Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Anton Maximov is active.

Publication


Featured researches published by Anton Maximov.


Cell | 2006

A Complexin/Synaptotagmin 1 Switch Controls Fast Synaptic Vesicle Exocytosis

Jiong Tang; Anton Maximov; Ok Ho Shin; Han Dai; Josep Rizo; Thomas C. Südhof

Ca(2+) binding to synaptotagmin 1 triggers fast exocytosis of synaptic vesicles that have been primed for release by SNARE-complex assembly. Besides synaptotagmin 1, fast Ca(2+)-triggered exocytosis requires complexins. Synaptotagmin 1 and complexins both bind to assembled SNARE complexes, but it is unclear how their functions are coupled. Here we propose that complexin binding activates SNARE complexes into a metastable state and that Ca(2+) binding to synaptotagmin 1 triggers fast exocytosis by displacing complexin from metastable SNARE complexes. Specifically, we demonstrate that, biochemically, synaptotagmin 1 competes with complexin for SNARE-complex binding, thereby dislodging complexin from SNARE complexes in a Ca(2+)-dependent manner. Physiologically, increasing the local concentration of complexin selectively impairs fast Ca(2+)-triggered exocytosis but retains other forms of SNARE-dependent fusion. The hypothesis that Ca(2+)-induced displacement of complexins from SNARE complexes triggers fast exocytosis accounts for the loss-of-function and gain-of-function phenotypes of complexins and provides a molecular explanation for the high speed and synchronicity of fast Ca(2+)-triggered neurotransmitter release.


Neuron | 2003

Huntingtin and huntingtin-associated protein 1 influence neuronal calcium signaling mediated by inositol-(1,4,5) triphosphate receptor type 1.

Tie-Shan Tang; Huiping Tu; Edmond Chan; Anton Maximov; Zhengnan Wang; Cheryl L. Wellington; Michael R. Hayden; Ilya Bezprozvanny

Huntingtons disease (HD) is caused by polyglutamine expansion (exp) in huntingtin (Htt). The type 1 inositol (1,4,5)-triphosphate receptor (InsP3R1) is an intracellular calcium (Ca2+) release channel that plays an important role in neuronal function. In a yeast two-hybrid screen with the InsP3R1 carboxy terminus, we isolated Htt-associated protein-1A (HAP1A). We show that an InsP3R1-HAP1A-Htt ternary complex is formed in vitro and in vivo. In planar lipid bilayer reconstitution experiments, InsP3R1 activation by InsP3 is sensitized by Httexp, but not by normal Htt. Transfection of full-length Httexp or caspase-resistant Httexp, but not normal Htt, into medium spiny striatal neurons faciliates Ca2+ release in response to threshold concentrations of the selective mGluR1/5 agonist 3,5-DHPG. Our findings identify a novel molecular link between Htt and InsP3R1-mediated neuronal Ca2+ signaling and provide an explanation for the derangement of cytosolic Ca2+ signaling in HD patients and mouse models.


Journal of Biological Chemistry | 1999

Association of Neuronal Calcium Channels with Modular Adaptor Proteins

Anton Maximov; Thomas C. Südhof; Ilya Bezprozvanny

Presynaptic voltage-gated calcium (Ca2+) channels mediate Ca2+ influx into the presynaptic terminal that triggers synaptic vesicle fusion and neurotransmitter release. The immediate proximity of Ca2+channels to the synaptic vesicle release apparatus is critical for rapid and efficient synaptic transmission. In a series of biochemical experiments, we demonstrate a specific association of the cytosolic carboxyl terminus of the N-type Ca2+ channel pore-forming α1B subunit with the modular adaptor proteins Mint1 and CASK. The carboxyl termini of α1B bind to the first PDZ domain of Mint1 (Mint1-1). The proline-rich region present in the carboxyl termini of α1B binds to the SH3 domain of CASK. Mint1-1 is specific for the E/D-X-W-C/S-COOH consensus, which defines a novel class of PDZ domains (class III). The Mint1-1 PDZ domain-binding motif is present only in the “long” carboxyl-terminal splice variants of N-type (α1B) and P/Q-type (α1A) Ca2+ channels, but not in R-type (α1E) or L-type (α1C) Ca2+ channels. Our results directly link presynaptic Ca2+ channels to a macromolecular complex formed by modular adaptor proteins at synaptic junction and advance our understanding of coupling between cell adhesion and synaptic vesicle exocytosis.


Science | 2009

Complexin Controls the Force Transfer from SNARE Complexes to Membranes in Fusion

Anton Maximov; Jiong Tang; Xiaofei Yang; Zhiping P. Pang; Thomas C. Südhof

Trans–SNAP receptor (SNARE, where SNAP is defined as soluble NSF attachment protein, and NSF is defined as N-ethylmaleimide–sensitive factor) complexes catalyze synaptic vesicle fusion and bind complexin, but the function of complexin binding to SNARE complexes remains unclear. Here we show that in neuronal synapses, complexin simultaneously suppressed spontaneous fusion and activated fast calcium ion–evoked fusion. The dual function of complexin required SNARE binding and also involved distinct amino-terminal sequences of complexin that localize to the point where trans-SNARE complexes insert into the fusing membranes, suggesting that complexin controls the force that trans-SNARE complexes apply onto the fusing membranes. Consistent with this hypothesis, a mutation in the membrane insertion sequence of the v-SNARE synaptobrevin/vesicle-associated membrane protein (VAMP) phenocopied the complexin loss-of-function state without impairing complexin binding to SNARE complexes. Thus, complexin probably activates and clamps the force transfer from assembled trans-SNARE complexes onto fusing membranes.


Neuron | 2013

The CAMKK2-AMPK Kinase Pathway Mediates the Synaptotoxic Effects of Aβ Oligomers through Tau Phosphorylation

Georges Mairet-Coello; Julien Courchet; Simon Pieraut; Virginie Courchet; Anton Maximov; Franck Polleux

Amyloid-β 1-42 (Aβ42) oligomers are synaptotoxic for excitatory cortical and hippocampal neurons and might play a role in early stages of Alzheimers disease (AD) progression. Recent results suggested that Aβ42 oligomers trigger activation of AMP-activated kinase (AMPK), and its activation is increased in the brain of patients with AD. We show that increased intracellular calcium [Ca²⁺](i) induced by NMDA receptor activation or membrane depolarization activates AMPK in a CAMKK2-dependent manner. CAMKK2 or AMPK overactivation is sufficient to induce dendritic spine loss. Conversely, inhibiting their activity protects hippocampal neurons against synaptotoxic effects of Aβ42 oligomers in vitro and against the loss of dendritic spines observed in the human APP(SWE,IND)-expressing transgenic mouse model in vivo. AMPK phosphorylates Tau on KxGS motif S262, and expression of Tau S262A inhibits the synaptotoxic effects of Aβ42 oligomers. Our results identify a CAMKK2-AMPK-Tau pathway as a critical mediator of the synaptotoxic effects of Aβ42 oligomers.


The Journal of Neuroscience | 2002

Synaptic Targeting of N-Type Calcium Channels in Hippocampal Neurons

Anton Maximov; Ilya Bezprozvanny

N-type calcium (Ca2+) channels play a critical role in synaptic function, but the mechanisms responsible for their targeting in neurons are poorly understood. N-type channels are formed by an α1B (CaV2.2) pore-forming subunit associated with β and α2δ auxiliary subunits. By expressing epitope-tagged recombinant α1B subunits in rat hippocampal neuronal cultures, we demonstrate here that synaptic targeting of N-type channels depends on neuronal contacts and synapse formation. We also establish that the C-terminal 163 aa (2177–2339) of the α1B-1 (CaV2.2a) splice variant contain sequences that are both necessary and sufficient for synaptic targeting. By site-directed mutagenesis, we demonstrate that postsynaptic density-95/discs large/zona occludens-1 and Src homology 3 domain-binding motifs located within this region of the α1B subunit (Maximov et al., 1999) act as synergistic synaptic targeting signals. We also show that the recombinant modular adaptor proteins Mint1 and CASK colocalize with N-type channels in synapses. We found that the α1B-2(CaV2.2b) splice variant is restricted to soma and dendrites and postulated that somatodendritic and axonal/presynaptic isoforms of N-type channels are generated via alternative splicing of α1B C termini. These data lead us to propose that during synaptogenesis, the α1B-1 (CaV2.2a) splice variant of the N-type Ca2+ channel pore-forming subunit is recruited to presynaptic locations by means of interactions with modular adaptor proteins Mint1 and CASK. Our results provide a novel insight into the molecular mechanisms responsible for targeting of Ca2+ channels and other synaptic proteins in neurons.


The EMBO Journal | 2006

Genetic analysis of synaptotagmin 2 in spontaneous and Ca2+-triggered neurotransmitter release.

Zhiping P. Pang; Jianyuan Sun; Josep Rizo; Anton Maximov; Thomas C. Südhof

Synaptotagmin 2 resembles synaptotagmin 1, the Ca2+ sensor for fast neurotransmitter release in forebrain synapses, but little is known about synaptotagmin 2 function. Here, we describe a severely ataxic mouse strain that harbors a single, destabilizing amino‐acid substitution (I377N) in synaptotagmin 2. In Calyx of Held synapses, this mutation causes a delay and a decrease in Ca2+‐induced but not in hypertonic sucrose‐induced release, suggesting that synaptotagmin 2 mediates Ca2+ triggering of evoked release in brainstem synapses. Unexpectedly, we additionally observed in synaptotagmin 2 mutant synapses a dramatic increase in spontaneous release. Synaptotagmin 1‐deficient excitatory and inhibitory cortical synapses also displayed a large increase in spontaneous release, demonstrating that this effect was shared among synaptotagmins 1 and 2. Our data suggest that synaptotagmin 1 and 2 perform equivalent functions in the Ca2+ triggering of action potential‐induced release and in the restriction of spontaneous release, consistent with a general role of synaptotagmins in controlling ‘release slots’ for synaptic vesicles at the active zone.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Impaired insulin secretion and glucose intolerance in synaptotagmin-7 null mutant mice

Natalia Gustavsson; Ye Lao; Anton Maximov; Jen Chieh Chuang; Elena Kostromina; Joyce J. Repa; Cai Li; George K. Radda; Thomas C. Südhof; Weiping Han

Vertebrates express at least 15 different synaptotagmins with the same domain structure but diverse localizations and tissue distributions. Synaptotagmin-1,-2, and -9 act as calcium sensors for the fast phrase of neurotransmitter release, and synaptotagmin-12 acts as a calcium-independent modulator of release. The exact functions of the remaining 11 synaptotagmins, however, have not been established. By analogy to the role of synaptotagmin-1, -2, and -9 in neurotransmission, these other synaptotagmins may serve as Ca2+ transducers regulating other Ca2+-dependent membrane processes, such as insulin secretion in pancreatic β-cells. Of these other synaptotagmins, synaptotagmin-7 is one of the most abundant and is present in pancreatic β-cells. To determine whether synaptotagmin-7 regulates Ca2+-dependent insulin secretion, we analyzed synaptotagmin-7 null mutant mice for glucose tolerance and insulin release. Here, we show that synaptotagmin-7 is required for the maintenance of systemic glucose tolerance and glucose-stimulated insulin secretion. Mutant mice have normal insulin sensitivity, insulin production, islet architecture and ultrastructural organization, and metabolic and calcium responses but exhibit impaired glucose-induced insulin secretion, indicating a calcium-sensing defect during insulin-containing secretory granule exocytosis. Taken together, our findings show that synaptotagmin-7 functions as a positive regulator of insulin secretion and may serve as a calcium sensor controlling insulin secretion in pancreatic β cells.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Synaptotagmin-1 and -7 are functionally overlapping Ca2+ sensors for exocytosis in adrenal chromaffin cells

Jean-Sébastien Schonn; Anton Maximov; Ye Lao; Thomas C. Südhof; Jakob B. Sørensen

Synaptotagmin-1, the canonical isoform of the synaptotagmin family, is a Ca2+ sensor for fast synchronous neurotransmitter release in forebrain neurons and chromaffin cells. Even though deletion of synaptotagmin-1 abolishes fast exocytosis in chromaffin cells, it reduces overall secretion by only 20% because of the persistence of slow exocytosis. Therefore, another Ca2+ sensor dominates release in these cells. Synaptotagmin-7 has a higher Ca2+ affinity and slower binding kinetics than synaptotagmin-1, matching the proposed properties for the second, slower Ca2+ sensor. Here, we examined Ca2+-triggered exocytosis in chromaffin cells from KO mice lacking synaptotagmin-7, and from knockin mice containing normal levels of a mutant synaptotagmin-7 whose C2B domain does not bind Ca2+. In both types of mutant chromaffin cells, Ca2+-triggered exocytosis was decreased dramatically. Moreover, in chromaffin cells lacking both synaptotagmin-1 and -7, only a very slow release component, accounting for ≈30% of WT exocytosis, persisted. These data establish synaptotagmin-7 as a major Ca2+ sensor for exocytosis in chromaffin cells, which, together with synaptotagmin-1, mediates almost all of the Ca2+ triggering of exocytosis in these cells, a surprising result, considering the lack of a role of synaptotagmin-7 in synaptic vesicle exocytosis.


Cell | 2012

HDAC4 governs a transcriptional program essential for synaptic plasticity and memory.

Richard Sando; Natalia Gounko; Simon Pieraut; Lujian Liao; John R. Yates; Anton Maximov

Neuronal activity influences genes involved in circuit development and information processing. However, the molecular basis of this process remains poorly understood. We found that HDAC4, a histone deacetylase that shuttles between the nucleus and cytoplasm, controls a transcriptional program essential for synaptic plasticity and memory. The nuclear import of HDAC4 and its association with chromatin is negatively regulated by NMDA receptors. In the nucleus, HDAC4 represses genes encoding constituents of central synapses, thereby affecting synaptic architecture and strength. Furthermore, we show that a truncated form of HDAC4 encoded by an allele associated with mental retardation is a gain-of-function nuclear repressor that abolishes transcription and synaptic transmission despite the loss of the deacetylase domain. Accordingly, mice carrying a mutant that mimics this allele exhibit deficits in neurotransmission, spatial learning, and memory. These studies elucidate a mechanism of experience-dependent plasticity and define the biological role of HDAC4 in the brain.

Collaboration


Dive into the Anton Maximov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simon Pieraut

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Richard Sando

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Josep Rizo

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Julien Courchet

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ye Lao

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jiong Tang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ok Ho Shin

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge