Network


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

Hotspot


Dive into the research topics where David Krizaj is active.

Publication


Featured researches published by David Krizaj.


The Journal of Physiology | 2008

Depletion of calcium stores regulates calcium influx and signal transmission in rod photoreceptors

T. Szikra; Karen Cusato; Wallace B. Thoreson; Peter Barabas; Theodore M. Bartoletti; David Krizaj

Tonic synapses are specialized for sustained calcium entry and transmitter release, allowing them to operate in a graded fashion over a wide dynamic range. We identified a novel plasma membrane calcium entry mechanism that extends the range of rod photoreceptor signalling into light‐adapted conditions. The mechanism, which shares molecular and physiological characteristics with store‐operated calcium entry (SOCE), is required to maintain baseline [Ca2+]i in rod inner segments and synaptic terminals. Sustained Ca2+ entry into rod cytosol is augmented by store depletion, blocked by La3+ and Gd3+ and suppressed by organic antagonists MRS‐1845 and SKF‐96365. Store depletion and the subsequent Ca2+ influx directly stimulated exocytosis in terminals of light‐adapted rods loaded with the activity‐dependent dye FM1–43. Moreover, SOCE blockers suppressed rod‐mediated synaptic inputs to horizontal cells without affecting presynaptic voltage‐operated Ca2+ entry. Silencing of TRPC1 expression with small interference RNA disrupted SOCE in rods, but had no effect on cone Ca2+ signalling. Rods were immunopositive for TRPC1 whereas cone inner segments immunostained with TRPC6 channel antibodies. Thus, SOCE modulates Ca2+ homeostasis and light‐evoked neurotransmission at the rod photoreceptor synapse mediated by TRPC1.


PLOS ONE | 2009

Calcium Homeostasis and Cone Signaling Are Regulated by Interactions between Calcium Stores and Plasma Membrane Ion Channels

T. Szikra; Peter Barabas; Theodore M. Bartoletti; Wei Huang; Abram Akopian; Wallace B. Thoreson; David Krizaj

Calcium is a messenger ion that controls all aspects of cone photoreceptor function, including synaptic release. The dynamic range of the cone output extends beyond the activation threshold for voltage-operated calcium entry, suggesting another calcium influx mechanism operates in cones hyperpolarized by light. We have used optical imaging and whole-cell voltage clamp to measure the contribution of store-operated Ca2+ entry (SOCE) to Ca2+ homeostasis and its role in regulation of neurotransmission at cone synapses. Mn2+ quenching of Fura-2 revealed sustained divalent cation entry in hyperpolarized cones. Ca2+ influx into cone inner segments was potentiated by hyperpolarization, facilitated by depletion of intracellular Ca2+ stores, unaffected by pharmacological manipulation of voltage-operated or cyclic nucleotide-gated Ca2+ channels and suppressed by lanthanides, 2-APB, MRS 1845 and SKF 96365. However, cation influx through store-operated channels crossed the threshold for activation of voltage-operated Ca2+ entry in a subset of cones, indicating that the operating range of inner segment signals is set by interactions between store- and voltage-operated Ca2+ channels. Exposure to MRS 1845 resulted in ∼40% reduction of light-evoked postsynaptic currents in photopic horizontal cells without affecting the light responses or voltage-operated Ca2+ currents in simultaneously recorded cones. The spatial pattern of store-operated calcium entry in cones matched immunolocalization of the store-operated sensor STIM1. These findings show that store-operated channels regulate spatial and temporal properties of Ca2+ homeostasis in vertebrate cones and demonstrate their role in generation of sustained excitatory signals across the first retinal synapse.


The Journal of Physiology | 2010

Glutamate-induced internalization of Cav1.3 L-type Ca2+ channels protects retinal neurons against excitotoxicity

Fengxia Mizuno; Peter Barabas; David Krizaj; Abram Akopian

Glutamate‐induced rise in the intracellular Ca2+ level is thought to be a major cause of excitotoxic cell death, but the mechanisms that control the Ca2+ overload are poorly understood. Using immunocytochemistry, electrophysiology and Ca2+ imaging, we show that activation of ionotropic glutamate receptors induces a selective internalization of Cav1.3 L‐type Ca2+ channels in salamander retinal neurons. The effect of glutamate on Cav1.3 internalization was blocked in Ca2+‐free external solution, or by strong buffering of internal Ca2+ with BAPTA. Downregulation of L‐type Ca2+ channel activity in retinal ganglion cells by glutamate was suppressed by inhibitors of dynamin‐dependent endocytosis. Stabilization of F‐actin by jasplakinolide significantly reduced the ability of glutamate to induce internalization suggesting it is mediated by Ca2+‐dependent reorganization of actin cytoskeleton. We showed that the Cav1.3 is the primary L‐type Ca2+ channel contributing to kainate‐induced excitotoxic death of amacrine and ganglion cells. Block of Cav1.3 internalization by either dynamin inhibition or F‐actin stabilization increased vulnerability of retinal amacrine and ganglion cells to kainate‐induced excitotoxicity. Our data show for the first time that Cav1.3 L‐type Ca2+ channels are subject to rapid glutamate‐induced internalization, which may serve as a negative feedback mechanism protecting retinal neurons against glutamate‐induced excitotoxicity.


Journal of Biological Chemistry | 2017

Endoplasmic reticulum (ER) Ca2+-channel activity contributes to ER stress and cone death in cyclic nucleotide-gated channel deficiency

Michael R. Butler; Hongwei Ma; Fan Yang; Joshua Belcher; Yun-Zheng Le; Katsuhiko Mikoshiba; Martin Biel; Stylianos Michalakis; Anthony Iuso; David Krizaj; Xi-Qin Ding

Endoplasmic reticulum (ER) stress and mislocalization of improperly folded proteins have been shown to contribute to photoreceptor death in models of inherited retinal degenerative diseases. In particular, mice with cone cyclic nucleotide-gated (CNG) channel deficiency, a model for achromatopsia, display both early-onset ER stress and opsin mistrafficking. By 2 weeks of age, these mice show elevated signaling from all three arms of the ER-stress pathway, and by 1 month, cone opsin is improperly distributed away from its normal outer segment location to other retinal layers. This work investigated the role of Ca2+-release channels in ER stress, protein mislocalization, and cone death in a mouse model of CNG-channel deficiency. We examined whether preservation of luminal Ca2+ stores through pharmacological and genetic suppression of ER Ca2+ efflux protects cones by attenuating ER stress. We demonstrated that the inhibition of ER Ca2+-efflux channels reduced all three arms of ER-stress signaling while improving opsin trafficking to cone outer segments and decreasing cone death by 20–35%. Cone-specific gene deletion of the inositol-1,4,5-trisphosphate receptor type I (IP3R1) also significantly increased cone density in the CNG-channel-deficient mice, suggesting that IP3R1 signaling contributes to Ca2+ homeostasis and cone survival. Consistent with the important contribution of organellar Ca2+ signaling in this achromatopsia mouse model, significant differences in dynamic intraorganellar Ca2+ levels were detected in CNG-channel-deficient cones. These results thus identify a novel molecular link between Ca2+ homeostasis and cone degeneration, thereby revealing novel therapeutic targets to preserve cones in inherited retinal degenerative diseases.


The Journal of Neuroscience | 2018

Retinal detachment-induced Müller glial cell swelling activates TRPV4 ion channels and triggers photoreceptor death at body temperature

Hidetaka Matsumoto; Shouta Sugio; François Seghers; David Krizaj; Hideo Akiyama; Yasuki Ishizaki; Philippe Gailly; Koji Shibasaki

Using region-specific injection of hyaluronic acid, we developed a mouse model of acute retinal detachment (RD) to investigate molecular mechanisms of photoreceptor cell death triggered by RD. We focused on the transient receptor potential vanilloid 4 (TRPV4) ion channel, which functions as a thermosensor, osmosensor, and/or mechanosensor. After RD, the number of apoptotic photoreceptors was reduced by ∼50% in TRPV4KO mice relative to wild-type mice, indicating the possible involvement of TRPV4 activation in RD-induced photoreceptor cell death. Furthermore, TRPV4 expressed in Müller glial cells can be activated by mechanical stimuli caused by RD-induced swelling of these cells, resulting in release of the cytokine MCP-1, which is reported as a mediator of Müller glia-derived strong mediator for RD-induced photoreceptor death. We also found that the TRPV4 activation by the Müller glial swelling was potentiated by body temperature. Together, our results suggest that RD adversely impacts photoreceptor viability via TRPV4-dependent cytokine release from Müller glial cells and that TRPV4 is part of a novel molecular pathway that could exacerbate the effects of hypoxia on photoreceptor survival after RD. SIGNIFICANCE STATEMENT Identification of the mechanisms of photoreceptor death in retinal detachment is required for establishment of therapeutic targets for preventing loss of visual acuity. In this study, we found that TRPV4 expressed in Müller glial cells can be activated by mechanical stimuli caused by RD-induced swelling of these cells, resulting in release of the cytokine MCP-1, which is reported as a mediator of Müller glia-derived strong mediator for RD-induced photoreceptor death. We also found that the TRPV4 activation by the Müller glial swelling was potentiated by body temperature. Hence, TRPV4 inhibition could suppress cell death in RD pathological conditions and suggests that TRPV4 in Müller glial cells might be a novel therapeutic target for preventing photoreceptor cell death after RD.


Advances in Experimental Medicine and Biology | 2010

Do calcium channel blockers rescue dying photoreceptors in the Pde6b rd1 mouse

Peter Barabas; Carolee Cutler Peck; David Krizaj


Investigative Ophthalmology & Visual Science | 2014

NONRETROGRADE ENDOCANNABINOID SIGNALING MODULATES RETINAL GANGLION CELL CALCIUM HOMEOSTASIS THROUGH THE TRPV1 CATION CHANNEL

Andrew O. Jo; Daniel A. Ryskamp; Sarah Redmon; Peter Barabas; David Krizaj


Investigative Ophthalmology & Visual Science | 2016

MECHANICAL STRETCH ALTERS THE EXPRESSION PROFILE OF MECHANOSENSITIVE CHANNELS IN MOUSE RETINAL GANGLION CELLS VIA ACTIVATION OF TRPV4

Tam T. T. Phuong; Monika Lakk; David Krizaj


Investigative Ophthalmology & Visual Science | 2015

TRPV4 channels regulate the inflow pathway in the anterior eye

Andrew O. Jo; Daniel A. Ryskamp; Amber M. Frye; Tam T. T. Phuong; Bruce A. Berkowitz; David Krizaj


Investigative Ophthalmology & Visual Science | 2013

Molecular coupling between TRPV4 and aquaporin 4 channels mediates osmosensation in Müller glia

David Krizaj; Daniel A. Ryskamp; Andrew O. Jo; A. S. Verkman; Nanna MacAulay

Collaboration


Dive into the David Krizaj'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

Claudio Punzo

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge