You are on page 1of 17

Pharmacology & Therapeutics 225 (2021) 107840

Contents lists available at ScienceDirect

Pharmacology & Therapeutics

journal homepage: www.elsevier.com/locate/pharmthera

Purinergic signaling in nervous system health and disease:


Focus on pannexin 1
Juan C. Sanchez-Arias ⁎,1, Emma van der Slagt 1, Haley A. Vecchiarelli, Rebecca C. Candlish, Nicole York,
Penelope A. Young, Olga Shevtsova, Afnan Juma, Marie-Ève Tremblay, Leigh Anne Swayne ⁎
Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada

a r t i c l e i n f o a b s t r a c t

Available online 19 March 2021 Purinergic signaling encompasses the cycle of adenosine 5′ triphosphate (ATP) release and its metabolism into
nucleotide and nucleoside derivatives, the direct release of nucleosides, and subsequent receptor-triggered
downstream intracellular pathways. Since the discovery of nerve terminal and glial ATP release into the neuropil,
Keywords: purinergic signaling has been implicated in the modulation of nervous system development, function, and dis-
Pannexin ease. In this review, we detail our current understanding of the roles of the pannexin 1 (PANX1) ATP-release
Purinergic signaling channel in neuronal development and plasticity, glial signaling, and neuron-glial-immune interactions. We addi-
Nucleotides tionally provide an overview of PANX1 structure, activation, and permeability to orientate readers and highlight
Nervous system recent research developments. We identify areas of convergence between PANX1 and purinergic receptor ac-
Neurodevelopment
tions. Additional highlights include data on PANX1's participation in the pathophysiology of nervous system de-
Neurological diseases
velopmental, degenerative, and inflammatory disorders. Our aim in combining this knowledge is to facilitate the
movement of our current understanding of PANX1 in the context of other nervous system purinergic signaling
mechanisms one step closer to clinical translation.
© 2021 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
2. PANX1 in the healthy nervous system. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. PANX1 in neurological disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Funding sources . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Author contributions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11

Abbreviations: AD, Alzheimer's disease; ADO, adenosine; ADP, adenosine 5′ diphosphate; APP, amyloid precursor protein; Arg, arginine; ARP2/3, actin-related protein 2/3 protein com-
plex; Asp, aspartate; ATP, adenosine 5′ triphosphate; C, carboxyl; CD, cluster of differentiation; COVID-19, coronavirus disease of 2019; CRMP2, collapsin response mediator protein 2;
CX3CR1, CX3 chemokine receptor 1; ENT1, equilibrative nucleoside transport 1; ECL1/2, extracellular loop 1/2; GFAP, glial fibrillary acidic protein; HIV, human immunodeficiency
virus; ICL1, intracellular loop 1; IL, interleukin; KO, knockout; N, amino; N2a, Neuro-2a; NBTI, Nitrobenzylthioinosine; NMDA, N-methyl-D-aspartate; PANX1, pannexin 1; PANX1CT,
pannexin 1 carboxy-terminus; poly(I:C), polyinosinic:polycytidylic acid; PS1, presenilin 1; Tyr, tyrosine; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TM1-4, transmem-
brane 1–4; TNF-α, tumour necrosis factor-α; Trp, tryptophan; UDP, uridine 5′ diphosphate; UTP, uridine 5′ triphosphate..
⁎ Corresponding authors.
E-mail addresses: juansa@uvic.ca (J.C. Sanchez-Arias), lswayne@uvic.ca (L.A. Swayne).
1
Equal contribution.

https://doi.org/10.1016/j.pharmthera.2021.107840
0163-7258/© 2021 Elsevier Inc. All rights reserved.
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

1. Introduction pannexin family (for a general review see Boyce, Epp, Nagarajan, &
Swayne, 2018). PANX1 monomers oligomerize into heptameric chan-
Pannexin 1 (PANX1) is a nervous system-enriched protein with di- nels permeable to small and large anions (such as chloride and adeno-
verse physiological and pathophysiological roles exerted throughout sine 5′ triphosphate (ATP)) as well as cations (e.g., Narahari et al.,
the body. PANX1 is the most widely studied of the three-membered 2021; see BOX 1 for additional details and references in Supplementary

Box 1
PANX1 channel structure and pharmacology.
Structure
PANX1 monomers oligomerize to form heptameric channels with extracellular, intracellular and transmembrane domains systematic mutagenesis
study revealed that conserved residues in this loop also play important roles in CBX function, potentially by (Deng et al., 2020; Michalski et al.,
2020; Mou et al., 2020; Navis, Fan, Trang, Thompson, & Derksen, 2020; Qu et al., 2020; Ruan, Orozco, Du, & Lü, 2020). Each PANX1 subunit con-
tains an amino (N)-terminal loop, four membrane spanning helices (TM1- 4), two extracellular loops (ECL1/2), an intracellular loop (ICL1), and a
carboxy- terminus (PANX1CT) (Deng et al., 2020; Michalski et al., 2020; Mou et al., 2020; Penuela et al., 2007; Qu et al., 2020; Ruan et al.,
2020; Sosinsky et al., 2011). Both the C-terminus and N-terminus are intracellular, and the main pore is positively charged at both ends (Deng
et al., 2020; Mou et al., 2020; Ruan et al., 2020). Seven positive residues pairs (Trp74-Arg75) line the narrowest portion (less than 5 Å) of the chan-
nel near the extracellular domain (Michalski et al., 2020; Mou et al., 2020; Qu et al., 2020; Ruan et al., 2020). Computational models and electro-
physiology data suggest that PANX1 channels contain seven narrow side tunnels within the intracellular domain at the inter-subunit interface that
provide an additional pathway for small ion flux (López et al., 2020; Ruan et al., 2020).

Activation
Under normal physiological conditions the main pore of the PANX1 channel is blocked by the PANX1CT (Y.-H. Chiu, Schappe, Desai, & Bayliss, 2018;
Deng et al., 2020; Ruan et al., 2020; Sandilos et al., 2012). Individual PANX1CT regions can be displaced in a step-wise manner leading to progres-
sive increases in channel activity and allowing for fine-tuning of channel flux (Y. H. Chiu et al., 2017). This phenomenon has been observed during
reversible a1-adrenoceptor activation of the channel, though it remains unclear if the PANX1CT are physically or allosterically displaced (Y. H. Chiu
et al., 2017). Irreversible activation of the channel has been demonstrated to occur during apoptosis via caspase-mediated cleavage at a caspase
cleavage site (Asp378; Yang et al. 2015) on the PANX1CT (Chekeni et al., 2010; Y.-H. Chiu et al., 2018; Ruan et al., 2020; Sandilos et al.,
2012). Additional stimuli have been proposed to activate PANX1 channels, such as changes in membrane potential, increased intracellular calcium,
increased extracellular potassium, mechanical stretching, and phosphorylation by Src family kinases (at residues Tyr198 and Tyr308; reviewed by
(Y.-H. Chiu et al., 2018; Whyte-Fagundes & Zoidl, 2018)). In a recent report, Lopez et al. (2020) identified two additional putative PKA phosphory-
lation sites (at residues Tyr302 and Ser328) that regulate stretch induced PANX1 activation. Given that these residues are located near the inter-sub-
unit interface, their phosphorylation may affect ion movement through the channel’s side tunnels. Further studies are required to fully understand the
mechanisms by which PANX1 is modulated under various physiological and tissue-specific contexts.
Selectivity
Both structural and functional data support that activated PANX1 channels favour the passage of anions and large anionic molecules such as ATP and
glutamate (Y. H. Chiu et al., 2017; Ma et al., 2012; Narahari et al., 2021). For example, the positively charged Arg75 residues in the main pore of the
channel form a cation-π interaction with Trp74 that constrain the pore and play a role in anion permeability (Deng et al., 2020; Michalski et al., 2020;
Ruan et al., 2020). In a recent study by Yang et al. (2020), increased PANX1 expression levels and membrane localization after TNF-a stimulation was
correlated with increased intracellular Ca2+ in endothelial cells. Molecular dynamics modelling revealed negatively charged residues (Asp and Glu) lo-
cated in the channel tunnels that could facilitate the transport of cations or Ca2+ (Sanchez Arias et al., 2020). Moreover, under caspase-mediated
activation of the channel, positively charged metabolites and dyes can slowly flux through the channel, supporting a role for PANX1 in the release
of signaling metabolites during apoptosis, such as spermidine (Medina et al., 2020; Narahari et al., 2021).

2
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

PANX1 inhibitors: common uses, other targets and structures

Inhibitor Common Use Other Receptor Targets References Structure


Probenecid Gout P2X7 receptor (Dahl et al., 2013; Díaz et al., 2019; Silverman et al.,
treatment 2008; Willebrords et al., 2017)

C13H19NO4S
PubChem Identifier:
CID 4911.
Spironolactone Hypertension Nuclear receptor subfamily (Good et al., 2018)
treatment 3 group C member 2 (Navis et al., 2020)

C24H32O4S
PubChem Identifier:
CID 5833.
Trovafloxacin Antibiotic DNA gyrase (Garg et al., 2018; Poon et al., 2014)
Topoisomerase IV

C20H15F3N4O3
PubChem Identifier:
CID 62959.
Carbenoxolone Gap channel Hemichannels (connexin (Michalski et al., 2020; Michalski & Kawate, 2016;
(CBX) inhibitor 26, connexin 38) Ruan et al., 2020; Willebrords et al., 2017)
Gap junctions

C34H50O7
PubChem Identifier:
CID 636403.
10
Panx Peptide Hemichannels (connexin (Pelegrin & Surprenant, 2006; Wang, Ma, Locovei,
46) Keane, & Dahl, 2007)

C58H79N15O16
Amino acid sequence:
WRQAAFVDSY
PubChem Identifier:
CID 44233736.
Brilliant Blue Food dye Voltage gated sodium (Jo & Bean, 2011; Wang, Jackson, & Dahl, 2013)
FCF channels

C37H34N2Na2O9S3
PubChem Identifier:
CID 19700.

material). PANX1 channels can be activated by a number of different Tang, 2020; Young, Yao, Sun, Cass, & Baldwin, 2008), PANX1 has
mechanisms and blocked by several drugs (BOX 1). Their well- emerged as a key convergence point of multiple signaling pathways in-
characterized role in ATP release implicates PANX1 channels as new volved in nervous system health and disease (reviewed in Yeung, Patil,
players in the intricate game of purinergic signaling within the nervous & Jackson, 2020).
system (BOX 2). In line with the ever-expanding ways that nervous sys- In this review we focus on recent advances in our understanding of
tem cell and tissue development and function are controlled by the vast PANX1 regulation of neuronal morphological and functional plasticity,
repertoire of purinergic signaling players (e.g., ATP-release channels, glial biology, behaviour, and neurological disease. We highlight exam-
nucleotide P2X & P2Y receptors, extracellular nucleotidases, nucleoside ples of similarities and differences between PANX1 and the spectrum
transporters, and nucleoside P1 receptors, see BOX 2 for summary and of purinergic signaling players, and examine emerging links to inflam-
for reviews see Burnstock, 2020; Parkinson et al., 2011; Illes, Xu, & mation and infection within the nervous system.

3
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

Box 2
Cell type specific PANX1 expression and associated purinergic signaling systems in health and disease.

2. PANX1 in the healthy nervous system et al., 1992). In follow-up work, (Wicki-Stordeur et al., 2016), we ob-
served selective loss of adult mouse brain ventricular zone neural pre-
2.1. Neural precursor cells and PANX1 in neuronal development cursor cells that underwent virus-mediated Panx1 knockout (KO),
suggesting PANX1 was critical for their maintenance. This finding was
Neuronal development is comprised of a complex set of processes expected since proliferation is a critical factor for maintaining cell pop-
occurring both embryonically and postnatally, beginning with neural ulation size. Our methods did not detect evidence for PANX1 regulation
precursor cell proliferation, specification, migration, differentiation, of cell proliferation in vivo; albeit we cannot completely rule out this
and integration into circuitry (for reviews see Castello & Gleeson, possibility. In addition to comparing and contrasting the roles of
2021; Jurkowski, Bettio, Woo, Patten, Yau, & Gil-Mohapel, 2020; Reh PANX1 and its purinergic receptor counterparts in neural precursor bi-
et al., 2020). Neural precursor cells express PANX1 (Wicki-Stordeur ology, a significant knowledge gap that remains is the potential
et al., 2016; Wicki-Stordeur, Dzugalo, Swansburg, Suits, & Swayne, crosstalk between PANX1 and P2 receptors in these cells both in vitro
2012; Wicki-Stordeur & Swayne, 2013) and several different P2 recep- and in vivo. For example, as we have previously proposed (Wicki-
tor proteins (reviewed in Franke & Illes, 2006; Oliveira, Illes, & Ulrich, Stordeur et al., 2016), ATP released via PANX1 could act as a “don't eat
2016; Ulrich & Illes, 2014). Purinergic signaling, including ATP release me” signal for neural precursor cells. Neural precursor cells mediate
by PANX1 as well as both ionotropic and metabotropic purinergic (P2) phagocytosis for the clearance of cells during the early stages of neuro-
receptor signaling, regulates many aspects of neuronal development nal development by a non-canonical P2X7 receptor-dependent process
(for reviews see Cavaliere, Donno, & D’Ambrosi, 2015; Oliveira et al., that is inhibited by ATP (Lovelace et al., 2015). It is tempting to speculate
2016; Rodrigues, Marques, & Cunha, 2019; Swayne & Bennett, 2016). that PANX1-mediated ATP release could repel roving phagocytic neural
With the established role of purinergic receptor signaling in neural precursors to prevent premature clearance via phagoptosis, death by
precursor cell biology and neuronal development, PANX1's role as an phagocytosis (Brown & Neher, 2012; Brown, Vilalta, & Fricker, 2015).
ATP-release channel (first described by Bao, Locovei, & Dahl, 2004; Another potential point of PANX1-P2X7 receptor crosstalk in neural
reviewed in Lohman & Isakson, 2014) provided the foundation for its precursor cells, as well as in neurons, is ATP-induced PANX1 internaliza-
detection in neural precursor cells by our group (Wicki-Stordeur et al., tion, a phenomenon we discovered (Boyce, Kim, Wicki-Stordeur, &
2012). We observed decreased neural precursor cell proliferation in Swayne, 2015; Boyce & Swayne, 2017) using a Neuro-2a (N2a)
the presence of the gout remedy and PANX1 blocker, probenecid (see neuroblastoma-derived neural precursor cell model (Truding,
BOX 1; Silverman, Locovei, & Dahl, 2008), or the P2 receptor-blocker, Shelanski, Daniels, & Morell, 1974). We proposed (Swayne & Boyce,
pyridoxalphosphate-6-azophenyl-2′,4′-disulfonic acid (Lambrecht 2017) that this process, which depends on an ATP-triggered interaction

4
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

between PANX1 and P2X7 receptors (Boyce & Swayne, 2017), could actin-interacting protein collapsin response mediator protein 2
provide fine-tuning of purinergic receptor regulation of neural precur- (CRMP2) (Frederiksen, Wicki-Stordeur, Sanchez-Arias, & Swayne,
sor cell proliferation and differentiation by downregulating PANX1 2019; Wicki-Stordeur & Swayne, 2013; Xu et al., 2018). Probenecid
ATP-release channels when extracellular ATP levels are in excess. A re- disrupted the PANX1-CRMP2 interaction, and increased microtubule
cent study by our group (Boyce et al., 2020) suggests that ATP- stability and neurite outgrowth in N2a cells (Xu et al., 2018). Notably,
induced PANX1 internalization likely occurs via macropinocytosis, and CRMP2 is an important positive regulator of dendritic spines (Zhang
furthermore, PANX1 itself could regulate macropinocytosis of ATP. et al., 2016). Our current working model states that high levels of so-
Additionally, we also found that PANX1 regulates neural precursor matosensory cortical neuronal PANX1 at the beginning of the postnatal
cell differentiation (Wicki-Stordeur & Swayne, 2013). PANX1 protein critical period for dendritic spine development in mice (Schlaggar, Fox,
expression levels decreased significantly in N2a cells and primary neo- & O'Leary, 1993) allows PANX1 to physically sequester CRMP2 and
natal ventricular zone neural precursor cells upon retinoic acid- and ARP2/3, thereby preventing aberrant dendritic spine stabilization and
low-serum-induced differentiation (Wicki-Stordeur & Swayne, 2013). synapse development. Developmental downregulation of PANX1,
As follows, PANX1 disruption (via siRNA-mediated Panx1 knockdown through mechanisms under investigation, triggers the release of
or probenecid) stimulated marked neurite formation in N2a cells and CRMP2 and ARP2/3, allowing them to promote cytoskeletal changes
primary neonatal ventricular zone neural precursor cells, while overex- that enhance spine growth and stability. Whether purinergic receptor
pression of PANX1 (transient transfection with PANX1-EGFP) decreased crosstalk is involved in aspects of this mechanism, including perhaps
neurite extension. Relatedly, others observed decreased expression of the downregulation of PANX1 through ATP-mediated internalization
P2X7 receptors with neuroblastoma-derived cell line and neural precur- involving ionotropic P2X7 receptors, as outlined above, remains to be
sor cell neuronal differentiation, and disruption of P2X7 receptors pro- investigated.
moted neuronal differentiation and neurite formation (Miras-Portugal,
Sebastián-Serrano, De Diego García, & Díaz-Hernández, 2017; Wu 2.2. PANX1 regulates synaptic plasticity
et al., 2009). Notably, in the context of peripheral nerves, Panx1 KO
did not appear to affect their development in situ; however, Panx1 KO In addition to regulating the structural development of synapses, as
increased neurite outgrowth from dorsal root ganglion explants, and described above, PANX1 modulates hippocampal synaptic transmission
this effect was mimicked by application of apyrase (an enzyme that hy- and consequently synapse strength and plasticity. In hippocampal brain
drolyzes extracellular ATP) and the PANX1 blocker, 10Panx (BOX 1; slices, PANX1 disruption (Panx1 KO and inhibition of PANX1 channels)
Horton et al., 2017). Altogether, these findings suggest that PANX1 reg- enhanced excitability: induction of long-term potentiation was facili-
ulates neurite formation and/or stability, potentially through crosstalk tated, frequency of spontaneous excitatory postsynaptic currents was
with purinergic signaling and through interactions with the cytoskele- increased, and induction of long-term depression was hindered
ton and its regulators. (Ardiles et al., 2014; Bialecki et al., 2020; Gajardo3 et al., 2018;
Consistent with a role in neurite formation, our recent findings sug- Prochnow et al., 2012), suggesting that PANX1 modulates hippocampal
gest that PANX1 regulates dendritic spine development (Sanchez-Arias synaptic plasticity mechanisms that play a critical role in learning and
et al., 2019; Sanchez-Arias, Candlish, van der Slagt, & Swayne, 2020). As memory (Takeuchi, Duszkiewicz, & Morris, 2014). Panx1 KO-
neurons mature and form physically and functionally connected net- associated changes in synaptic plasticity were normalized by adenosine
works, highly dynamic protrusions arise from dendritic shafts and ma- supplementation (Prochnow et al., 2012), suggesting PANX1 regulates
ture into dendritic spines that express ionotropic postsynaptic synaptic adenosine receptor activity. Once in the extracellular space,
glutamate receptors and thereby receive excitatory synaptic input ATP is hydrolyzed into adenosine by extracellular nucleotidases
(Berry & Nedivi, 2017). Using confocal microscopy to image fluorescent (Hamann & Attwell, 1996; Wieraszko, Goldsmith, & Seyfried, 1989)
lipophilic cationic indocarbocyanine dye-labeled neuronal membranes, resulting in activation of A1 and A2A adenosine receptors (BOX 2)
we detected increased cortical neuron dendritic spine density associ- which inhibit neuronal activity (Dunwiddie & Masino, 2001). This ten-
ated with Panx1 KO both in situ (layer 5 pyramidal neurons), as well tative mechanism is consistent with the modulatory effect of adenosine
as in primary cortical neuron cultures at 12 to 13 days in vitro on neuronal networks (Fredholm, Chen, Masino, & Vaugeois, 2005).
(Sanchez-Arias et al., 2019). Accordingly, the expression levels of The studies revealing PANX1 regulation of synaptic plasticity build
ionotropic glutamatergic receptors (glutamate receptor 1 and gluta- on prior work demonstrating that ATP itself acts as an important
matergic receptor ionotropic, N-methyl-D-aspartate (NMDA) 2A) and synaptic modulator (Fields & Stevens, 2000; Zhang et al., 2003). When
postsynaptic density protein-95 were elevated in cortical synaptosomes co-released with other neurotransmitters at nerve terminals or by as-
from Panx1 KO mice. To determine if the Panx1 KO-mediated increase in trocytes, ATP can enhance or decrease excitability via a variety of
dendritic spine density was due to increased dendritic protrusion stabil- purinergic signaling pathways, such as ionotropic P2X receptors, metab-
ity, we developed new methods to measure dendritic protrusion dy- otropic P2Y receptors, and adenosine receptors (for examples see Lalo
namics in immature cortical neurons at 10 days in vitro (Sanchez- et al., 2014; Lalo, Palygin, Verkhratsky, Grant, & Pankratov, 2016; Tan
Arias et al., 2020). At this relatively early in vitro developmental stage et al., 2017; Zhang et al., 2003; and reviewed in Guzman & Gerevich,
(Takano, Xu, Funahashi, Namba, & Kaibuchi, 2015), Panx1 KO neurons 2016; Hnasko & Edwards, 2012; Illes, Burnstock, & Tang, 2019;
already exhibited increased dendritic protrusion density when com- Pankratov, Lalo, Krishtal, & Verkhratsky, 2009; Pankratov, Lalo,
pared to wild-type controls. Transient expression of PANX1-EGFP in Verkhratsky, & North, 2006; Pascual et al., 2005; Sebastião & Ribeiro,
both wild-type and KO cultures was associated with decreased dendritic 2015; Zimmermann, 2016). Although, Panx1 mRNA levels are very
protrusion density (Sanchez-Arias et al., 2020). In terms of dynamics, low in astrocytes and other glial cells (Zhang et al., 2014) under naïve
increased dendritic protrusion movement and turnover were observed conditions, new, more highly-selective, astrocytic Cre lines, such as
with transient PANX1-EGFP expression, while dendritic protrusions Aldh1l1-CreERT2 (Srinivasan et al., 2016), will help to disentangle the
were relatively stable in Panx1 KO cultures. Overall, these results sug- contributions of astrocytic and neuronal PANX1 to physiological and
gest that PANX1 acts as a developmental brake for the formation of den- pathophysiological (i.e., seizure) neuronal network activity. Rafael,
dritic spines. Cairus, Tizzoni, Abudara, and Vitureira (2020) found that neuronal-
Cytoskeletal dynamics play key roles in the processes involved in specific, but not astrocytic, Panx1 KO impaired P2X7 receptor-
dendritic spine development (Borovac, Bosch, & Okamoto, 2018; Dent, mediated presynaptic terminal expansion induced by chronic inactivity
2017). Our previous proteomic study identified two key cytoskeletal (tetrodotoxin) in hippocampal cultures; however, compensatory ho-
regulating proteins as novel PANX1 interactors: actin-related protein 3 meostatic synaptic plasticity changes to nerve terminal density (based
(ARP3, a component of the ARP2/3 complex) and the microtubule and on vesicular glutamate transporter 1 puncta) depended on both

5
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

neuronal and astrocytic Panx1 KO. We recently found that global Panx1 precursors. While Panx1 mRNA and PANX1 protein were not detected
KO primary cortical neuron cultures (with low astrocyte density) exhib- in glial cells in the retina (Dvoriantchikova et al., 2006), the velocity
ited larger and more abundant neuronal networks (Sanchez-Arias et al., by which retinal microglial processes extended and retracted was re-
2019), which could in part be explained by increased cortical dendritic duced in the presence of probenecid (PANX1 blocker, see BOX 1)
spine density observed both in situ and in vitro described above. More (Fontainhas et al., 2011). Peripherally, Panx1 mRNA was not detected
recently, ablation of the Panx1a gene in zebrafish disrupted expression in a Schwann cell-like cell line (RT4-B5) (Ray, Zoidl, Weickert, Wahle,
of genes involved in visual system development, visual processing, & Dermietzel, 2005); however, more recently, PANX1 protein was de-
and pre- and post-synaptic dopaminergic signaling. This also resulted tected in myelin and blood vessels (as well as in the axon) in sections
in abnormal locomotion in the dark and locomotion responses and from peripheral (sciatic) nerves of adult mice (Horton et al., 2017). Ad-
brain wave frequency transitions in response to light, suggesting that ditionally, Panx1 transcripts and PANX1 protein were detected in pri-
zebrafish Panx1a plays an important role in the development of neuro- mary mouse Schwann cell cultures, where they contributed to
nal networks involved in supporting visual-motor functions (Safarian, hypotonicity-induced ATP release dependent on intracellular RhoA
Whyte-Fagundes, Zoidl, Grigull, & Zoidl, 2020). and the cytoskeleton (Wei et al., 2021). Type II cells in the carotid
Together, these results suggest PANX1 plays an important role in body, which resemble glia, express PANX1 ex vivo (Zhang, Piskuric,
neuronal network development, and both neuronal and astrocytic Vollmer, & Nurse, 2012). Therefore, PANX1 has been detected in a spec-
PANX1 can contribute to purinergic modulation depending on the phys- trum of glial cell types both in the central and peripheral nervous
iological or pathophysiological context. systems.
In terms of its functional role in glial cells, PANX1 has been impli-
2.3. Glial and endothelial cell PANX1 cated in the release of ATP and other metabolites. For example, PANX1
regulates ATP and D-serine release from cultured cortical astrocytes in
There are mixed reports of Panx1 gene expression in glial cells under response to P2X7 receptor activation (Iglesias et al., 2009; Pan, Chou,
naïve conditions. According to the RNA-sequencing transcriptome and & Sun, 2015). PANX1 and P2X7 receptor inhibition reduced propidium
splicing database of glia, neurons, and vascular cells of the mouse cere- iodide uptake (Kovács et al., 2018) and glucocorticoid-induced cytosolic
bral cortex (https://www.brainrnaseq.org/), PANX1 is strongly translocation and release of the high mobility group box-1 pro-
expressed in oligodendrocyte precursor cells and ‘newly-formed oligo- inflammatory protein in primary cortical astrocyte cultures (Hisaoka-
dendrocytes’ at postnatal day 17, while very low levels are found in Nakashima et al., 2020). In the 1321-N1 astrocyte cell line transiently
myelinating oligodendrocytes at this timepoint, and microglia and en- expressing the rat P2X7 receptor, the PANX1 inhibitor, 10Panx, blocked
dothelial cells at postnatal day 7 (Zhang et al., 2014). It should be ATP-induced ethidium bromide dye uptake, a readout of large-pore
noted that the oligodendrocyte lineages for this database were obtained channel activity (Pelegrin & Surprenant, 2006); although it should be
from postnatal day 17 brains because it is the earliest timepoint at noted that 10Panx has also been reported to affect channels formed by
which the full spectrum of these cells is present; additional information connexin 46 (Patel, Zhang, & Veenstra, 2014; Wang, Ma, Locovei,
about the procedures and timepoints at which the various cell types Keane, & Dahl, 2007) and could have other offsite targets. Calcium
were purified can be found in Zhang et al. (2014). Consistent with the waves in astrocyte cultures were reduced by siRNA-mediated Panx1
results of Zhang and colleagues, Panx1 transcripts and PANX1 protein knock-down (Scemes, Suadicani, Dahl, & Spray, 2007), and elevated ex-
have also been detected at appreciable levels in primary oligodendro- tracellular potassium activated PANX1 channels (potassium levels were
cyte cultures (Domercq et al., 2010; Huang, Grinspan, Abrams, & elevated to pathologically high levels ranging from 20 mM to 130 mM,
Scherer, 2007). Additionally, Panx1 (and Panx2) mRNA were not de- which are levels detected in the context of seizure, cortical spreading
tected in glial fibrillary acidic protein (GFAP)-positive glia in adult rats depression or stroke (Ayata & Lauritzen, 2015; Ito, Ohno, Nakamura,
(Vogt, Hormuzdi, & Monyer, 2005), and Panx1 mRNA was not found in Suganuma, & Inaba, 1979))), leading to ATP release and subsequent
mouse brain astrocytes in situ under naïve conditions (Zappala et al., neuronal P2X7 receptor activation and increased neuronal excitability
2006). Yet, there are numerous reports that pannexins are expressed (Silverman et al., 2009). Long term exposure of astrocytes to patholog-
in astrocytic cells in culture conditions. PANX1 was detected in an astro- ically elevated extracellular potassium reduced calcium waves in a
cyte cell line (1231-N1) (Pelegrin & Surprenant, 2006), as well as in pri- PANX1 dependent manner, particularly when P2X7 receptors were im-
mary astrocyte cultures (Huang et al., 2007; Iglesias, Dahl, Qiu, Spray, & plicated (Scemes & Spray, 2012). Notably, extracellular ATP feeds back
Scemes, 2009; Lai et al., 2007), where there is evidence of functional to inhibit PANX1-mediated ATP release from cultured cortical astro-
PANX1 channels (Iglesias et al., 2009). In terms of location on hippo- cytes, which is likely a homeostatic response to prevent ATP-mediated
campal astrocytes, PANX1 was distributed on processes as discrete cell death (Jackson, Wang, Keane, Scemes, & Dahl, 2014); elevated ex-
puncta (Boassa, Nguyen, Hu, Ellisman, & Sosinsky, 2015). Sorted mi- tracellular potassium was able to attenuate this homeostatic response.
croglia from the spinal cord on embryonic day 13.5 also express Moreover, lactate dehydrogenase and caspase-3 were increased in re-
PANX1 (Rigato et al., 2012). In the blood-brain barrier, PANX1 has sponse to elevated extracellular potassium, but this did not occur in
been detected in smooth muscle (Burns, Phillips, & Sokoya, 2012), Panx1 KO astrocytes (Jackson et al., 2014), confirming an important
though there are conflicting reports on endothelial cell PANX1 expres- role for PANX1 responses to pathological stimuli. In mouse spinal cord
sion. As mentioned above, endothelial Panx1 transcripts are sparse ac- astrocytes (cultured and acute slice), fibroblast growth factor-1 led to
cording to a brain cell-type specific transcriptomic database (Zhang calcium induced ATP release, P2X7 receptor activation and subsequent
et al., 2014), and PANX1 was not detected in RBE4 immortalized cul- PANX1-mediated ATP release (Garré et al., 2010). In primary cortical as-
tured endothelial cells from rat brain capillaries (De Bock et al., 2012). trocyte cultures, pre-treatment with tumour necrosis factor-α (TNF-α)
Conversely, PANX1 was detected in the immortalized human endothe- and Thy-1 or β3 integrin increased PANX1 levels and altered its cellular
lial cell line (hCMEC/D3) (Kaneko et al., 2015), and endothelial cells of distribution (Lagos-Cabré et al., 2017). Further supporting a role for
the mouse retina (Dvoriantchikova, Ivanov, Panchin, & Shestopalov, PANX1 in ATP release from primary astrocyte cultures, ATP release
2006). was reduced in primary optic nerve astrocyte cultures from Panx1 KO
There are similarly mixed results in terms of PANX1 expression in mice (Beckel et al., 2014).
other types of glia, namely retinal and peripheral glia. Panx1 transcripts Whereas low levels of PANX1 were detected in mature astrocytes
were detected in cerebellar Bergmann glia (Zappala et al., 2006), which and microglia, its functional relevance in these cells could be enhanced
are thought to be a type of neural precursor cell, consistent with the ex- in pathologies associated with injury and inflammation where in-
pression of PANX1 in ventricular zone and hippocampal neural creased PANX1 activity and levels have been observed (e.g., stroke, for

