Network


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

Hotspot


Dive into the research topics where Aleksandr Shcheglovitov is active.

Publication


Featured researches published by Aleksandr Shcheglovitov.


Nature | 2011

MicroRNA-mediated conversion of human fibroblasts to neurons

Andrew S. Yoo; Alfred X. Sun; Li Li; Aleksandr Shcheglovitov; Thomas Portmann; Yulong Li; Chris Lee-Messer; Ricardo E. Dolmetsch; Richard W. Tsien; Gerald R. Crabtree

Neurogenic transcription factors and evolutionarily conserved signalling pathways have been found to be instrumental in the formation of neurons. However, the instructive role of microRNAs (miRNAs) in neurogenesis remains unexplored. We recently discovered that miR-9* and miR-124 instruct compositional changes of SWI/SNF-like BAF chromatin-remodelling complexes, a process important for neuronal differentiation and function. Nearing mitotic exit of neural progenitors, miR-9* and miR-124 repress the BAF53a subunit of the neural-progenitor (np)BAF chromatin-remodelling complex. After mitotic exit, BAF53a is replaced by BAF53b, and BAF45a by BAF45b and BAF45c, which are then incorporated into neuron-specific (n)BAF complexes essential for post-mitotic functions. Because miR-9/9* and miR-124 also control multiple genes regulating neuronal differentiation and function, we proposed that these miRNAs might contribute to neuronal fates. Here we show that expression of miR-9/9* and miR-124 (miR-9/9*-124) in human fibroblasts induces their conversion into neurons, a process facilitated by NEUROD2. Further addition of neurogenic transcription factors ASCL1 and MYT1L enhances the rate of conversion and the maturation of the converted neurons, whereas expression of these transcription factors alone without miR-9/9*-124 was ineffective. These studies indicate that the genetic circuitry involving miR-9/9*-124 can have an instructive role in neural fate determination.


Cell Stem Cell | 2011

LRRK2 Mutant iPSC-Derived DA Neurons Demonstrate Increased Susceptibility to Oxidative Stress

Ha Nam Nguyen; Blake Byers; Branden Cord; Aleksandr Shcheglovitov; James A. Byrne; Prachi Gujar; Kehkooi Kee; Birgitt Schüle; Ricardo E. Dolmetsch; William Langston; Theo D. Palmer; Renee A. Reijo Pera

Studies of Parkinsons disease (PD) have been hindered by lack of access to affected human dopaminergic (DA) neurons. Here, we report generation of induced pluripotent stem cells that carry the p.G2019S mutation (G2019S-iPSCs) in the Leucine-Rich Repeat Kinase-2 (LRRK2) gene, the most common PD-related mutation, and their differentiation into DA neurons. The high penetrance of the LRRK2 mutation and its clinical resemblance to sporadic PD suggest that these cells could provide a valuable platform for disease analysis and drug development. We found that DA neurons derived from G2019S-iPSCs showed increased expression of key oxidative stress-response genes and α-synuclein protein. The mutant neurons were also more sensitive to caspase-3 activation and cell death caused by exposure to stress agents, such as hydrogen peroxide, MG-132, and 6-hydroxydopamine, than control DA neurons. This enhanced stress sensitivity is consistent with existing understanding of early PD phenotypes and represents a potential therapeutic target.


Nature Medicine | 2011

Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome

Sergiu P. Paşca; Thomas Portmann; Irina Voineagu; Masayuki Yazawa; Aleksandr Shcheglovitov; Anca M. Pasca; Branden Cord; Theo D. Palmer; Sachiko Chikahisa; Seiji Nishino; Jonathan A. Bernstein; Joachim Hallmayer; Daniel H. Geschwind; Ricardo E. Dolmetsch

Monogenic neurodevelopmental disorders provide key insights into the pathogenesis of disease and help us understand how specific genes control the development of the human brain. Timothy syndrome is caused by a missense mutation in the L-type calcium channel Cav1.2 that is associated with developmental delay and autism. We generated cortical neuronal precursor cells and neurons from induced pluripotent stem cells derived from individuals with Timothy syndrome. Cells from these individuals have defects in calcium (Ca2+) signaling and activity-dependent gene expression. They also show abnormalities in differentiation, including decreased expression of genes that are expressed in lower cortical layers and in callosal projection neurons. In addition, neurons derived from individuals with Timothy syndrome show abnormal expression of tyrosine hydroxylase and increased production of norepinephrine and dopamine. This phenotype can be reversed by treatment with roscovitine, a cyclin-dependent kinase inhibitor and atypical L-type–channel blocker. These findings provide strong evidence that Cav1.2 regulates the differentiation of cortical neurons in humans and offer new insights into the causes of autism in individuals with Timothy syndrome.


Nature | 2013

SHANK3 and IGF1 restore synaptic deficits in neurons from 22q13 deletion syndrome patients

Aleksandr Shcheglovitov; Olesya Shcheglovitova; Masayuki Yazawa; Thomas Portmann; Rui Shu; Vittorio Sebastiano; Anna Krawisz; Wendy Froehlich; Jonathan A. Bernstein; Joachim Hallmayer; Ricardo E. Dolmetsch

Phelan–McDermid syndrome (PMDS) is a complex neurodevelopmental disorder characterized by global developmental delay, severely impaired speech, intellectual disability, and an increased risk of autism spectrum disorders (ASDs). PMDS is caused by heterozygous deletions of chromosome 22q13.3. Among the genes in the deleted region is SHANK3, which encodes a protein in the postsynaptic density (PSD). Rare mutations in SHANK3 have been associated with idiopathic ASDs, non-syndromic intellectual disability, and schizophrenia. Although SHANK3 is considered to be the most likely candidate gene for the neurological abnormalities in PMDS patients, the cellular and molecular phenotypes associated with this syndrome in human neurons are unknown. We generated induced pluripotent stem (iPS) cells from individuals with PMDS and autism and used them to produce functional neurons. We show that PMDS neurons have reduced SHANK3 expression and major defects in excitatory, but not inhibitory, synaptic transmission. Excitatory synaptic transmission in PMDS neurons can be corrected by restoring SHANK3 expression or by treating neurons with insulin-like growth factor 1 (IGF1). IGF1 treatment promotes formation of mature excitatory synapses that lack SHANK3 but contain PSD95 and N-methyl-d-aspartate (NMDA) receptors with fast deactivation kinetics. Our findings provide direct evidence for a disruption in the ratio of cellular excitation and inhibition in PMDS neurons, and point to a molecular pathway that can be recruited to restore it.


Science | 2010

The CRAC Channel Activator STIM1 Binds and Inhibits L-Type Voltage-Gated Calcium Channels

Chan Young Park; Aleksandr Shcheglovitov; Ricardo E. Dolmetsch

Channel STIMulation The STIM1 protein functions as a calcium sensor and regulates entry of calcium into cells across the plasma membrane. When cell surface receptors are stimulated and cause release of calcium from internal stores in the endoplasmic reticulum (ER), STIM proteins in the ER membrane interact with the Orai channel pore protein in the plasma membrane to allow calcium entry from the outside of the cell (see the Perspective by Cahalan). Park et al. (p. 101) and Wang et al. (p. 105) now show that STIM also acts to suppress conductance by another calcium channel—the voltage-operated CaV1.2 channel. STIM1 appeared to interact directly with CaV1.2 channels in multiple cell types, including vascular smooth muscle cells, neurons, and cultured cells derived from T lymphocytes. The interaction inhibited opening of the CaV1.2 channels and caused depletion of the channel from the cell surface. The sensor protein that monitors depletion of intracellular calcium regulates two classes of calcium entry channels. Voltage- and store-operated calcium (Ca2+) channels are the major routes of Ca2+ entry in mammalian cells, but little is known about how cells coordinate the activity of these channels to generate coherent calcium signals. We found that STIM1 (stromal interaction molecule 1), the main activator of store-operated Ca2+ channels, directly suppresses depolarization-induced opening of the voltage-gated Ca2+ channel CaV1.2. STIM1 binds to the C terminus of CaV1.2 through its Ca2+ release–activated Ca2+ activation domain, acutely inhibits gating, and causes long-term internalization of the channel from the membrane. This establishes a previously unknown function for STIM1 and provides a molecular mechanism to explain the reciprocal regulation of these two channels in cells.


Nature Neuroscience | 2013

Timothy syndrome is associated with activity-dependent dendritic retraction in rodent and human neurons

Jocelyn F. Krey; Sergiu P. Paşca; Aleksandr Shcheglovitov; Masayuki Yazawa; Rachel Schwemberger; Randall Rasmusson; Ricardo E. Dolmetsch

L-type voltage gated calcium channels have an important role in neuronal development by promoting dendritic growth and arborization. A point mutation in the gene encoding CaV1.2 causes Timothy syndrome, a neurodevelopmental disorder associated with autism spectrum disorders (ASDs). We report that channels with the Timothy syndrome alteration cause activity-dependent dendrite retraction in rat and mouse neurons and in induced pluripotent stem cell (iPSC)-derived neurons from individuals with Timothy syndrome. Dendrite retraction was independent of calcium permeation through the mutant channel, was associated with ectopic activation of RhoA and was inhibited by overexpression of the channel-associated GTPase Gem. These results suggest that CaV1.2 can activate RhoA signaling independently of Ca2+ and provide insights into the cellular basis of Timothy syndrome and other ASDs.


The Journal of Physiology | 2014

Mechanisms by which a CACNA1H mutation in epilepsy patients increases seizure susceptibility

Veit-Simon Eckle; Aleksandr Shcheglovitov; Iuliia Vitko; Deblina Dey; Chan Choo Yap; Bettina Winckler; Edward Perez-Reyes

Mutations in the Cav3.2 T‐type Ca2+ channel were found in patients with idiopathic generalized epilepsies, yet the mechanisms by which these mutations increase neuronal excitability and susceptibility to seizures remain to be determined. Using electrophysiological and transfection methods, we validate in cultured hippocampal neurons the hypothesis that an epilepsy mutation increases neuronal excitability. Mutations in the I–II loop of the channel increase trafficking to the plasma membrane without altering trafficking into dendrites. Mutations also enhance dendritic arborization. Additionally, we provide the first evidence that Cav3.2 can signal to Ca2+‐regulated transcription factors, which are known to play important roles in neuronal development and gene expression.


The Journal of General Physiology | 2012

Molecular and biophysical basis of glutamate and trace metal modulation of voltage-gated Cav2.3 calcium channels

Aleksandr Shcheglovitov; Iuliia Vitko; Roman M. Lazarenko; Peihan Orestes; Slobodan M. Todorovic; Edward Perez-Reyes

Here, we describe a new mechanism by which glutamate (Glu) and trace metals reciprocally modulate activity of the Cav2.3 channel by profoundly shifting its voltage-dependent gating. We show that zinc and copper, at physiologically relevant concentrations, occupy an extracellular binding site on the surface of Cav2.3 and hold the threshold for activation of these channels in a depolarized voltage range. Abolishing this binding by chelation or the substitution of key amino acid residues in IS1–IS2 (H111) and IS2–IS3 (H179 and H183) loops potentiates Cav2.3 by shifting the voltage dependence of activation toward more negative membrane potentials. We demonstrate that copper regulates the voltage dependence of Cav2.3 by affecting gating charge movements. Thus, in the presence of copper, gating charges transition into the “ON” position slower, delaying activation and reducing the voltage sensitivity of the channel. Overall, our results suggest a new mechanism by which Glu and trace metals transiently modulate voltage-dependent gating of Cav2.3, potentially affecting synaptic transmission and plasticity in the brain.


NeuroImage | 2015

Direct in vivo assessment of human stem cell graft–host neural circuits

Blake Byers; Hyunjoo J. Lee; Jia Liu; Andrew J. Weitz; Peter Lin; Pengbo Zhang; Aleksandr Shcheglovitov; Ricardo E. Dolmetsch; Renee A. Reijo Pera; Jin Hyung Lee

Despite the potential of stem cell-derived neural transplants for treating intractable neurological diseases, the global effects of a transplants electrical activity on host circuitry have never been measured directly, preventing the systematic optimization of such therapies. Here, we overcome this problem by combining optogenetics, stem cell biology, and neuroimaging to directly map stem cell-driven neural circuit formation in vivo. We engineered human induced pluripotent stem cells (iPSCs) to express channelrhodopsin-2 and transplanted resulting neurons to striatum of rats. To non-invasively visualize the function of newly formed circuits, we performed high-field functional magnetic resonance imaging (fMRI) during selective stimulation of transplanted cells. fMRI successfully detected local and remote neural activity, enabling the global graft-host neural circuit function to be assessed. These results demonstrate the potential of a novel neuroimaging-based platform that can be used to identify how a grafts electrical activity influences the brain network in vivo.


Biochimica et Biophysica Acta | 2007

Selectivity signatures of three isoforms of recombinant T-type Ca2+ channels

Aleksandr Shcheglovitov; P. G. Kostyuk; Yaroslav Shuba

Collaboration


Dive into the Aleksandr Shcheglovitov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge