Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2017 Feb 8;93(3):587-605.e7.
doi: 10.1016/j.neuron.2016.12.034. Epub 2017 Jan 26.

Transient Opening of the Mitochondrial Permeability Transition Pore Induces Microdomain Calcium Transients in Astrocyte Processes

Affiliations

Transient Opening of the Mitochondrial Permeability Transition Pore Induces Microdomain Calcium Transients in Astrocyte Processes

Amit Agarwal et al. Neuron. .

Abstract

Astrocytes extend highly branched processes that form functionally isolated microdomains, facilitating local homeostasis by redistributing ions, removing neurotransmitters, and releasing factors to influence blood flow and neuronal activity. Microdomains exhibit spontaneous increases in calcium (Ca2+), but the mechanisms and functional significance of this localized signaling are unknown. By developing conditional, membrane-anchored GCaMP3 mice, we found that microdomain activity that occurs in the absence of inositol triphosphate (IP3)-dependent release from endoplasmic reticulum arises through Ca2+ efflux from mitochondria during brief openings of the mitochondrial permeability transition pore. These microdomain Ca2+ transients were facilitated by the production of reactive oxygen species during oxidative phosphorylation and were enhanced by expression of a mutant form of superoxide dismutase 1 (SOD1 G93A) that causes astrocyte dysfunction and neurodegeneration in amyotrophic lateral sclerosis (ALS). By localizing mitochondria to microdomains, astrocytes ensure local metabolic support for energetically demanding processes and enable coupling between metabolic demand and Ca2+ signaling events.

Keywords: ATP; astrocyte; calcium; cortex; in vivo; metabolism; microdomain; mitochondria; two-photon.

PubMed Disclaimer

Figures

Figure 1
Figure 1. Conditional expression of mGCaMP3 in astrocytes
(A) Gene trap strategy used to insert conditional mGCaMP3 transgene into murine Rosa26 gene. (Top) mGCaMP3 targeting vector which harbors 5′ short (R26-SA) and 3′ long (R26-LA) homology arms from Rosa26, the CMV enhancer chicken β-actin hybrid (CAG) promoter (black box), loxP (blue triangles) flanked neomycin resistance cassette (NeoR, gray box) flanked by two FRT sites (yellow triangles) and 3x SV40-polyA sequence (red hexagon), the mGCaMP3 cDNA (light green box), woodchuck post-transcriptional response element (WPRE, brown box), bovine growth hormone poly A sequence (red box) and negative selection cassette with diphtheria toxin fragment A (DTA, black box). (Bottom) Rosa26 allele with conditional mGCaMP3 cassette after homologous recombination. NeoR cassette was removed by in vivo site-specific recombination. (B) Coronal section of brain from a GLAST-CreER;Rosa26-lsl-mGCaMP3 (GLAST-mGC3) mouse immunostained for mGCaMP3. (C) Coronal hemi-section of brain from a GLAST-CreER;Rosa26-lsl-mGCaMP3;Rosa26-lsl-tdTomato (GLAST-mGC3/tdT) mouse immunostained for tdTomato, mGCaMP3 and GFAP. Inset shows one cortical astrocyte at higher magnification co-expressing mGCaMP3 and GFAP. (D) High magnification images from boxed area in (C). CC: corpus callosum, CgCx: cingulate cortex. (E) Images showing one cortical astrocyte from a GLAST-mGC3/tdT mouse (maximum intensity projected confocal z-stack) immunostained for mGCaMP3 and tdT. (F) Coronal hemi-section of brain from a GFAP-CreER;Rosa26-lsl-mGCaMP3 (GFAP-CreER;mGC3) mouse immunostained for mGCaMP3 and GFAP. (G) Coronal section of brain from a GLAST-mGC3 mouse showing silver intensified immunogold labeling of mGCaMP3. Inset shows one astrocyte at higher magnification. (H, I) High magnification images of silver intensified immunogold labeling of mGCaMP3 in an astrocyte soma (H) and peri-synaptic processes (I) from GLAST-mGC3 mice. NT: nerve terminal, Nu: nucleus, Sp: spine.
Figure 2
Figure 2. Quantification of spontaneous microdomain Ca2+ transients in cortical astrocytes (See also Figure S1, Movie S1, S2 and Data S1)
(A) Image analysis based on CaSCaDe (Ca2+ Signal Classification and Decoding) algorithm. (B) Image of one astrocyte from a GLAST-mGC3 mouse. Median intensity projection (pseudocolored) from 540 frames. (C, D) Maps showing 125 spontaneously active microdomains that occurred in 260 s identified using the CaSCaDe algorithm. (C) All microdomains overlaid on median intensity projected image of astrocyte in (B). (D) Color-coded microdomain map used to uniquely identify each active region. Dashed line indicates cell border. (E) Intensity versus time traces for 10 microdomains (from 125) (colors correspond to locations shown in D), showing characteristics of spontaneous Ca2+ transients in individual microdomains. (F) Intensity versus time traces for 125 spontaneously active microdomains. (G) Raster plot of microdomain activity, color-coded according to fluorescence change (z-score). (H) Histogram illustrating the number of events that occurred per microdomain in five minutes. Data shown as mean ± SEM. 8445 events were analyzed from 114 cells. (I) Spatial map and intensity versus time traces for six adjacent microdomains (area highlighted by red box in D) showing that they can exhibit both uncorrelated and correlated spontaneous activity.
Figure 3
Figure 3. Neurotransmitters activate distinct microdomains in astrocyte processes (See also Figure S2 and Movie S2)
(A) Median intensity projection image (pseudocolored) of 540 frames from one astrocyte from a GLAST-mGC3 mouse (left). Map of spontaneously active microdomains (in TTX (1 μM)) overlaid on image (right). (B) Map of microdomains recorded in control (119 domains, left) and after application of norepinephrine (0.5 μM NE, 152 domains, right). Dashed lines indicate cell border. (C) Intensity versus time traces for five microdomains (corresponding to colors in B), showing characteristics of Ca2+ transients in control and following application of NE. (D) Examples of microdomain Ca2+ transients from traces #2 and #5 (asterisks in C). (E) Raster plots displaying timing and intensity of Ca2+ transients (119 microdomains in control, left, and 152 microdomains in NE, right). (F) Graph of change in temporal correlation of Ca2+ transients among microdomains induced by NE (N = 24 cells, GLAST-mGC3 mice). Data shown as mean ± SEM. *** p < 0.0001 and * p = 0.01, paired two-tailed Student’s t-test. (G – I) Graphs showing mean duration per event (G), frequency of events per domain (H) and spatial co-localization index (I) in control (+ TTX) and NE (+TTX). Data shown as mean ± SEM (n = 18 cells, GLAST-mGC3 mice). *** p < 0.0001, paired two-tailed Student’s t-test. (J) Map showing distribution of active microdomains recorded after application of ATP (blue) and NE (red). Microdomains that exhibited activity in both conditions shown in green. (K – M) Graphs comparing co-localization between microdomains recorded in NE (+ TTX) and ATP (+ TTX) (K), NE (+ TTX) and DHPG (+ TTX) (L), ATP (+ TTX) and DHPG (+ TTX) (M). After randomization, the spatial co-localization of microdomains between two conditions was significantly reduced. Data shown as mean ± SEM. N = 9 cells from GLAST-mGC3 mice. *** p < 0.0004, paired two-tailed Student’s t-test. (N) Graph of number of active domains per cell in control and during two successive applications of ATP (ATP1, ATP2). Data shown as mean ± SEM. N = 15 cells from GLAST-mGC3 mice. *** p < 0.0001 and ** p < 0.001 repeated measure one-way ANOVA analysis with Tukey’s multiple comparisons post hoc test. (O) Graph of spatial co-localization between active microdomains during two ATP applications. All experiments were done in the presence of TTX. After randomization, the spatial co-localization of microdomains between two conditions was significantly reduced. Data shown as mean ± SEM. N = 15 cells from GLAST-mGC3 mice. *** p < 0.0001, paired two-tailed Student’s t-test. (P) Temporal correlation of Ca2+ transients among microdomains recorded during baseline (Control) and in the presence of metabotropic receptor agonists (ATP, DHPG, and NE; all in TTX). Data shown as mean ± SEM. N = 9 cells from GLAST-mGC3 mice. *** p = 0.0002, Repeated measures one-way ANOVA with Tukey’s Multiple Comparison Test.
Figure 4
Figure 4. Spontaneous microdomain Ca2+ transients persist in the absence of neurotransmitter release and ER-dependent Ca2+ release (See also Figure S3 and Movie S3)
(A) Spontaneous EPSCs recorded from cortical pyramidal neurons in control conditions (black trace) and after treatment with veratidine (10 μM) and bafilomycin A1 (4 μM, BafA1, red trace). (B) Histogram of the frequency of spontaneous EPSCs recorded in control conditions and after treatment with veratidine and BafA1. Data shown as mean ± SEM. N = 7 (untreated) N = 5 (BafA1) cells from control mice (GLAST-mGC3 or mGCaMP3/+). **p < 0.009, unpaired two-tailed Student’s t-test. (C) Histogram of the amplitudes of spontaneous EPSCs recorded in control conditions and after treatment with veratidine and BafA1. (D) Image of one astrocyte from a GLAST-mGC3 mouse showing median intensity projection (pseudocolored) from 540 frames (left) in an acute brain slice treated with veratridine and BafA1. (E) Map of 99 spontaneously active microdomains recorded in veratridine and BafA1. (F) Intensity versus time traces of 5 microdomains (corresponding to colors in E) showing characteristics of Ca2+ transients in veratridine and BafA1. (G) Raster plot displaying Ca2+ transients from 99 microdomains in veratridine and BafA1. (H, I) Graphs of number of microdomains per cell (H) and frequency of events per microdomain (I) recorded in control and in veratridine and BafA1. Data shown as mean ± SEM. N = 33 (untreated) and 36 (BafA1) cells from GLAST-mGC3 mice. ns: not significant, two-tailed Student’s t-test. (J) Image of one astrocyte from a GLAST-mGC3;IP3R2−/−mouse showing median intensity projection (pseudocolored) from 260 s (left). Map of spontaneously active microdomains (in TTX (1μM)) overlaid on image (right). (K) Map of all spontaneously active microdomains in 260 s (35 domains). Dashed line indicates cell border. (L) Intensity versus time traces of five microdomains (corresponding to colors in B) showing characteristics of Ca2+ transients. (M) Raster plot displaying Ca2+ transients from active regions in 260 s. (N – Q) Graphs of number of microdomains per cell (N), frequency of events per domain (O), number of Ca2+ transients observed per microdomain (P) and mean amplitude (Q) in GLAST-mGC3 (Control, 94 cells) and GLAST-mGC3;IP3R2−/−(104 cells) mice. Data shown as mean ± SEM. *** p < 0.0001, unpaired two-tailed non-parametric Mann-Whitney test. (R – T) Intensity versus time plots for 5 microdomains (top) and raster plots (bottom) displaying spontaneous Ca2+ transients (in control, black) and NE (10 μM, green) and ATP (100 μM, orange) from GLAST-mGC3;IP3R2−/−mice. All experiments were done in the presence of 1 μM TTX. (U – W) Graphs of number of microdomains per cell (U), frequency of events per domain (V), and mean amplitude (W) of spontaneous Ca2+ transients (in TTX, black) and NE (10 μM, green) and ATP (100 μM, orange) from GLAST-mGC3;IP3R2−/− mice. Data shown as mean ± SEM. N = 12 cells for each condition. ns: not significant, p > 0.05, ** p < 0.01 and * p < 0.01 repeated measure one-way ANOVA analysis with Tukey’s multiple comparisons post hoc test.
Figure 5
Figure 5. Microdomain Ca2+ transients persist in vivo in IP3R2−/−mice (See also Movie S4, S5)
(A) In vivo two photon imaging configuration in which mice were allowed to walk on a linear treadmill. (B) Median intensity projection image (pseudocolored) of one astrocyte in vivo from the primary visual cortex (V1) in a GLAST-mGC3;IP3R2+/+ mouse showing active regions during 260 s (left). Map of spontaneously active microdomains is overlaid on image (right). Dashed red line highlights blood vessel. (C) Image of one astrocyte in vivo from visual cortex (V1) in a GLAST-mGC3;IP3R2−/−mouse, showing median intensity projection (pseudocolored) from 260 s (left). Map of all spontaneously active microdomains (101) that occurred in 286 s is overlaid on image (right). (D) Map of all microdomains (101) that occurred in 286 s in astrocyte shown in (B) (GLAST-mGC3; IP3R2+/+) mouse. (E) Intensity versus time plots for five microdomains (corresponding to colors in D) showing characteristics of Ca2+ transients in a GLAST-mGC3;IP3R2+/+ mouse (top). Raster plot displaying the activity of all microdomains (bottom). Timing of enforced locomotion highlighted by dashed line. (F) Map of all microdomains (51) that occurred in 286 s in astrocyte shown in (C) (GLAST-mGC3;IP3R2−/−) mouse. (G) Intensity versus time plots for five microdomains (corresponding to colors in F) showing characteristics of Ca2+ transients from cell in (F) (top). Raster plot displaying the activity of all microdomains (bottom). Timing of enforced locomotion highlighted by dashed line. (H) Histograms of the average number of active microdomains per cell (left) and number of Ca2+ transients (Events) during 50 s of imaging (right), in IP3R2+/+ (GLAST-mGC3;IP3R2+/+) and IP3R2−/−(GLAST-mGC3;IP3R2−/−) mice during baseline activity. Data shown as mean ± SEM. N = 14 cells from GLAST-mGC3;IP3R2+/+ mice and 25 cells from GLAST-mGC3;IP3R2−/−mice were analyzed. ns: not significant, * p > 0.05, unpaired two-tailed Student’s t-test. (I – J) Graphs showing the number of Ca2+ transients (Events) observed during 50 s of imaging (I) and mean amplitude (z-score) for microdomain Ca2+ transients (J) without stimulation (spontaneous, Spont) and during enforced locomotion on the treadmill (TM) for Control (IP3R2+/+) (left) and IP3R2−/−mice (right). Data shown as mean ± SEM. Note change in scale for the IP3R2−/−mice in (I). N = 13 cells from GLAST-mGC3;IP3R2+/+ mice and 14 cells from GLAST-mGC3;IP3R2−/−mice were analyzed. ns: not significant, p > 0.05, ** p < 0.009, *** p < 0.0001, paired two-tailed Student’s t-test.
Figure 6
Figure 6. Microdomain Ca2+ transients co-localize with mitochondria (See also Figure S4 and Movie S6)
(A) Electron micrograph showing silver enhanced gold immunolabeling of mGCaMP3 in a GLAST-mGC3 mouse. Astrocyte processes are colored red and mitochondria in astrocyte processes are colored green. Yellow arrows highlight excitatory synapses. Image at right shows higher magnification image of area highlighted by boxed area at left. (B) Electron micrographs showing spatial relationship between mitochondria and ER in astrocyte processes located in the vicinity of the excitatory nerve terminals. (C) Histogram showing area within fine astrocyte processes and nerve terminals occupied by mitochondria. Data shown as mean ± SEM. ns: not significant, p > 0.05 unpaired two-tailed Student’s t-test. (D) Images of cortical astrocytes from a GLAST-CreER;Rosa26-lsl-tdTomato;Rosa26-lsl-mito-EGFP (GLAST-tdT;mito-EGFP) mouse labeled with anti-GFAP (GFAP, blue), anti-mCherry (tdT, red) and anti-GFP antibodies. (E) High magnification images of one astrocyte (highlighted by box in E) showing tdT, mGCaMP3 and GFAP localization. (F) Image of one astrocyte in a GLAST-mGC3;mito-EGFP mouse showing median intensity projection (pseudocolored) from 260 s (top). Map of spontaneously active microdomains is overlaid on image (below). Dashed white line highlights cell border. (G) Image of fluorescently tagged mitochondria from cell in (F) (top). Map of spontaneously active microdomains (mD, outlined in black) and mitochondria (Mito, green). (H) Image of fluorescently tagged mitochondria in which z plane has been expanded to visualize mitochondria just above and below plane of imaging from cell in (F) (top). Map of spontaneously active microdomains (mD, outlined in black) and mitochondria (Mito, green). Additional co-localized areas highlighted in red. (I) Intensity versus time plots for Ca2+ transients from five microdomains (colors correspond to locations shown in F, bottom), showing characteristics of spontaneous Ca2+ signals. (J) Plot of co-localization between spontaneously active microdomains and mitochondria and co-localization after randomization of mitochondrial locations within the imaging plane (blue) and after z adjustment (green). Data shown as mean ± SEM. N = 7 cells (single plane) and N = 12 cells (z-plane corrected) from GLAST-mGC;mito-EGFP mice. *** p < 0.0001, paired two-tailed Student’s t-test. (K) Histogram showing the decrease in active microdomains that did not co-localize with mitochondria after treatment with thapsigargin (Tpg, 1 μM, 60 minutes) to deplete ER Ca2+ stores. Data shown as mean ± SEM, N = 12 (untreated) and 11 (Tpg treated) cells. * p < 0.03, unpaired two-tailed Student’s t-test.
Figure 7
Figure 7. Mitochondrial membrane permeability transition pore (mPTP) regulates spontaneous Ca2+ transients (See also Figure S5–S7 and Movie S7, S8)
(A) Schematic showing configuration of membrane permeability transition pore (mPTP, red), mitochondrial Ca2+ uniporter (MCU, yellow) and the electron transport chain (ETC, blue). Pharmacological inhibition of mPTP (mPTP-I; cyclosporin A, 20 μM and rotenone, 10 μM) inhibits mPTP opening and prevents Ca2+ efflux from mitochondrial matrix into the cytosol. (B) Images of astrocytes (median intensity projection of time-series image stack) showing active regions during 286 s in control (left) and after exposure to mPTP-I (right) (GLAST-mGC3;IP3R2−/−mouse). (C) Maps of all spontaneously active microdomains in control (28) and after mPTP inhibition (4). (D) Intensity versus time plots for five microdomains in control and four microdomains in mPTP-I treated slices (colors correspond to locations shown in C). (E) Raster plot displaying Ca2+ transients from all active regions in 286 s in control (top) and mPTP-I (bottom) treated slices. (F) Graphs showing relative frequency (left) and mean amplitude (z-score) of Ca2+ transients in control and mPTP-I treated slices. Data shown as mean ± SEM. N = 18 cells. *** p < 0.0001, ** p < 0.001, unpaired two-tailed Student’s t-test. (G) Schematic showing enhancement of mPTP opening by carboxyatratyloside (CAtr, 20 μM), leading to enhanced Ca2+ efflux from the mitochondrial matrix to the cytosol. (H) Image of an astrocyte (median intensity projection of time-series image stack) showing active regions during 286 s in a GLAST-mGC3;IP3R2−/−mouse. (I) Maps of all spontaneously active microdomains during 260 s in control (19) and after mPTP activation (70) (CAtr treated). (J) Intensity versus time plots of microdomain activity in control (left) and after CAtr treatment (right) (colors correspond to locations shown in I). (K) Raster plots displaying Ca2+ transients in an astrocyte in control (top) and after CAtr treatment (bottom). (L) Graphs showing changes in number of active microdomains per cell (left), event frequency per microdomain (middle), and mean amplitude of Ca2+ transients (right) recorded in control and after CAtr treatment. Data shown as mean ± SEM. For each condition 10 individual cells from GLAST-mGC3;IP3R2−/−mice were analyzed. ns: not significant, ** p < 0.006, paired two-tailed Student’s t-test. (M) Image of an astrocyte (median intensity projection of time-series image stack) showing active regions during 286 s in a GLAST-mGC3;IP3R2−/−mouse. (N) Maps of all spontaneously active microdomains during 260 s in control (left), after light exposure (Photoact., middle), and after mPTP-I treatment (right). (O) Raster plots displaying Ca2+ transients in an astrocyte in control (left), after light exposure (Photoact., middle), and after mPTP-I treatment (right). (P) Graphs showing changes in number of domains per cell (top) and event frequency/domain (bottom) in control, after light exposure (Photoact.), and after mPTP-I treatment. Data shown as mean ± SEM. N = 10 cells from GLAST-mGC3;IP3R2−/−mice. ns: not significant, *** p <0.0001, * p < 0.01 repeated measure one-way ANOVA analysis with Tukey’s multiple comparisons post hoc test.
Figure 8
Figure 8. Enhanced mitochondrial Ca2+ efflux from astrocytes in ALS model (SOD1G93A) mice (See also Figure S8)
(A) Schematic showing the in vivo two photon imaging configuration in which mice were allowed to walk on a linear treadmill. (B) Intensity versus time plots of microdomain activity in an astrocyte from a control mouse (top) and from a SOD1G93A mouse (bottom). (C) Raster plots displaying Ca2+ transients in astrocyte microdomains from a control mouse (left) and from a SOD1G93A mouse (right). (D, E) Graphs of the number of active microdomains per cell (D) and frequency of events per microdomain (E) in control and SOD1G93A mice. Data shown as mean ± SEM. N = 11 cells each from Control and SOD1G93A mice. * p < 0.01, ** p < 0.001 unpaired two-tailed Student’s t-test. (F) Images of single astrocytes in acute slices of motor cortex from a GLAST-mGC3 (top) and a SOD1G93A mouse (bottom) showing median intensity projection (pseudocolored) from 260 s after treatment with thapsigargin (Tpg, 1 μM, 60 minutes). (G) Maps of all active microdomains (Control: 22; SOD1G93A: 39) that occurred in 286 s in astrocytes shown in (F). (H) Intensity versus time plots of microdomain activity in a control (top) and in a SOD1G93A mouse (bottom) (colors correspond to locations shown in corresponding maps in G). (I) Raster plots displaying microdomain Ca2+ transients imaged over a period of 260 s recorded in Tpg in a control (top) and in a SOD1G93A mouse (bottom). (J) Graphs comparing the frequency (left) and mean amplitude (z-score, right) of microdomain Ca2+ transients in Tpg treated cortical slices from aged matched control and SOD1G93A mice. Data shown as mean ± SEM. N = 14 cells each from Control and SOD1G93A mice. * p < 0.03, unpaired two-tailed Student’s t-test. (K) Graphs comparing the frequency (left) and mean amplitude (z-score, right) of microdomain Ca2+ transients in SOD1G93A mice before (baseline) and after mPTP inhibition (mPTP-I, 30 min). Data shown as mean ± SEM. N = 10 cells each from Control and SOD1G93A mice. * p <0.02, ** p < 0.002, paired two-tailed Student’s t-test.

Comment in

Similar articles

Cited by

References

    1. Araque A, Carmignoto G, Haydon PG, Oliet SHR, Robitaille R, Volterra A. Gliotransmitters travel in time and space. Neuron. 2014;81:728–739. - PMC - PubMed
    1. Bánsághi S, Golenár T, Madesh M, Csordás G, RamachandraRao S, Sharma K, Yule DI, Joseph SK, Hajnóczky G. Isoform- and species-specific control of inositol 1,4,5-trisphosphate (IP3) receptors by reactive oxygen species. J Biol Chem. 2014;289:8170–8181. - PMC - PubMed
    1. Bernardi P, Petronilli V. The permeability transition pore as a mitochondrial calcium release channel: a critical appraisal. J Bioenerg Biomembr. 1996;28:131–138. - PubMed
    1. Bélanger M, Allaman I, Magistretti PJ. Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation. Cell Metabolism. 2011;14:724–738. - PubMed
    1. Cavelier P, Attwell D. Neurotransmitter depletion by bafilomycin is promoted by vesicle turnover. Neurosci Lett. 2007;412:95–100. - PMC - PubMed

Publication types

MeSH terms

Substances