6
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

review see Brocardo, Acosta, Piantanida, & Rela, 2019). Therefore, astro- AD (Flores-Muñoz et al., 2020). Moreover, probenecid reversed synaptic
cytic and microglial cultures in which PANX1 levels may be elevated plasticity impairments and normalized dendritic spine densities in APP/
compared to in situ, could still provide important insight into cellular PS1 transgenic mice (Flores-Muñoz et al., 2020). In hippocampal slices
PANX1 cell biology related to pathological contexts. It is also important from APP/PS1 transgenic mice, PANX1 contributed (albeit relatively
to note that the low levels of astrocytic and microglial PANX1 in situ less than did connexin 43) to dye uptake in astrocytes associated with
under naïve conditions could still be functionally important. The puta- plaques (Yi et al., 2016). Notably, the broad spectrum anti-bacterial/
tive role(s) of PANX1 in oligodendrocytes, where appreciable Panx1 fungal/inflammatory agent minocycline (known to act on microglia, as-
transcripts are detected, is an area of future investigation with trocytes, endothelial cells, and other immune cells) blocked this effect,
important clinical relevance. Finally, it is important to note that our un- suggesting inflammation drives the opening of PANX1 channels. In cul-
derstanding of glial PANX1 will be greatly aided by the enhanced cell- tured astrocytes, amyloid-β increased surface PANX1 levels (Orellana
type specificity of several relatively new Cre lines (e.g., Aldh1l1-driven et al., 2011). Additionally, chronic inflammation associated with AD pa-
Cre for astrocytic KO (Srinivasan et al., 2016; Winchenbach et al., thology produced an excess of pro-inflammatory molecules, including
2016))). TNF-α (Decourt, Lahiri, & Sabbagh, 2016). It was recently discovered
that application of TNF-α upregulated PANX1 expression in peripheral
3. PANX1 in neurological disease endothelial cells (Yang et al., 2020). Moreover, brain mast cells (which
store various pro-inflammatory mediators in secretory granules, includ-
3.1. PANX1 in neurodevelopmental disorders ing TNF-α; reviewed in Hendriksen, van Bergeijk, Oosting, & Redegeld,
2017) undergo rapid degranulation upon exposure to amyloid-β in a
Adverse changes in synaptic function and dendritic spine structure PANX1-dependent mechanism (Harcha et al., 2015). These neuro-
and density have been implicated in a range of developmental, degener- inflammatory mechanisms and their effects in neuron-glial interactions
ative and injury-based neuropsychiatric disorders (for reviews see should be further investigated in the context of the role of PANX1 in the
Forrest, Parnell, & Penzes, 2018; Nishiyama, 2019; Penzes, Cahill, AD brain. Similarly, chronic inflammation associated with AD pathology
Jones, VanLeeuwen, & Woolfrey, 2011). Thus, given PANX1's role in increased extracellular ATP levels leading to purinergic receptor activa-
dendritic spine development and synaptic plasticity, it is perhaps not tion (reviewed in Cieślak & Wojtczak, 2018). The distribution and ex-
surprising that Panx1 KO mice exhibited increased anxiety-like behav- pression levels of several P2Y receptor subtypes, and the levels of
iours, impaired spatial memory and object recognition (Gajardo et al., P2X7 receptors were reportedly altered in tissues from AD-affected
2018; Prochnow et al., 2012), increased day-long wakefulness and individuals and in several transgenic mouse models of AD expressing
motor activity (Kovalzon, Moiseenko, Kurtenbach, Shestopalov, & different mutations associated with familial forms of the disease
Panchin, 2017), as well as, reduced restraint stress-induced ATP release (reviewed in Erb, Cao, Ajit, & Weisman, 2015; Francistiová et al.,
within the hippocampus (Orellana et al., 2015). PANX1 and P2X7 re- 2020). Furthermore, within an APP/PS1 transgenic mouse background,
ceptors were also implicated in stress-induced astrocytic release P2rx7 KO was associated with reduced amyloid-β plaque load, normal-
of high-mobility group box-1, a neuroinflammatory protein implicated ization of cognitive and synaptic plasticity deficits, and modulation of
in major depressive disorder (Hisaoka-Nakashima et al., 2020). In cluster of differentiation (CD)8-positive T cell recruitment, but had no
contrast to the relatively minor neurological phenotypes associated impact on microglial activation or cytokine production (Martin et al.,
with Panx1 KO in mice, a human germline PANX1 variant, PANX1 2019). While the roles of glial PANX1 in AD remain largely elusive,
Arginine217Histidine (which results in reduced channel activity when PANX1 blockers (probenecid and the PANX1 mimetic peptides 10Panx
characterized in vitro in a heterologous expression system), was associ- and E1b) partially reversed amyloid-β induced dye uptake and current
ated with multi-system dysfunction, including hearing loss and in primary cortical microglia cultures, but not primary cortical astrocyte
neurodevelopmental delay (Shao et al., 2016). PANX1 single nucleotide cultures (Orellana, Shoji, et al., 2011). In the same study, exposure to
polymorphisms have also been linked to autism spectrum disorders conditioned media from amyloid-β treated microglia induced astrocytic
(Davis et al., 2012), but no association was found with schizophrenia connexin 43-mediated ATP and glutamate release that together trig-
(Gawlik, Wagner, Pfuhlmann, & Stöber, 2016). PANX1 variants have gered neuronal PANX1 activation via P2 and NMDA receptor activation
also been shown to affect platelet aggregation (Molica et al., 2015), oo- (Orellana, Shoji, et al., 2011).
cyte biology (Sang et al., 2019; Wang et al., 2021), and channel- PANX1 is also altered in other neurodegenerative disorders. In pri-
trafficking in melanoma tumors (Nouri-Nejad et al., 2021), but nervous mary cortical astrocyte cultures, α-synuclein (a protein linked to
system implications for these are currently unknown. The basis for the Parkinson's disease (Stefanis, 2012)) stimulated PANX1-mediated ATP
range in severity of phenotypes associated with mouse Panx1 KO and release in a complex mechanism implicating cytokine and nitric oxide
different human variants is not yet known, but it is important to caution production, as well as purinergic (P2X7 and/or P2Y1) and glutamatergic
against comparison between complete Panx1 KO (Panx1tm1.1Fama receptor signaling (Díaz et al., 2019). Treatment of primary cultured as-
(Anselmi et al., 2008)); Panx1tm1.2Vmd (Qu et al., 2011)); Panx1tm1.1Vshe trocytes with α-synuclein also decreased intracellular PANX1 levels
((Dvoriantchikova et al., 2012)) or partial protein loss (Panx1tm1a (Díaz et al., 2019). Selective purinergic and adenosine receptor inhibi-
(KOMP)Wtsi
(Seminario-Vidal et al., 2011); discussed in Cone, Ambrosi, tion is used to provide symptomatic relief in Huntington's and
Scemes, Martone, & Sosinsky, 2013; Hanstein et al., 2013)) versus ex- Parkinson's disease (Oliveira-Giacomelli et al., 2018). In particular, inhi-
pression of full-length PANX1 variant at single amino acids. In the latter bition of adenosine A2A receptors in Huntington's disease is associated
case, the protein is still expressed and would likely still interact with key with symptom mitigation (Blum et al., 2003, 2018; Oliveira-Giacomelli
proteins and signaling partners. The consequences of variants on et al., 2018). Though A2A receptors were downregulated in
PANX1-interactions and its role as a signaling hub (Frederiksen, Huntington's disease, their downstream signaling pathways were am-
Wicki-Stordeur, Sanchez-Arias, & Swayne, 2019; Wicki-Stordeur & plified, suggesting these receptors or their signaling pathways could
Swayne, 2014) remain to be determined. be targeted for therapeutic intervention (Glass, Dragunow, & Faull,
2000; Varani et al., 2001). The precise mechanistic details of the
3.2. PANX1 in neurodegenerative disorders crosstalk between PANX1 and receptor systems associated with
purinergic signaling in AD and Huntington's disease, as well as in
PANX1 has also been also implicated in Alzheimer's disease (AD). Al- other neurodegenerative diseases associated with abnormalities in den-
though PANX1 is downregulated postnatally in healthy brain, its ex- dritic spines represent clinically-relevant knowledge gaps to bridge in
pression was maintained at high levels in the amyloid precursor future work (reviewed in Penzes et al., 2011; Forrest et al., 2018;
protein (APP)/presenilin 1 (PS1) transgenic mouse model of familial Nishiyama, 2019).

7
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

3.3. PANX1 in stroke, seizure, and traumatic brain injury (Dossi et al., 2018; Santiago et al., 2011; Scemes, Velíšek, & Velíšková,
2019; Wellmann, Álvarez-Ferradas, Maturana, Sáez, & Bonansco,
In accordance with its modulation of synaptic plasticity and its up- 2018). Microglial PANX1 was upregulated by proinflammatory cyto-
regulation during disease states, PANX1 is also important acutely during kines (e.g., TNF-α, interferon-γ (Sáez et al., 2013)). Trovafloxacin, a
cerebral ischemia, as well as in the short- and long-term changes that broad-spectrum antibiotic and PANX1 channel inhibitor (see BOX1), ex-
occur in the brain after stroke and seizure. The first indication of a link hibited anti-inflammatory and neuroprotective effects in mice suffering
between brain injury and PANX1 came with the discovery of the activa- from brain trauma (Garg et al., 2018). There was a reduction in pro-
tion of PANX1 channels under ischemic and excitotoxic conditions inflammatory cytokine levels and fewer microglia localized to the site
(Thompson, 2006; Thompson et al., 2008). NMDA receptor activation of injury associated with trovafloxacin treatment (Garg et al., 2018).
of Src kinases results in PANX1 C-terminus phosphorylation at tyrosine The authors hypothesized that trovafloxacin-mediated inhibition of
(Tyr)308, thereby promoting channel activation (Weilinger et al., 2016; microglial PANX1-mediated ATP release hindered the ability for microg-
Weilinger, Tang, & Thompson, 2012). Disrupting PANX1 by blocking lia to respond to injury (Garg et al., 2018). In a follow-up study,
PANX1 channels (e.g., probenecid) or via genetic ablation (global KO microglial-specific conditional Panx1 KO reduced neuroinflammation
or endothelial-specific KO) reduced infarct volume in models of ische- and other deleterious effects associated with traumatic brain injury
mic (Bargiotas et al., 2011; Freitas-Andrade, Bechberger, MacVicar, (Seo, Dalal, Calderon, & Contreras, 2020). While microglia express neg-
Viau, & Naus, 2017; Good et al., 2018; Xiong et al., 2014) and hemor- ligible levels of Panx1 transcripts in the naïve mouse brain (Zhang et al.,
rhagic (Zhou et al., 2018) stroke, and prevented cell death in in vitro 2014), these findings suggest that inflammation and injury could upreg-
models (Jian et al., 2016; Weilinger et al., 2016; Wicki-Stordeur et al., ulate PANX1 expression and functional protein levels (Seo et al., 2020).
2016). It is reasonable to speculate that ischemia-induced PANX1 chan- Collectively, these studies implicate astrocytic and microglial PANX1 in
nel opening could also trigger irreversible C-terminus caspase cleavage, neurological diseases associated with neuroinflammation such as stroke
a phenomenon that has been observed in other non-nervous system and seizure.
contexts (Chekeni et al., 2010; Medina et al., 2020; Sandilos et al., Nucleoside transporters have also been implicated in stroke and sei-
2012), but whether this occurs in the brain in the context of injury or zure, particularly equilibrative nucleoside transporter 1 (ENT1). Expres-
stroke remains to be confirmed. The increase in ATP released in the ex- sion levels of ENT1 peaked 12 h after middle cerebral artery occlusion in
tracellular space following stroke was associated with activation of the rat, and administration (30 min after stroke induction) of an ENT1
P2X7 receptors, as well as excessive calcium influx and neuronal inhibitor, Nitrobenzylthioinosine (NBTI), reduced infarct volume, im-
death (Arbeloa, Pérez-Samartín, Gottlieb, & Matute, 2012; Martín, proved neurological function (measured at 24 h and 72 h post-stroke in-
Domercq, & Matute, 2018). The P2X7 receptor antagonist, Brilliant duction), and prevented neuronal apoptosis (and associated signaling
Blue G, markedly reduced brain damage associated with transient pathways), likely by elevating extracellular adenosine (Zhang et al.,
focal cerebral ischemia in a rat model (Arbeloa et al., 2012). Taken to- 2020). Similarly, in the context of seizure, ENT1 protein levels were el-
gether, these findings suggest that in the context of brain injury, both evated in the temporal neocortex from patients with temporal lobe ep-
NMDA receptors and P2X7 receptors are linked to PANX1 channels ilepsy, as well as in hippocampal lysates prepared from a rat model of
with dire consequences for the neurons involved. seizure (lithium chloride, followed by pilocarpine administration).
In addition to affecting neuronal survival, several studies suggest NBTI suppressed seizure susceptibility (longer onset latency and re-
glial PANX1 is also important in stroke and seizure. For example, duced seizure severity) in the rat model both when applied locally via
ischemia-triggered PANX1-mediated ATP release from oligodendro- microinjection and when administered intraperitoneally (Xu et al.,
cytes led to P2X7 receptor-dependent oligodendrocyte death 2015). NBTI also decreased rat hippocampal CA1 pyramidal neuron ex-
(Domercq et al., 2010). Several studies have shown that astrocyte and citability and mini excitatory postsynaptic currents in slices prepared
microglia responses to stroke are tempered by blocking PANX1, which 24 h after the onset of seizure (Xu et al., 2015). A follow-up study re-
could not necessarily be attributed to glial PANX1 per se. For example, vealed that the impact of seizure on ENT1 expression levels is regulated
probenecid reversed transient ischemia/reperfusion increases in GFAP by dynamic-related protein 1, a key protein involved in mitochondrial
and ionized calcium binding adaptor molecule 1 reactivity (Wei et al., fission (Luo et al., 2020). Selective inhibition of dynamic-related protein
2015), and in female Panx1 KO mice, there were fewer GFAP-positive 1 function by intraperitoneal administration of mitochondrial division
astrocytes and ionized calcium binding adaptor molecule 1-positive mi- inhibitor 1, 30 min prior to induction of status epilepticus, attenuated
croglia (Freitas-Andrade et al., 2017). Furthermore, probenecid reduced seizures and prevented the increase in ENT1 expression levels (Luo
lactate dehydrogenase activity, cell death, reactive oxygen species et al., 2020). The same group also showed that inhibition of p38
levels, NOD-, LRR-, and pyrin domain-containing protein 3 levels, mitogen-activated protein kinase blocked seizure-induced expression
caspase-1 levels, aquaporin 4 levels, high mobility group BOX 1 levels, level increases in the A1 receptor and ENT1, reduced seizure onset la-
and interleukin (IL)-1β secretion in primary astrocyte cultures follow- tency, and decreased the number of seizures in the same rat model
ing oxygen and glucose deprivation (6 h) and reoxygenation (24 (Zhou et al., 2020).
h) (Jian et al., 2016). Additionally, probenecid reversed focal cerebral The potential crosstalk between PANX1, purinergic receptors, aden-
ischemia-induced increases in CD68-positive cells (microglia or macro- osine receptors, and nucleoside transporters between various brain cell
phages), and partially normalized increased GFAP/aquaporin 4 levels types in stroke and seizure represents an important knowledge gap
(Xiong et al., 2014). Despite low astrocytic and microglial PANX1 ex- with key translational implications.
pression levels under naïve conditions, given that various types of injury
and inflammation have been associated with increases in PANX1 ex- 3.4. PANX1 in pain
pression levels (e.g., inflammation (Yang et al., 2020)) and stroke
(Freitas-Andrade et al., 2017; Jiang et al., 2012)), one possible mecha- Consistent with its importance in shaping basal plasticity and
nism for astrocytic and microglial PANX1 involvement in stroke/injury neuronal networks as well as its implication in maladaptive plasticity
and seizure could be a possible injury/inflammation-mediated PANX1 in seizure, PANX1 also plays a role in neuropathic and mechanically-
upregulation in these cells. Relatively higher PANX1 expression levels induced chronic pain (Bravo, Maturana, Pelissier, Hernández, &
were detected in GFAP-positive astrocytes in lesions from patients Constandil, 2015; von Hehn, Baron, & Woolf, 2012; Woolf, 2011). Vari-
with three types of focal cortical dysplasia compared to healthy controls ous central and peripheral mechanisms link PANX1 with biological pro-
(Li et al., 2017). Astrocytic and neuronal PANX1 activation has also been cesses involved in hypersensitivity (mechanical allodynia) and central
implicated in ictal-like activity in brain tissue from patients with epi- sensitization. For instance, in dorsal root ganglia, ATP released from sen-
lepsy and kainate-induced and kindling-induced seizures in rodents sory neurons activated satellite glial cell P2X7 receptors, eliciting

8
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

calcium entry and pro-inflammatory cytokine (TNF-α and IL-1β) re- other purinergic signaling players. For example, sustained P2X7 recep-
lease, thereby increasing neuronal network excitability (Mousseau tor activation has been associated with the formation of a high conduc-
et al., 2018; Zhang, Chen, Wang, & Huang, 2007). Dorsal root ganglion tance pore causing cell death (for review see Khakh & Alan North,
PANX1 expression levels were elevated in rodent models of neuropathic 2006), but there is also evidence that extracellular ATP promotes cancer
and chronic pain, contributing to an increase in extracellular ATP. In progression in myriad ways (Di Virgilio et al., 2018; Di Virgilio &
light of this increase in PANX1 associated with neuropathic and chronic Adinolfi, 2017; Vultaggio-Poma, Sarti, & Di Virgilio, 2020). Further
pain, it is perhaps not surprising that blocking PANX1 function de- support of the hypothesis that PANX1 may be involved in nervous
creased hyperalgesia, mechanical allodynia, and release of pro- system cancers, comes from the emergent neural precursor-like
inflammatory cytokines (Bravo et al., 2014; Mousseau et al., 2018; nature of nervous system tumour cells (Couturier et al., 2020; Jessa
Zhang, Laumet, Chen, Hittelman, & Pan, 2015). Panx1 KO mice subjected et al., 2019) taken together with our previous discovery of PANX1
to sciatic nerve injury were protected from development of mechanical regulation of neural stem cell proliferation, maintenance and differenti-
allodynia, a process requiring PANX1 expression in circulating hemato- ation (Wicki-Stordeur et al., 2012, 2016; Wicki-Stordeur & Swayne,
poietic cells (Weaver et al., 2017). Spinal cord microglial PANX1 was re- 2013). We found that PANX1 overexpression increased N2a mouse
cently shown to be critically important for mediating aspects of pain neuroblastoma-derived cell proliferation that was blocked by probene-
and responses to analgesics (Burma et al., 2017; Mousseau et al., cid treatment (Wicki-Stordeur et al., 2012) and siRNA-mediated Panx1
2018). In a mouse model of arthritis, genetic deletion of microglial knock-down and probenecid treatment resulted in neurite outgrowth
PANX1 attenuated the development of mechanical allodynia (Wicki-Stordeur & Swayne, 2013), reminiscent of the effects of retinoic
(Mousseau et al., 2018). Moreover, probenecid treatment suppressed acid treatment on these cells (Shea, Fischer, & Sapirstein, 1985). Nota-
mechanical allodynia without affecting acute nociception (Mousseau bly, retinoic acid is used clinically for the treatment of neuroblastoma
et al., 2018). Repeated morphine administration increased PANX1 ex- (Reynolds, Matthay, Villablanca, & Maurer, 2003), suggesting PANX1
pression levels in spinal cord microglia (Burma et al., 2017). Mice lack- could be an important therapeutic target for nervous system cancers
ing PANX1 in CX3 chemokine receptor 1 (CX3CR1)-positive microglia like neuroblastoma. More recently, PANX1 was detected in human
showed attenuated morphine withdrawal symptoms and naloxone- neuroblastoma tumour specimens and high-risk patient derived cell
induced synaptic facilitation in the dorsal horn, and naloxone-induced lines and probenecid reduced neuroblastoma progression in vitro and
ATP release was mediated by PANX1 (Burma et al., 2017). Despite in vivo (Holland, 2020). We previously showed that ATP induced
these findings, CX3CR1-driven Panx1 KO microglia did not prevent the PANX1-P2X7 receptor clustering and internalization in N2a cells
development of opioid-induced tolerance or hyperalgesia (Burma (Boyce et al., 2015; Boyce & Swayne, 2017). Building on that work, we
et al., 2017). Overall, these results implicate PANX1 in microglial mech- recently discovered that PANX1 may regulate macropinocytosis of ATP
anisms underlying the spinal cord processing of pain and suggest that in N2a cells (Boyce et al., 2020). Macropinocytosis of ATP confers drug
PANX1 pharmacological manipulation holds potential to treat these resistance to cancer cells by facilitating uptake of ATP and extracellular
complex disorders. debris (Jayashankar & Edinger, 2020; Qian et al., 2014) as a means of
Although there is no evidence supporting a role for PANX1 in acute obtaining additional energy (Di Virgilio et al., 2018; Recouvreux &
nociception, PANX1's mechanosensitive properties, first described in Commisso, 2017; Vultaggio-Poma et al., 2020), adding new metabolic
(Bao et al., 2004), raise the possibility that PANX1 could play a role in implications of PANX1 in cancer cell biology.
touch sensation. Intriguingly, in Caenorhabditis elegans touch receptor Cancer cells also use ectonucleotidases and nucleoside transporters
neurons, mouse PANX1 rescued touch sensation that was lost via KO to increase adenosine concentrations in the extracellular milieu. Glioma
of the gene encoding for the uncoordinated-7 mechanosensitive cells express high levels of membrane-bound ectonucleotidases, specif-
innexin hemichannel-forming protein (i.e., a half gap junction forming ically CD39 and CD73, whose sequential activity produces adenosine
a single membrane channel), suggesting PANX1 and its invertebrate from ATP (reviewed in Ceruti & Abbracchio, 2020). Additionally, rela-
innexin relatives could play role(s) in neuronal mechanotransduction tively low levels of ENT1 were detected in primary stem-like cells and
under physiological conditions as well (Walker & Schafer, 2020). U87MG glioblastoma cells, further raising extracellular adenosine by
preventing its re-uptake (Alarcón et al., 2020). High levels of extracellu-
3.5. PANX1 in nervous system cancers lar adenosine allow glioma cells to modulate the host immune response
(primarily through A1 and A2A receptors in peri-tumour tissue) and
In healthy tissues, extracellular ATP and adenosine concentrations boost their tumorgenicity, by promoting angiogenesis via A2 and A3 re-
are typically at nanomolar levels (Blay, White, & Hoskin, 1997; ceptors (Ceruti & Abbracchio, 2020; Gessi, Merighi, Sacchetto, Simioni,
Falzoni, Donvito, & Di Virgilio, 2013; Yegutkin, 2014); however, their & Borea, 2011). Given the intricate connection between PANX1 and
concentrations in the tumour microenvironment are significantly purinergic receptors (in particular P2X7 receptors) in tumorigenicity
higher, reaching hundreds of micromolar levels (Alarcón et al., 2020; and associated host immune response (modulated by adenosine recep-
Ohta et al., 2006; Pellegatti et al., 2008). Depending on their concentra- tors), further pharmacological or genetic manipulations that dampen
tions and subsequent receptor subtype affinities, ATP and adenosine ATP release (e.g., by targeting PANX1) and signaling (e.g., by targeting
play roles in the promotion and suppression of tumour growth and cy- P2 receptors, ectonucleotidases and nucleoside transporters) represent
totoxicity, as well as in the activation of the host immune system (Di novel potential therapeutic approaches to cancer.
Virgilio & Adinolfi, 2017; Di Virgilio, Sarti, Falzoni, De Marchi, &
Adinolfi, 2018). In support of the idea that PANX1 might play a role in 3.6. Highlight on PANX1 and purinergic signaling in neuron-microglial
brain cancer, rat C6 glioma cells overexpressing PANX1 exhibited in- interactions
creased tumorigenicity via accelerated assembly of three-dimensional
compact aggregates (Bao, Lai, Naus, & Morgan, 2012), although it should Microglia play a critical role in maintaining homeostatic neuronal ac-
be noted that PANX1 was previously suggested to be a tumour suppres- tivity during both physiological and inflammatory conditions (reviewed
sor in C6 glioma cells (Lai et al., 2007). In human glioblastoma U87-MG in Illes, Rubini, Ulrich, Zhao, & Tang, 2020). Under naïve conditions,
cells, PANX1 levels were upregulated in mitotic cells and Panx1 knock- microglial processes are highly dynamic and reach out to survey their
down decreased cell proliferation (Wei, Yang, Shi, & Chen, 2015). Nota- environment where they regulate synaptic pruning and network plas-
bly, coincident application of ATP in the presence of Panx1 knock-down ticity (Tremblay et al., 2011). In the context of inflammation associated
resulted in a greater reduction in cell proliferation (Wei, Yang, et al., with infection and several different neurological diseases, microglia are
2015), implying a level of divergence of PANX1 and ATP-dependent triggered to move towards sites of injury to phagocytose dead or dying
mechanisms in cancer that could be dependent on the expression of cells and take part in antigen presentation and cytokine production

9
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

(reviewed by Bachiller et al., 2018). Increased extracellular ATP associ- regulator of the venous endothelium inflammatory response. Similarly,
ated with inflammation acts on ionotropic P2X and metabotropic P2Y endothelial-specific deletion of Panx1 resulted in reduced infarct vol-
receptors (see BOX2, reviewed by Illes, Rubini, Ulrich, Zhao, & Tang, ume, leukocyte infiltration, and vascular tone (Good et al., 2018).
2020) on microglial membranes, contributing to their migration and in- Given that integrity of the vascular system is paramount to sustain nor-
creased inflammatory function (Illes, Xu, & Tang, 2020; Tremblay et al., mal brain function and that inflammation and vascular pathology are
2011; Walz, Ilschner, Ohlemeyer, Banati, & Kettenmann, 1993). For in- important contributors to neurodegenerative disease (Chitnis &
stance, expression of microglial P2Y12 receptors, associated with the Weiner, 2017; Mangiafico, Sarnataro, Mangiafico, & Fiore, 2006;
ramified microglial morphology observed under physiological condi- Phillips & Mate-Kole, 1997; Sweeney, Kisler, Montagne, Toga, &
tions (Sipe et al., 2016), is reduced significantly following microglial re- Zlokovic, 2018; Tasci et al., 2018), PANX1's role in purinergic and cyto-
activity to ATP (Haynes et al., 2006). Haynes et al. (2006) found that kine signaling during neuroinflammatory responses marks PANX1
microglia lacking P2Y12 receptors were unable to migrate or project modulation as a possible treatment of cerebrovascular pathologies.
their processes towards extracellular nucleotides in vivo and in vitro, PANX1 channels are strongly associated with inflammation. PANX1
suggesting that these receptors are important for microglia chemotaxis, channels are, in part, responsible for inflammatory ATP release
particularly in response to nucleotides (Kyrargyri et al., 2020; Madry (Chitnis & Weiner, 2017; Dahl, Qiu, & Wang, 2013; Linden, Koch-
et al., 2018). It is tempting to speculate that neuronal, astrocytic, or oli- Nolte, & Dahl, 2019; Makarenkova, Shah, & Shestopalov, 2018;
godendroglial PANX1 channels could be potential sources of ATP release Pelegrin & Surprenant, 2006; Penuela, Gehi, & Laird, 2013) and PANX1
for communication with microglial P2Y12 receptors. For instance, ATP also regulates inflammasome activation and regulation of cytokine re-
release induced by neuronal NMDA receptor activation following lease in a variety of cell types (reviewed by Makarenkova, Shah, &
kainate-induced seizure activity or glutamate application promoted Shestopalov, 2018). Inflammasomes are intracellular protein complexes
microglial process extension, which was abolished with P2ry12 KO or that form in response to pathogen-associated molecular patterns or
probenecid treatment (Eyo et al., 2014). Notably, ATP and NMDA- danger-associated molecular patterns (reviewed by Zheng, Liwinski, &
triggered microglial process extension was still observed with Elinav, 2020) and induce caspase activation as well as an inflammatory
carbenoxolone treatment (PANX1 and gap junction inhibitor, BOX1) response, including pyroptosis, a type of inflammatory cell death dis-
and Panx1 global KO (Dissing-Olesen et al., 2014); future investigations tinct from apoptosis (Yang, He, Muñoz-Planillo, Liu, & Núñez, 2015;
using more selective pharmacological agents and cell-type specific KO Zheng et al., 2020). ATP promotes IL-1β secretion and release during
models could shed some light on the mechanisms underlying neuron- pathogen-associated molecular pattern-mediated responses through
glial communication. In addition to injury and inflammation, we de- activation of P2X7 receptors and by acting as an enhancer signal
scribed how neuronal activity itself can result in changes to microglial (Crespo Yanguas et al., 2017; Di Virgilio, Dal Ben, Sarti, Giuliani, &
morphology and function (Tremblay, Lowery, & Majewska, 2010; Falzoni, 2017; Makarenkova, Shah, & Shestopalov, 2018; Zhou, Green,
reviewed in Tremblay et al., 2011), in part by increasing local concentra- Bennet, Gunn, & Davidson, 2019). Nuclear factor-κβ activation follow-
tions of extracellular ATP (Badimon et al., 2020). Primary cultured neo- ing TNF-α-triggered signaling, upregulated PANX1 expression levels
natal microglia convert neuronally-released ATP to adenosine, eliciting and increased trafficking of PANX1 channels to the membrane (Yang
neuronal adenosine receptor-mediated activity suppression, thereby et al., 2020). This led to increased intracellular calcium and subsequent
creating a microglial-driven neuronal homeostasis negative feedback upregulation of IL-1β expression, processing, and release, suggesting
loop (Badimon et al., 2020). Somewhat relatedly, Ent2 KO, which re- PANX1 signals in a feed-forward manner during sterile inflammation.
sulted in elevated extracellular adenosine levels, provided resistance As mentioned above, probenecid reduced inflammasome activation,
to lipopolysaccharide-induced neuroinflammation (as assessed by mi- IL-1β and high mobility group BOX 1 release, NOD-, LRR- and pyrin
croglia reactivity and astrogliosis), blood-brain barrier breakdown domain-containing protein 3 and caspase-1 expression, and reactive ox-
(tight junction integrity, leakage) and neurotoxicity (based on IgG accu- ygen species production in response to inflammatory stimuli (Jian et al.,
mulation and caspase-3 cleavage) via activation of A1 and A2A recep- 2016), suggesting that inhibiting PANX1 might be therapeutic during
tors (Wu, Lee, Chou, Chern, & Lin, 2020). Altogether, these studies inflammatory conditions. Importantly, numerous studies have identi-
highlight the importance of purinergic signaling and possibly PANX1 in- fied an association between PANX1 channels and P2X receptors both
volvement in the microglial dynamics associated with synaptic pruning upstream and downstream of inflammasome activation, though cell
and inflammation. specific mechanisms of activation are still unclear (Hung et al., 2013;
Jeong et al., 2020; Makarenkova, Shah, & Shestopalov, 2018; Silverman
3.7. Peripheral PANX1 signaling with implications for nervous system et al., 2009). Given that PANX1-induced P2X7 receptor-mediated
diseases inflammasome signaling is amenable to therapeutic intervention, fur-
ther understanding of the mechanism behind activation is paramount
Along with roles in development and cell function in the nervous to translate these findings into interventions. Understanding the mech-
system, PANX1-associated processes in peripheral tissues have impor- anism will allow for treatment of heightened inflammatory states such
tant implications for nervous system function. These processes include as: sepsis, ischemic stroke, cortical spreading depression in migraine
regulation of vascular tone, immune cell migration, and inflammatory headache, chronic pain, and neuroimmune and neurodegenerative dis-
responses. For instance, α-1D-adrenergic receptor-mediated vasocon- orders (Dvoriantchikova et al., 2012; Pronin et al., 2019; Yeung et al.,
striction in vascular smooth muscle cells involves PANX1 activation 2020; Zhang et al., 2015; Zhou et al., 2019).
via phosphorylation at Tyr198 by Src kinases, and subsequent release Not only does PANX1 have a role in the host immune response, but it
of ATP (Billaud et al., 2011, 2015; DeLalio et al., 2019), implicating also plays a critical role in viral infection itself. For example, human im-
PANX1 in hypertensive vascular pathology (DeLalio et al., 2019). munodeficiency virus (HIV)-binding to its receptors on CD4-positive T
Notably, PANX1 phosphorylation at Tyr198 has also been linked to lymphocytes and primary human peripheral blood mononuclear cells
PANX1-mediated ATP release in venous endothelial cells following stimulated a long-lasting and biphasic opening of PANX1 channels
TNF-α stimulation (Lohman et al., 2015). Under these inflammatory (Orellana et al., 2013). HIV-binding to its cellular receptors has also
conditions, PANX1 activation promoted leukocyte adhesion and trans- been found to induce ATP release via PANX1 which subsequently signals
migration through venous vessel walls, in part, by upregulating the P2Y2 receptors to induce viral uptake and initiate replication (Séror
expression of vascular cell adhesion protein 1 in endothelial cells et al., 2011). Of interest, Swartz, Esposito, Durham, Hartmann, and
and also by facilitating ATP release (Chen et al., 2006; Grassi, 2010). Chen (2014) found that inhibition of P2X hindered HIV infection of
ATP subsequently activated P2Y2 and A3 receptors on leukocytes CD4-positive T lymphocytes. Notably, the elevation of circulating ATP
(Chen et al., 2006; Grassi, 2010), suggesting that PANX1 is a positive levels was recently identified as a biomarker for HIV-associated

10
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

cognitive impairment (Velasquez et al., 2020). It has been speculated astrocytes, or other glial cells (i.e., oligodendroglial lineage cells and mi-
that in the brain, particularly in glial cells, similar to the periphery croglia). Finally, further evaluation and validation of the PANX1 interac-
(Orellana et al., 2013), PANX1 facilitated purinergic receptor- tome in neuronal and non-neuronal nervous system cells will allow us
dependent cellular HIV infection (Malik & Eugenin, 2019). Once cells to bridge important knowledge gaps in our understanding of the molec-
are infected with HIV, HIV may interact further with PANX1 leading to ular mechanisms underlying the roles attributed to PANX1 and other
extracellular increases in glutamate and ATP, which drive downstream purinergic signaling mechanisms in nervous system health and disease.
effects of HIV infection including inflammation, cognitive deficits, and
excitotoxicity (Malik & Eugenin, 2019). In cultured astrocytes in a Funding sources
PANX1-dependent manner, HIV altered their permeability and calcium
dynamics, and increased release of ATP and inflammatory mediators This work was supported by operating grants from the Canadian
(Gajardo-Gómez et al., 2020). Institutes of Health Research (CIHR Grant MOP142215) and from the
Stemming from the critical roles of PANX1 in viral infection and in- Natural Sciences and Engineering Research Council (NSERC) [RGPIN-
flammation, we recently proposed PANX1 inhibition as a potential 2017-03889] to L.A.S. L.A.S. was also supported by a Michael Smith
treatment for the coronavirus disease of 2019 (COVID-19) (Swayne Foundation for Health Research (MSFHR) and British Columbia Schizo-
et al., 2020). Though there is currently no evidence showing an interac- phrenia Society Foundation Scholar Award (5900). J.C.S.A. was sup-
tion between PANX1 and severe acute respiratory syndrome coronavi- ported by a University of Victoria Fellowship Graduate Award. P.Y.
rus 2 (SARS-CoV-2), PANX1 activity has been implicated in the was supported by an NSERC CGS-M. H.A.V. was supported by a CIHR
propagation of inflammatory responses in tissues susceptible to Postdoctoral Fellowship and a MSFHR Research Trainee Award. M.E.T.
hyperinflammation following severe acute respiratory syndrome coro- is a Tier 2 Canada Research Chair in Neurobiology of Aging and
navirus 2 infection. In various models of injury, endothelial cell PANX1 Cognition.
disruption reduced inflammation (Good et al., 2018; Jankowski et al.,
2018; Sharma et al., 2018). As such, PANX1 inhibition could minimize Author contributions
damaging effects of inflammation and reduce the cytokine storm seen
in COVID-19 patients (Hui et al., 2020). Given the role of PANX1 in the The manuscript idea was conceived by LAS in collaboration with
regulation of neurological plasticity and its links to viral infection and JCSA, EVDS, HAV and MET. JCSA and EVDS made equal contributions.
inflammation, PANX1 should also be investigated in the context of the JCSA, EVDS, HAV, RCC and LAS were the primary writers. All authors re-
neurological effects of COVID-19 (Li, Bai, Hashikawa, & Yan-Chao Li, vised the manuscript.
2020).
Maternal infection during pregnancy and subsequent maternal im- Declaration of Competing Interest
mune activation underlies a number of neurological disorders
(reviewed in Brown & Conway, 2019). Polyinosinic:polycytidylic acid The authors have no conflicts of interest to declare.
(poly(I:C)), a viral infection mimic that induces maternal immune acti-
vation as a model for autism spectrum disorders (Reisinger et al., 2015), References
was used to investigate the role of P2X7 receptors in this process. Mater-
Alarcón, S., Toro, M.Á., Villarreal, C., Melo, R., Fernández, R., Ayuso Sacido, A., ... Quezada, C.
nal poly(I:C) injection produced an autism-like phenotype in offspring, (2020). Decreased Equilibrative Nucleoside Transporter 1 (ENT1) activity contributes
as well as increased levels of the pro-inflammatory cytokine, IL-6, in the to the high extracellular adenosine levels in mesenchymal glioblastoma stem-like
fetal brain (Horváth et al., 2019), and these effects were normalized by cells. Cells 9(8), 1914. https://doi.org/10.3390/cells9081914.
Anselmi, F., Hernandez, V. H., Crispino, G., Seydel, A., Ortolanoa, S., Roper, S. D., ...
genetic or pharmacological disruption of the P2X7 receptor. Addition- Mammano, F. (2008). ATP release through connexin hemichannels and gap junction
ally, several adverse outcomes in poly(I:C)-treated mice were reversed transfer of second messengers propagate Ca2+ signals across the inner ear.
by administration of suramin, a non-selective purinergic antagonist, or Proceedings of the National Academy of Sciences of the United States of America 105
(48), 18770–18775. https://doi.org/10.1073/pnas.0800793105.
via downregulation of purinergic receptors (Naviaux et al., 2013).
Arbeloa, J., Pérez-Samartín, A., Gottlieb, M., & Matute, C. (2012). P2X7 receptor blockade
PANX1's role in ATP release, pro-inflammatory cytokine signaling and prevents ATP excitotoxicity in neurons and reduces brain damage after ischemia.
viral infection, together with the identification of PANX1 single- Neurobiology of Disease 45(3), 954–961. https://doi.org/10.1016/j.nbd.2011.12.014.
nucleotide polymorphisms associated with risk for autism spectrum Ardiles, A. O., Flores-Muñoz, C., Toro-Ayala, G., Cárdenas, A. M., Palacios, A. G., Muñoz, P., ...
Martínez, A. D. (2014). Pannexin 1 regulates bidirectional hippocampal synaptic plas-
disorders (Davis et al., 2012), support further investigation of a potential ticity in adult mice. Frontiers in Cellular Neuroscience 8, 326. https://doi.org/10.3389/
link between PANX1 and maternal immune activation. fncel.2014.00326.
Ayata, C., & Lauritzen, M. (2015). Spreading depression, spreading depolarizations, and
the cerebral vasculature. Physiological Reviews 95(3), 953–993. https://doi.org/10.
4. Future directions 1152/physrev.00027.2014.
Bachiller, S., Jiménez-Ferrer, I., Paulus, A., Yang, Y., Swanberg, M., Deierborg, T., & Boza-
As an ATP-release channel, it is not surprising that PANX1 partici- Serrano, A. (2018). Microglia in neurological diseases: A road map to brain-disease
dependent-inflammatory response. Frontiers in Cellular Neuroscience 12, 488.
pates in myriad physiological and pathophysiological nervous system
https://doi.org/10.3389/fncel.2018.00488.
contexts. The recent high-resolution cryo-EM characterization of Badimon, A., Strasburger, H. J., Ayata, P., Chen, X., Nair, A., Ikegami, A., ... Schaefer, A.
PANX1 structure (BOX1; Deng et al., 2020; Jin et al., 2020; Michalski (2020). Negative feedback control of neuronal activity by microglia. Nature 586
et al., 2020; Mou et al., 2020; R. Qu et al., 2020; Ruan, Orozco, Du, & (7829), 417–423. https://doi.org/10.1038/s41586-020-2777-8.
Bao, B. A., Lai, C. P., Naus, C. C., & Morgan, J. R. (2012). Pannexin1 drives multicellular ag-
Lü, 2020) will undoubtedly facilitate the design of PANX1-targeting gregate compaction via a signaling Cascade that remodels the actin cytoskeleton. The
molecules with enhanced selectivity and specificity. Development of Journal of Biological Chemistry 287(11), 8407–8416. https://doi.org/10.1074/jbc.
improved PANX1 blockers will provide new options for treating chal- M111.306522.
Bao, L., Locovei, S., & Dahl, G. (2004). Pannexin membrane channels are mechanosensitive
lenging neurological disorders such as neurodegeneration, conduits for ATP. FEBS Letters 572(1–3), 65–68. https://doi.org/10.1016/j.febslet.2004.
neurodevelopmental disorders, stroke, and pain. Moreover, our grow- 07.009.
ing appreciation of the contribution of PANX1 and other purinergic sig- Bargiotas, P., Krenz, A., Hormuzdi, S. G., Ridder, D. A., Herb, A., Barakat, W., ... Schwaninger,
M. (2011). Pannexins in ischemia-induced neurodegeneration. Proceedings of the
naling systems to nervous system cellular development throughout the
National Academy of Sciences of the United States of America 108(51), 20772–20777.
lifespan, under both healthy and pathological conditions, will further https://doi.org/10.1073/pnas.1018262108.
serve to enable clinical translation related to PANX1 and purinergic sig- Beckel, J. M., Argall, A. J., Lim, J. C., Xia, J., Lu, W., Coffey, E. E., ... Mitchell, C. H. (2014).
naling. Improved, in-depth understanding of PANX1 and purinergic sig- Mechanosensitive release of adenosine 5′-triphosphate through pannexin channels
and mechanosensitive upregulation of pannexin channels in optic nerve head astro-
naling in nervous system cellular functions will also be facilitated by cytes: A mechanism for purinergic involvement in chronic strain. GLIA 62(9),
transgenic mouse lines that selectively target neurons, interneurons, 1486–1501. https://doi.org/10.1002/glia.22695.

