple disease related proteins to regulate synapse strength. It directly interacts with the schizophrenia risk factor DISC1 that can act to suppress kalirin function within the spine. [43] Furthermore, kalirin7 directly interacts with the GluN2B subunit of the NMDA receptor [40] and PSD95 [44] within the post-synaptic density.

The importance of KALRN in neurodevelopment is supported by knockout animal models that display profound deficiencies in multiple behavioral tasks. Kalirin knockout animals display reduced GEF activity, [13] dendritic arborization and spine density. [45] These deficits may be linked to the observed reductions in prepulse inhibition, sociability and increased locomotor activity. Notably, loss of kalirin results in deficits in working memory, but not reference memory. [13] [46] The generation of a kalirin7 specific knockout animal model revealed similar deficits in spine density, [46] [47] suggesting a central role of kalirin7 in regulating neuronal connectivity. Both full and kalirin7 specific knockout animals show decreased anxiety-like behavior and impaired contextual fear learning. [47] [48] [10]

Expression

Multiple isoforms, arising from alternate splicing and promoter usage, display varying tissue and developmental expression. [49] [50] Control over kalirin expression is exerted through the use of alternate promoters which produce alternate start sites and restrict expression to specific neuronal subpopulations, and alter kalirin activity within neurons. [51] [52] During early development, the long kalirin9 and 12 isoforms are predominant in the brain. These isoforms contain both a Rac and a RhoA selective GEF domain, and control axonal growth and dendritic branching. Kalirin9 and 12 are also expressed ubiquitously throughout the body [53] and have functions outwith the brain. However, during neurodevelopment the kalirin7 isoform is preferentially expressed during periods of synaptogenesis, and this isoform displays highly restricted cortical expression. [53] [54] Kalirin7 expresses only the N-terminal domains, including the Rac-GEF domain along with a c-terminal PDZ-binding domain that strongly targets kalirin7 to the post-synaptic density. [44] It is likely this subcellular distribution is vital to kalirin7 function, as this isoform exerts control dendritic spine density and synaptic plasticity. It is likely that mutations that result in deregulation of kalirin function within the brain is responsible for the role of kalirin within multiple neurological disorders.

Notes

Related Research Articles

<span class="mw-page-title-main">Dendritic spine</span> Small protrusion on a dendrite that receives input from a single axon

A dendritic spine is a small membranous protrusion from a neuron's dendrite that typically receives input from a single axon at the synapse. Dendritic spines serve as a storage site for synaptic strength and help transmit electrical signals to the neuron's cell body. Most spines have a bulbous head, and a thin neck that connects the head of the spine to the shaft of the dendrite. The dendrites of a single neuron can contain hundreds to thousands of spines. In addition to spines providing an anatomical substrate for memory storage and synaptic transmission, they may also serve to increase the number of possible contacts between neurons. It has also been suggested that changes in the activity of neurons have a positive effect on spine morphology.

In neuroscience, synaptic plasticity is the ability of synapses to strengthen or weaken over time, in response to increases or decreases in their activity. Since memories are postulated to be represented by vastly interconnected neural circuits in the brain, synaptic plasticity is one of the important neurochemical foundations of learning and memory.

<span class="mw-page-title-main">Brain-derived neurotrophic factor</span> Protein found in humans

Brain-derived neurotrophic factor (BDNF), or abrineurin, is a protein that, in humans, is encoded by the BDNF gene. BDNF is a member of the neurotrophin family of growth factors, which are related to the canonical nerve growth factor (NGF), a family which also includes NT-3 and NT-4/NT-5. Neurotrophic factors are found in the brain and the periphery. BDNF was first isolated from a pig brain in 1982 by Yves-Alain Barde and Hans Thoenen.

Immediate early genes (IEGs) are genes which are activated transiently and rapidly in response to a wide variety of cellular stimuli. They represent a standing response mechanism that is activated at the transcription level in the first round of response to stimuli, before any new proteins are synthesized. IEGs are distinct from "late response" genes, which can only be activated later, following the synthesis of early response gene products. Thus IEGs have been called the "gateway to the genomic response". The term can describe viral regulatory proteins that are synthesized following viral infection of a host cell, or cellular proteins that are made immediately following stimulation of a resting cell by extracellular signals.

Synaptogenesis is the formation of synapses between neurons in the nervous system. Although it occurs throughout a healthy person's lifespan, an explosion of synapse formation occurs during early brain development, known as exuberant synaptogenesis. Synaptogenesis is particularly important during an individual's critical period, during which there is a certain degree of synaptic pruning due to competition for neural growth factors by neurons and synapses. Processes that are not used, or inhibited during their critical period will fail to develop normally later on in life.

<span class="mw-page-title-main">Tau protein</span> Group of six protein isoforms produced from the MAPT gene

The tau proteins are a group of six highly soluble protein isoforms produced by alternative splicing from the gene MAPT. They have roles primarily in maintaining the stability of microtubules in axons and are abundant in the neurons of the central nervous system (CNS), where the cerebral cortex has the highest abundance. They are less common elsewhere but are also expressed at very low levels in CNS astrocytes and oligodendrocytes.

<span class="mw-page-title-main">SNAP25</span> Protein-coding gene in the species Homo sapiens

Synaptosomal-Associated Protein, 25kDa (SNAP-25) is a Target Soluble NSF (N-ethylmaleimide-sensitive factor) Attachment Protein Receptor (t-SNARE) protein encoded by the SNAP25 gene found on chromosome 20p12.2 in humans. SNAP-25 is a component of the trans-SNARE complex, which accounts for membrane fusion specificity and directly executes fusion by forming a tight complex that brings the synaptic vesicle and plasma membranes together.

<span class="mw-page-title-main">Protocadherin</span>

Protocadherins (Pcdhs) are the largest mammalian subgroup of the cadherin superfamily of homophilic cell-adhesion proteins. They were discovered by Shintaro Suzuki's group, when they used PCR to find new members of the cadherin family. The PCR fragments that corresponded to protocadherins were found in vertebrate and invertebrate species. This prevalence in a wide range of species suggested that the fragments were part of an ancient cadherin and were thus termed "Protocadherins" as the "first cadherins". Of the approximately 70 Pcdh genes identified in mammalian genomes, over 50 are located in tightly linked gene clusters on the same chromosome. Until recently, it was assumed that this kind of organization can only be found in vertebrates, but Octopus bimaculoides has 168 genes of which nearly three-quarters are found in tandem clusters with the two largest clusters compromising 31 and 17 genes, respectively.

<span class="mw-page-title-main">Neuregulin 1</span> Protein-coding gene in the species Homo sapiens

Neuregulin 1, or NRG1, is a gene of the epidermal growth factor family that in humans is encoded by the NRG1 gene. NRG1 is one of four proteins in the neuregulin family that act on the EGFR family of receptors. Neuregulin 1 is produced in numerous isoforms by alternative splicing, which allows it to perform a wide variety of functions. It is essential for the normal development of the nervous system and the heart.

<span class="mw-page-title-main">CX614</span> Chemical compound

CX-614 is an ampakine drug developed by Cortex Pharmaceuticals. It has been investigated for its effect on AMPA receptors.

<span class="mw-page-title-main">HOMER1</span> Protein and coding gene in humans

Homer protein homolog 1 or Homer1 is a neuronal protein that in humans is encoded by the HOMER1 gene. Other names are Vesl and PSD-Zip45.

<span class="mw-page-title-main">SYNGAP1</span> Protein in Homo sapiens

Synaptic Ras GTPase-activating protein 1, also known as synaptic Ras-GAP 1 or SYNGAP1, is a protein that in humans is encoded by the SYNGAP1 gene. SYNGAP1 is a ras GTPase-activating protein that is critical for the development of cognition and proper synapse function. Mutations in humans can cause intellectual disability, epilepsy, autism and sensory processing deficits.

<span class="mw-page-title-main">PPP1R9B</span> Protein found in humans

Neurabin-2 is a protein that in humans is encoded by the PPP1R9B gene.

<span class="mw-page-title-main">Brain mitochondrial carrier protein 1</span> Protein-coding gene in the species Homo sapiens

Brain mitochondrial carrier protein 1 is a protein that in humans is encoded by the SLC25A14 gene.

<span class="mw-page-title-main">SYNGR1</span> Protein-coding gene in the species Homo sapiens

Synaptogyrin-1 is a protein that in humans is encoded by the SYNGR1 gene.

<span class="mw-page-title-main">Dedicator of cytokinesis protein 4</span> Protein found in humans

Dedicator of cytokinesis protein 4 (Dock4), is a large protein encoded in the human by the DOCK4 gene, involved in intracellular signalling networks. It is a member of the DOCK-B subfamily of the DOCK family of guanine nucleotide exchange factors (GEFs) which function as activators of small G-proteins. Dock4 activates the small G proteins Rac and Rap1.

Activity-dependent plasticity is a form of functional and structural neuroplasticity that arises from the use of cognitive functions and personal experience; hence, it is the biological basis for learning and the formation of new memories. Activity-dependent plasticity is a form of neuroplasticity that arises from intrinsic or endogenous activity, as opposed to forms of neuroplasticity that arise from extrinsic or exogenous factors, such as electrical brain stimulation- or drug-induced neuroplasticity. The brain's ability to remodel itself forms the basis of the brain's capacity to retain memories, improve motor function, and enhance comprehension and speech amongst other things. It is this trait to retain and form memories that is associated with neural plasticity and therefore many of the functions individuals perform on a daily basis. This plasticity occurs as a result of changes in gene expression which are triggered by signaling cascades that are activated by various signaling molecules during increased neuronal activity.

Long-term potentiation (LTP), thought to be the cellular basis for learning and memory, involves a specific signal transmission process that underlies synaptic plasticity. Among the many mechanisms responsible for the maintenance of synaptic plasticity is the cadherin–catenin complex. By forming complexes with intracellular catenin proteins, neural cadherins (N-cadherins) serve as a link between synaptic activity and synaptic plasticity, and play important roles in the processes of learning and memory.

Memory allocation is a process that determines which specific synapses and neurons in a neural network will store a given memory. Although multiple neurons can receive a stimulus, only a subset of the neurons will induce the necessary plasticity for memory encoding. The selection of this subset of neurons is termed neuronal allocation. Similarly, multiple synapses can be activated by a given set of inputs, but specific mechanisms determine which synapses actually go on to encode the memory, and this process is referred to as synaptic allocation. Memory allocation was first discovered in the lateral amygdala by Sheena Josselyn and colleagues in Alcino J. Silva's laboratory.

<span class="mw-page-title-main">Synaptic stabilization</span> Modifying synaptic strength via cell adhesion molecules

Synaptic stabilization is crucial in the developing and adult nervous systems and is considered a result of the late phase of long-term potentiation (LTP). The mechanism involves strengthening and maintaining active synapses through increased expression of cytoskeletal and extracellular matrix elements and postsynaptic scaffold proteins, while pruning less active ones. For example, cell adhesion molecules (CAMs) play a large role in synaptic maintenance and stabilization. Gerald Edelman discovered CAMs and studied their function during development, which showed CAMs are required for cell migration and the formation of the entire nervous system. In the adult nervous system, CAMs play an integral role in synaptic plasticity relating to learning and memory.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000160145 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000061751 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. 1 2 Colomer V, Engelender S, Sharp AH, Duan K, Cooper JK, Lanahan A, et al. (September 1997). "Huntingtin-associated protein 1 (HAP1) binds to a Trio-like polypeptide, with a rac1 guanine nucleotide exchange factor domain". Human Molecular Genetics. 6 (9): 1519–25. doi: 10.1093/hmg/6.9.1519 . PMID   9285789.
  6. Kawai T, Sanjo H, Akira S (February 1999). "Duet is a novel serine/threonine kinase with Dbl-Homology (DH) and Pleckstrin-Homology (PH) domains". Gene. 227 (2): 249–55. doi:10.1016/S0378-1119(98)00605-2. PMID   10023074.
  7. Chakrabarti K, Lin R, Schiller NI, Wang Y, Koubi D, Fan YX, et al. (June 2005). "Critical role for Kalirin in nerve growth factor signaling through TrkA". Molecular and Cellular Biology. 25 (12): 5106–18. doi:10.1128/MCB.25.12.5106-5118.2005. PMC   1140581 . PMID   15923627.
  8. McPherson CE, Eipper BA, Mains RE (February 2002). "Genomic organization and differential expression of Kalirin isoforms". Gene. 284 (1–2): 41–51. doi:10.1016/S0378-1119(02)00386-4. PMID   11891045.
  9. Xie Z, Srivastava DP, Photowala H, Kai L, Cahill ME, Woolfrey KM, et al. (November 2007). "Kalirin-7 controls activity-dependent structural and functional plasticity of dendritic spines". Neuron. 56 (4): 640–56. doi:10.1016/j.neuron.2007.10.005. PMC   2118058 . PMID   18031682.
  10. 1 2 Ma XM, Kiraly DD, Gaier ED, Wang Y, Kim EJ, Levine ES, et al. (November 2008). "Kalirin-7 is required for synaptic structure and function". The Journal of Neuroscience. 28 (47): 12368–82. doi:10.1523/JNEUROSCI.4269-08.2008. PMC   2586970 . PMID   19020030.
  11. Sommer JE, Budreck EC (April 2009). "Kalirin-7: linking spine plasticity and behavior". The Journal of Neuroscience. 29 (17): 5367–9. doi:10.1523/JNEUROSCI.0235-09.2009. PMC   2684031 . PMID   19403804.
  12. 1 2 Penzes P, Jones KA (August 2008). "Dendritic spine dynamics--a key role for kalirin-7". Trends in Neurosciences. 31 (8): 419–27. doi:10.1016/j.tins.2008.06.001. PMC   3973420 . PMID   18597863.
  13. 1 2 3 4 5 6 7 Cahill ME, Xie Z, Day M, Photowala H, Barbolina MV, Miller CA, et al. (August 2009). "Kalirin regulates cortical spine morphogenesis and disease-related behavioral phenotypes". Proceedings of the National Academy of Sciences of the United States of America. 106 (31): 13058–63. Bibcode:2009PNAS..10613058C. doi: 10.1073/pnas.0904636106 . PMC   2722269 . PMID   19625617.
  14. 1 2 Youn H, Ji I, Ji HP, Markesbery WR, Ji TH (November 2007). "Under-expression of Kalirin-7 Increases iNOS activity in cultured cells and correlates to elevated iNOS activity in Alzheimer's disease hippocampus". Journal of Alzheimer's Disease. 12 (3): 271–81. doi:10.3233/jad-2007-12309. PMID   18057561.
  15. 1 2 Youn H, Jeoung M, Koo Y, Ji H, Markesbery WR, Ji I, Ji TH (June 2007). "Kalirin is under-expressed in Alzheimer's disease hippocampus". Journal of Alzheimer's Disease. 11 (3): 385–97. doi:10.3233/jad-2007-11314. PMID   17851188.
  16. Lek M, Diab N (2019-07-15). "Faculty Opinions recommendation of Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism". Faculty Opinions. doi: 10.3410/f.734542901.793562583 . S2CID   199641906.
  17. Lek M, Diab N (2019-07-15). "Faculty Opinions recommendation of Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism". Faculty Opinions. doi: 10.3410/f.734542901.793562583 . S2CID   199641906.
  18. Leblond CS, Cliquet F, Carton C, Huguet G, Mathieu A, Kergrohen T, et al. (2018-07-06). "Both rare and common genetic variants contribute to autism in the Faroe Islands". npj Genomic Medicine. 4: 1. bioRxiv   10.1101/363853 . doi:10.1038/s41525-018-0075-2. PMC   6341098 . PMID   30675382. S2CID   196670411.
  19. Makrythanasis P, Guipponi M, Santoni FA, Zaki M, Issa MY, Ansar M, et al. (July 2016). "Exome sequencing discloses KALRN homozygous variant as likely cause of intellectual disability and short stature in a consanguineous pedigree". Human Genomics. 10 (1): 26. doi: 10.1186/s40246-016-0082-2 . PMC   4947303 . PMID   27421267.
  20. "Prevalence and Architecture of De Novo Mutations in Developmental Disorders" (PDF). Obstetrical & Gynecological Survey. 72 (6): 340–341. June 2017. doi:10.1097/ogx.0000000000000460. ISSN   0029-7828. S2CID   79759435.
  21. Peña-Oliver Y, Carvalho FM, Sanchez-Roige S, Quinlan EB, Jia T, Walker-Tilley T, et al. (2016-04-07). "Mouse and Human Genetic Analyses Associate Kalirin with Ventral Striatal Activation during Impulsivity and with Alcohol Misuse". Frontiers in Genetics. 7: 52. doi: 10.3389/fgene.2016.00052 . PMC   4823271 . PMID   27092175.
  22. Kiraly DD, Nemirovsky NE, LaRese TP, Tomek SE, Yahn SL, Olive MF, et al. (October 2013). "Constitutive knockout of kalirin-7 leads to increased rates of cocaine self-administration". Molecular Pharmacology. 84 (4): 582–90. doi:10.1124/mol.113.087106. PMC   3781382 . PMID   23894151.
  23. "Correction: Wang et al., Kalirin-7 Mediates Cocaine-Induced AMPA Receptor and Spine Plasticity, Enabling Incentive Sensitization". Journal of Neuroscience. 34 (2): 688. 2014-01-08. doi: 10.1523/jneurosci.4822-13.2014 . ISSN   0270-6474. PMC   3953587 . S2CID   219222516.
  24. Kiraly DD, Ma XM, Mazzone CM, Xin X, Mains RE, Eipper BA (August 2010). "Behavioral and morphological responses to cocaine require kalirin7". Biological Psychiatry. 68 (3): 249–55. doi:10.1016/j.biopsych.2010.03.024. PMC   2907465 . PMID   20452575.
  25. Burdon KP (2014-03-07). "Faculty Opinions recommendation of A polygenic burden of rare disruptive mutations in schizophrenia". Faculty Opinions. doi: 10.3410/f.718252264.793491785 .
  26. Russell TA, Blizinsky KD, Cobia DJ, Cahill ME, Xie Z, Sweet RA, et al. (September 2014). "A sequence variant in human KALRN impairs protein function and coincides with reduced cortical thickness". Nature Communications. 5 (1): 4858. Bibcode:2014NatCo...5.4858R. doi:10.1038/ncomms5858. PMC   4166532 . PMID   25224588.
  27. Kushima I, Nakamura Y, Aleksic B, Ikeda M, Ito Y, Shiino T, et al. (May 2012). "Resequencing and association analysis of the KALRN and EPHB1 genes and their contribution to schizophrenia susceptibility". Schizophrenia Bulletin. 38 (3): 552–60. doi:10.1093/schbul/sbq118. PMC   3329972 . PMID   21041834.
  28. Narayan S, Tang B, Head SR, Gilmartin TJ, Sutcliffe JG, Dean B, Thomas EA (November 2008). "Molecular profiles of schizophrenia in the CNS at different stages of illness". Brain Research. 1239: 235–48. doi:10.1016/j.brainres.2008.08.023. PMC   2783475 . PMID   18778695.
  29. Hill JJ, Hashimoto T, Lewis DA (June 2006). "Molecular mechanisms contributing to dendritic spine alterations in the prefrontal cortex of subjects with schizophrenia". Molecular Psychiatry. 11 (6): 557–66. doi: 10.1038/sj.mp.4001792 . PMID   16402129. S2CID   614799.
  30. Nairn A, Leslie S (2018-12-19). "Faculty Opinions recommendation of Changes in the Synaptic Proteome in Tauopathy and Rescue of Tau-Induced Synapse Loss by C1q Antibodies". Faculty Opinions. doi: 10.3410/f.734327820.793554446 . S2CID   91384966.
  31. 1 2 Cissé M, Duplan E, Lorivel T, Dunys J, Bauer C, Meckler X, et al. (November 2017). "The transcription factor XBP1s restores hippocampal synaptic plasticity and memory by control of the Kalirin-7 pathway in Alzheimer model". Molecular Psychiatry. 22 (11): 1562–1575. doi:10.1038/mp.2016.152. PMC   5658671 . PMID   27646263.
  32. Xie Z, Shapiro LP, Cahill ME, Russell TA, Lacor PN, Klein WL, Penzes P (May 2019). "Kalirin-7 prevents dendritic spine dysgenesis induced by amyloid beta-derived oligomers". The European Journal of Neuroscience. 49 (9): 1091–1101. doi:10.1111/ejn.14311. PMC   6559832 . PMID   30565792.
  33. 1 2 Penzes P, Johnson RC, Kambampati V, Mains RE, Eipper BA (November 2001). "Distinct roles for the two Rho GDP/GTP exchange factor domains of kalirin in regulation of neurite growth and neuronal morphology". The Journal of Neuroscience. 21 (21): 8426–34. doi:10.1523/jneurosci.21-21-08426.2001. PMC   6762781 . PMID   11606631.
  34. Jones KA, Srivastava DP, Allen JA, Strachan RT, Roth BL, Penzes P (November 2009). "Rapid modulation of spine morphology by the 5-HT2A serotonin receptor through kalirin-7 signaling". Proceedings of the National Academy of Sciences of the United States of America. 106 (46): 19575–80. Bibcode:2009PNAS..10619575J. doi: 10.1073/pnas.0905884106 . PMC   2780750 . PMID   19889983.
  35. Yan Y, Eipper BA, Mains RE (October 2015). "Kalirin-9 and Kalirin-12 Play Essential Roles in Dendritic Outgrowth and Branching". Cerebral Cortex. 25 (10): 3487–501. doi:10.1093/cercor/bhu182. PMC   4585498 . PMID   25146373.
  36. May V, Schiller MR, Eipper BA, Mains RE (August 2002). "Kalirin Dbl-homology guanine nucleotide exchange factor 1 domain initiates new axon outgrowths via RhoG-mediated mechanisms". The Journal of Neuroscience. 22 (16): 6980–90. doi: 10.1523/jneurosci.22-16-06980.2002 . PMC   6757900 . PMID   12177196. S2CID   15927856.
  37. 1 2 Xie Z, Srivastava DP, Photowala H, Kai L, Cahill ME, Woolfrey KM, et al. (November 2007). "Kalirin-7 controls activity-dependent structural and functional plasticity of dendritic spines". Neuron. 56 (4): 640–56. doi:10.1016/j.neuron.2007.10.005. PMC   2118058 . PMID   18031682.
  38. 1 2 3 Herring BE, Nicoll RA (February 2016). "Kalirin and Trio proteins serve critical roles in excitatory synaptic transmission and LTP". Proceedings of the National Academy of Sciences of the United States of America. 113 (8): 2264–9. Bibcode:2016PNAS..113.2264H. doi: 10.1073/pnas.1600179113 . PMC   4776457 . PMID   26858404.
  39. 1 2 Lemtiri-Chlieh F, Zhao L, Kiraly DD, Eipper BA, Mains RE, Levine ES (December 2011). "Kalirin-7 is necessary for normal NMDA receptor-dependent synaptic plasticity". BMC Neuroscience. 12 (1): 126. doi: 10.1186/1471-2202-12-126 . PMC   3261125 . PMID   22182308.
  40. 1 2 3 Kiraly DD, Lemtiri-Chlieh F, Levine ES, Mains RE, Eipper BA (August 2011). "Kalirin binds the NR2B subunit of the NMDA receptor, altering its synaptic localization and function". The Journal of Neuroscience. 31 (35): 12554–65. doi:10.1523/jneurosci.3143-11.2011. PMC   3172699 . PMID   21880917.
  41. LaRese TP, Yan Y, Eipper BA, Mains RE (May 2017). "Using Kalirin conditional knockout mice to distinguish its role in dopamine receptor mediated behaviors". BMC Neuroscience. 18 (1): 45. doi: 10.1186/s12868-017-0363-2 . PMC   5442696 . PMID   28535798.
  42. Paskus JD, Tian C, Fingleton E, Shen C, Chen X, Li Y, et al. (December 2019). "Synaptic Kalirin-7 and Trio Interactomes Reveal a GEF Protein-Dependent Neuroligin-1 Mechanism of Action". Cell Reports. 29 (10): 2944–2952.e5. doi: 10.1016/j.celrep.2019.10.115 . PMC   9012321 . PMID   31801062.
  43. Hayashi-Takagi A, Takaki M, Graziane N, Seshadri S, Murdoch H, Dunlop AJ, et al. (March 2010). "Disrupted-in-Schizophrenia 1 (DISC1) regulates spines of the glutamate synapse via Rac1". Nature Neuroscience. 13 (3): 327–32. doi:10.1038/nn.2487. PMC   2846623 . PMID   20139976.
  44. 1 2 Penzes P, Johnson RC, Sattler R, Zhang X, Huganir RL, Kambampati V, et al. (January 2001). "The neuronal Rho-GEF Kalirin-7 interacts with PDZ domain-containing proteins and regulates dendritic morphogenesis". Neuron. 29 (1): 229–42. doi: 10.1016/s0896-6273(01)00193-3 . PMID   11182094. S2CID   7014018.
  45. Xie Z, Cahill ME, Penzes P (January 2010). "Kalirin loss results in cortical morphological alterations". Molecular and Cellular Neurosciences. 43 (1): 81–9. doi:10.1016/j.mcn.2009.09.006. PMC   2818244 . PMID   19800004.
  46. 1 2 Vanleeuwen JE, Penzes P (December 2012). "Long-term perturbation of spine plasticity results in distinct impairments of cognitive function". Journal of Neurochemistry. 123 (5): 781–9. doi:10.1111/j.1471-4159.2012.07899.x. PMC   3493825 . PMID   22862288.
  47. 1 2 Xie Z, Cahill ME, Radulovic J, Wang J, Campbell SL, Miller CA, et al. (January 2011). "Hippocampal phenotypes in kalirin-deficient mice". Molecular and Cellular Neurosciences. 46 (1): 45–54. doi:10.1016/j.mcn.2010.08.005. PMC   3576140 . PMID   20708080.
  48. Mandela P, Yankova M, Conti LH, Ma XM, Grady J, Eipper BA, Mains RE (November 2012). "Kalrn plays key roles within and outside of the nervous system". BMC Neuroscience. 13 (1): 136. doi: 10.1186/1471-2202-13-136 . PMC   3541206 . PMID   23116210.
  49. Johnson RC, Penzes P, Eipper BA, Mains RE (June 2000). "Isoforms of kalirin, a neuronal Dbl family member, generated through use of different 5'- and 3'-ends along with an internal translational initiation site". The Journal of Biological Chemistry. 275 (25): 19324–33. doi: 10.1074/jbc.m000676200 . PMID   10777487. S2CID   2718066.
  50. McPherson CE, Eipper BA, Mains RE (February 2002). "Genomic organization and differential expression of Kalirin isoforms". Gene. 284 (1–2): 41–51. doi:10.1016/s0378-1119(02)00386-4. PMID   11891045.
  51. Mains RE, Kiraly DD, Eipper-Mains JE, Ma XM, Eipper BA (February 2011). "Kalrn promoter usage and isoform expression respond to chronic cocaine exposure". BMC Neuroscience. 12 (1): 20. doi: 10.1186/1471-2202-12-20 . PMC   3048553 . PMID   21329509.
  52. Miller MB, Vishwanatha KS, Mains RE, Eipper BA (May 2015). "An N-terminal Amphipathic Helix Binds Phosphoinositides and Enhances Kalirin Sec14 Domain-mediated Membrane Interactions". The Journal of Biological Chemistry. 290 (21): 13541–55. doi: 10.1074/jbc.m115.636746 . PMC   4505600 . PMID   25861993.
  53. 1 2 Hansel DE, Quiñones ME, Ronnett GV, Eipper BA (July 2001). "Kalirin, a GDP/GTP exchange factor of the Dbl family, is localized to nerve, muscle, and endocrine tissue during embryonic rat development". The Journal of Histochemistry and Cytochemistry. 49 (7): 833–44. doi: 10.1177/002215540104900704 . PMID   11410608. S2CID   14973698.
  54. Mandela P, Ma XM (2012). "Kalirin, a key player in synapse formation, is implicated in human diseases". Neural Plasticity. 2012: 728161. doi: 10.1155/2012/728161 . PMC   3324156 . PMID   22548195.