11
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

Berry, K. P., & Nedivi, E. (2017). Spine dynamics: Are they all the same? Neuron 96(1), and membrane permeability during apoptosis. Nature 467(7317), 863–867 [pii]
43–55. https://doi.org/10.1016/j.neuron.2017.08.008. https://doi.org/10.1038/nature09413.
Bialecki, J., Werner, A., Weilinger, N. L., Tucker, C. M., Vecchiarelli, H. A., Egaña, J., ... Chen, Y., Corriden, R., Inoue, Y., Yip, L., Hashiguchi, N., Zinkernagel, A., ... Junger, W. G.
Thompson, R. J. (2020). Suppression of presynaptic glutamate release by postsynaptic (2006). ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors.
metabotropic NMDA receptor Signalling to Pannexin-1. The Journal of neuroscience: Science 314(5806), 1792–1795. https://doi.org/10.1126/science.1132559.
the official journal of the Society for Neuroscience 40(4), 729–742. https://doi.org/10. Chiu, Y. H., Jin, X., Medina, C. B., Leonhardt, S. A., Kiessling, V., Bennett, B. C., ... Bayliss, D. A.
1523/JNEUROSCI.0257-19.2019. (2017). A quantized mechanism for activation of pannexin channels. Nature
Billaud, M., Chiu, Y. -H., Lohman, A. W., Parpaite, T., Butcher, J. T., Mutchler, S. M., ... Communications 8. https://doi.org/10.1038/ncomms14324.
Isakson, B. E. (2015). A molecular signature in the pannexin1 intracellular loop con- Chiu, Y. -H., Schappe, M. S., Desai, B. N., & Bayliss, D. A. (2018). Revisiting multimodal ac-
fers channel activation by the α1 adrenoreceptor in smooth muscle cells. Science tivation and channel properties of Pannexin 1. The Journal of General Physiology 150
Signaling 8(364), Article ra17–ra17. https://doi.org/10.1126/scisignal.2005824. (1), 19–39. https://doi.org/10.1085/jgp.201711888.
Billaud, M., Lohman, A. W., Straub, A. C., Looft-Wilson, R., Johnstone, S. R., Araj, C. A., ... Chitnis, T., & Weiner, H. L. (2017). CNS inflammation and neurodegeneration. Journal of
Isakson, B. E. (2011). Pannexin1 regulates α1-adrenergic receptor- mediated vaso- Clinical Investigation 127(10), 3577–3587. https://doi.org/10.1172/JCI90609.
constriction. Circulation Research 109(1), 80–85. https://doi.org/10.1161/ Cieślak, M., & Wojtczak, A. (2018). Role of purinergic receptors in the Alzheimer’s disease.
CIRCRESAHA.110.237594. Purinergic Signalling 14(4), 331–344. https://doi.org/10.1007/s11302-018-9629-0.
Blay, J., White, T. D., & Hoskin, D. W. (1997). The extracellular fluid of solid carcinomas Cone, A. C., Ambrosi, C., Scemes, E., Martone, M. E., & Sosinsky, G. E. (2013). A comparative
contains immunosuppressive concentrations of adenosine. Cancer Research 57(13). antibody analysis of pannexin1 expression in four rat brain regions reveals varying
Blum, D., Chern, Y., Domenici, M. R., Buée, L., Lin, C. -Y., Rea, W., ... Popoli, P. (2018). The subcellular localizations. Frontiers in Pharmacology. https://doi.org/10.3389/fphar.
role of adenosine tone and adenosine receptors in Huntington’s disease. Journal of 2013.00006.
Caffeine and Adenosine Research 8(2), 43–58. https://doi.org/10.1089/caff.2018.0006. Couturier, C. P., Ayyadhury, S., Le, P. U., Nadaf, J., Monlong, J., Riva, G., ... Petrecca, K. (2020).
Blum, D., Galas, M. C., Pintor, A., Brouillet, E., Ledent, C., Muller, C. E., ... Schiffmann, S. N. Single-cell RNA-seq reveals that glioblastoma recapitulates a normal
(2003). A dual role of adenosine A2A receptors in 3-nitropropionic acid-induced neurodevelopmental hierarchy. Nature Communications 11(1), 3406. https://doi.org/
striatal lesions: Implications for the neuroprotective potential of A2A antagonists. 10.1038/s41467-020-17186-5.
Journal of Neuroscience 23(12), 5361–5369. https://doi.org/10.1523/jneurosci.23-12- Crespo Yanguas, S., Willebrords, J., Johnstone, S. R., Maes, M., Decrock, E., De Bock, M., ...
05361.2003. Vinken, M. (2017). Pannexin1 as mediator of inflammation and cell death.
Boassa, D., Nguyen, P., Hu, J., Ellisman, M. H., & Sosinsky, G. E. (2015). Pannexin2 oligo- Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 1864(1), 51–61.
mers localize in the membranes of endosomal vesicles in mammalian cells while https://doi.org/10.1016/j.bbamcr.2016.10.006.
Pannexin1 channels traffic to the plasma membrane. Frontiers in Cellular
Dahl, G., Qiu, F., & Wang, J. (2013). The bizarre pharmacology of the ATP release channel
Neuroscience 8(FEB), 468. https://doi.org/10.3389/fncel.2014.00468.
pannexin1. Neuropharmacology 75, 583–593. https://doi.org/10.1016/j.neuropharm.
Borovac, J., Bosch, M., & Okamoto, K. (2018). Regulation of actin dynamics during struc- 2013.02.019.
tural plasticity of dendritic spines: Signaling messengers and actin-binding proteins.
Davis, L. K., Gamazon, E. R., Kistner-Griffin, E., Badner, J. A., Liu, C., Cook, E. H., ... Cox, N. J.
Molecular and Cellular Neuroscience 91, 122–130. https://doi.org/10.1016/j.mcn.2018.
(2012). Loci nominally associated with autism from genome-wide analysis show en-
07.001.
richment of brain expression quantitative trait loci but not lymphoblastoid cell line
Boyce, A. K., van der Slagt, E., Sanchez-Arias, J. C., Anne Swayne, L., Boyce-andrewboyce, A.
expression quantitative trait loci. Molecular Autism 3(1), 3. https://doi.org/10.1186/
K., & Leigh Anne, U. (2020). ATP triggers macropinocytosis that internalizes and is
2040-2392-3-3.
regulated by PANX1. BioRxiv, Article 2020.11.19.389072. https://doi.org/10.1101/
De Bock, M., Wang, N., Bol, M., Decrock, E., Ponsaerts, R., Bultynck, G., ... Leybaert, L.
2020.11.19.389072.
(2012). Connexin-43 hemichannels contribute to cytoplasmic Ca2+ oscillations by
Boyce, A. K. J., Epp, A. L., Nagarajan, A., & Swayne, L. A. (2018). Transcriptional and post-
providing a bimodal Ca2+−dependent Ca2+−entry pathway. The Journal of
translational regulation of pannexins. Biochimica et Biophysica Acta (BBA) -
Biological Chemistry.. https://doi.org/10.1074/jbc.M111.299610.
Biomembranes 1860(1), 72–82. https://doi.org/10.1016/j.bbamem.2017.03.004.
Decourt, B., Lahiri, D. K., & Sabbagh, M. N. (2016). Targeting tumor necrosis factor alpha
Boyce, A. K. J., Kim, M. S., Wicki-Stordeur, L. E., & Swayne, L. A. (2015). ATP stimulates
for Alzheimer’s disease. Current Alzheimer Research 14(4), 412–425. https://doi.org/
pannexin 1 internalization to endosomal compartments. The Biochemical Journal
10.2174/1567205013666160930110551.
470(3), 319–330. https://doi.org/10.1042/BJ20141551.
Deng, Z., He, Z., Maksaev, G., Bitter, R. M., Rau, M., Fitzpatrick, J. A. J., & Yuan, P. (2020).
Boyce, A. K. J., & Swayne, L. A. (2017). P2X7 receptor cross-talk regulates ATP-induced
Cryo-EM structures of the ATP release channel pannexin 1. Nature Structural &
pannexin 1 internalization. Biochemical Journal 474(13), 2133–2144. https://doi.org/
Molecular Biology 27(4), 373–381. https://doi.org/10.1038/s41594-020-0401-0.
10.1042/BCJ20170257.
DeLalio, L. J., Billaud, M., Ruddiman, C. A., Johnstone, S. R., Butcher, J. T., Wolpe, A. G., ...
Bravo, D., Ibarra, P., Retamal, J., Pelissier, T., Laurido, C., Hernandez, A., & Constandil, L.
Isakson, B. E. (2019). Constitutive SRC-mediated phosphorylation of pannexin 1 at ty-
(2014). Pannexin 1: A novel participant in neuropathic pain signaling in the rat spinal
rosine 198 occurs at the plasma membrane. Journal of Biological Chemistry 294(17),
cord. Pain 155(10), 2108–2115. https://doi.org/10.1016/j.pain.2014.07.024.
6940–6956. https://doi.org/10.1074/jbc.RA118.006982.
Bravo, D., Maturana, C. J., Pelissier, T., Hernández, A., & Constandil, L. (2015). Interactions
of pannexin 1 with NMDA and P2X7 receptors in central nervous system pathologies: Dent, E. W. (2017). Of microtubules and memory: Implications for microtubule dynamics
Possible role on chronic pain. Pharmacological Research 101, 86–93. https://doi.org/ in dendrites and spines. Molecular Biology of the Cell 28(1), 1–8. https://doi.org/10.
10.1016/j.phrs.2015.07.016. 1091/mbc.e15-11-0769.
Brocardo, L., Acosta, L. E., Piantanida, A. P., & Rela, L. (2019). Beneficial and detrimental re- Di Virgilio, F., & Adinolfi, E. (2017). Extracellular purines, purinergic receptors and tumor
modeling of glial Connexin and Pannexin functions in rodent models of nervous sys- growth. Oncogene 36(3), 293–303. https://doi.org/10.1038/onc.2016.206.
tem diseases. Frontiers in Cellular Neuroscience 13. https://doi.org/10.3389/fncel.2019. Di Virgilio, F., Dal Ben, D., Sarti, A. C., Giuliani, A. L., & Falzoni, S. (2017). The P2X7 receptor
00491. in infection and inflammation. Immunity 47(1), 15–31. https://doi.org/10.1016/j.
Brown, A. S., & Conway, F. (2019). Maternal immune activation and related factors in the immuni.2017.06.020.
risk of offspring psychiatric disorders. Frontiers in Psychiatry 10(MAY), 1–6. https:// Di Virgilio, F., Sarti, A. C., Falzoni, S., De Marchi, E., & Adinolfi, E. (2018). Extracellular ATP
doi.org/10.3389/fpsyt.2019.00430. and P2 purinergic signalling in the tumour microenvironment. Nature Reviews Cancer
Brown, G., Vilalta, A., & Fricker, M. (2015). Phagoptosis - cell death by phagocytosis - plays 18(10), 601–618. https://doi.org/10.1038/s41568-018-0037-0.
central roles in physiology, host defense and pathology. Current Molecular Medicine Díaz, E. F., Labra, V. C., Alvear, T. F., Mellado, L. A., Inostroza, C. A., Oyarzún, J. E., ... Orellana,
15(9), 842–851. https://doi.org/10.2174/156652401509151105130628. J. A. (2019). Connexin 43 hemichannels and pannexin‐1 channels contribute to the
Brown, G. C., & Neher, J. J. (2012). Eaten alive! Cell death by primary phagocytosis: α‐synuclein‐induced dysfunction and death of astrocytes. Glia 67(8). https://doi.
“Phagoptosis”. Trends in Biochemical Sciences 37(8), 325–332. https://doi.org/10. org/10.1002/glia.23631 glia.23631.
1016/j.tibs.2012.05.002. Dissing-Olesen, L., LeDue, J. M., Rungta, R. L., Hefendehl, J. K., Choi, H. B., & MacVicar, B. A.
Burma, N. E., Bonin, R. P., Leduc-Pessah, H., Baimel, C., Cairncross, Z. F., Mousseau, M., ... (2014). Activation of neuronal NMDA receptors triggers transient ATP-mediated
Trang, T. (2017). Blocking microglial pannexin-1 channels alleviates morphine with- microglial process outgrowth. Journal of Neuroscience 34(32), 10511–10527. https://
drawal in rodents. Nature Medicine 23(3), 355–360. https://doi.org/10.1038/nm.4281. doi.org/10.1523/JNEUROSCI.0405-14.2014.
Burns, A. R., Phillips, S. C., & Sokoya, E. M. (2012). Pannexin protein expression in the rat Domercq, M., Perez-Samartin, A., Aparicio, D., Alberdi, E., Pampliega, O., & Matute, C.
middle cerebral artery. Journal of Vascular Research 49(2), 101–110. https://doi.org/ (2010). P2X7 receptors mediate ischemic damage to oligodendrocytes. Glia 58(6),
10.1159/000332329. 730–740. https://doi.org/10.1002/glia.20958.
Burnstock, G. (2020). Introduction to purinergic signalling in the brain. Advances in Dossi, E., Blauwblomme, T., Moulard, J., Chever, O., Vasile, F., Guinard, E., ... Rouach, N.
Experimental Medicine and Biology 1202, 1–12. https://doi.org/10.1007/978-3-030- (2018). Pannexin-1 channels contribute to seizure generation in human epileptic
30651-9_1. brain tissue and in a mouse model of epilepsy. Science Translational Medicine 10
Castello, M. A., & Gleeson, J. G. (2021). Insight into developmental mechanisms of global (443), 3796. https://doi.org/10.1126/scitranslmed.aar3796.
and focal migration disorders of cortical development. Current Opinion in Dunwiddie, T. V., & Masino, S. A. (2001). The role and regulation of adenosine in the cen-
Neurobiology 66, 77–84. https://doi.org/10.1016/j.conb.2020.10.005. tral nervous system. Annual Review of Neuroscience 24(1), 31–55. https://doi.org/10.
Cavaliere, F., Donno, C., & D’Ambrosi, N. (2015). Purinergic signaling: A common pathway 1146/annurev.neuro.24.1.31.
for neural and mesenchymal stem cell maintenance and differentiation. Frontiers in Dvoriantchikova, G., Ivanov, D., Barakat, D., Grinberg, A., Wen, R., Slepak, V. Z., &
Cellular Neuroscience 9(June), 211. https://doi.org/10.3389/fncel.2015.00211. Shestopalov, V. I. (2012). Genetic ablation of Pannexin1 protects retinal neurons
Ceruti, S., & Abbracchio, M. P. (2020). Adenosine signaling in glioma cells. Advances in from ischemic injury. PLoS One 7(2), Article e31991. https://doi.org/10.1371/
Experimental Medicine and Biology 1202, 13–33. https://doi.org/10.1007/978-3-030- journal.pone.0031991.
30651-9_2. Dvoriantchikova, G., Ivanov, D., Panchin, Y., & Shestopalov, V. I. (2006). Expression of
Chekeni, F. B., Elliott, M. R., Sandilos, J. K., Walk, S. F., Kinchen, J. M., Lazarowski, E. R., ... pannexin family of proteins in the retina. FEBS Letters 580(9), 2178–2182. https://
Ravichandran, K. S. (2010). Pannexin 1 channels mediate “find-me” signal release doi.org/10.1016/j.febslet.2006.03.026.

12
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

Erb, L., Cao, C., Ajit, D., & Weisman, G. A. (2015). P2Y receptors in Alzheimer’s disease. Hanstein, R., Negoro, H., Patel, N. K., Charollais, A., Meda, P., Spray, D. C., ... Scemes, E.
Biology of the Cell 107(1), 1–21. https://doi.org/10.1111/boc.201400043. (2013). Promises and pitfalls of a Pannexin1 transgenic mouse line. Frontiers in
Eyo, U. B., Peng, J., Swiatkowski, P., Mukherjee, A., Bispo, A., & Wu, L. J. (2014). Neuronal Pharmacology. https://doi.org/10.3389/fphar.2013.00061 4 MAY.
hyperactivity recruits microglial processes via neuronal NMDA receptors and Harcha, P. A., Vargas, A., Yi, C., Koulakoff, A. A., Giaume, C., & Sáez, J. C. (2015). Hemichan-
microglial P2Y12 receptors after status epilepticus. Journal of Neuroscience 34(32), nels are required for amyloid β-peptide-induced degranulation and are activated in
10528–10540. https://doi.org/10.1523/JNEUROSCI.0416-14.2014. brain mast cells of APPswe/PS1dE9 Mice. Journal of Neuroscience 35(25),
Falzoni, S., Donvito, G., & Di Virgilio, F. (2013). Detecting adenosine triphosphate in the 9526–9538. https://doi.org/10.1523/JNEUROSCI.3686-14.2015.
pericellular space. Interface Focus 3(3), 20120101. https://doi.org/10.1098/rsfs.2012. Haynes, S. E., Hollopeter, G., Yang, G., Kurpius, D., Dailey, M. E., Gan, W. B., & Julius, D.
0101. (2006). The P2Y12 receptor regulates microglial activation by extracellular nucleo-
Fields, R. D., & Stevens, B. (2000). ATP: An extracellular signaling molecule between neu- tides. Nature Neuroscience 9(12), 1512–1519. https://doi.org/10.1038/nn1805.
rons and glia. Trends in Neurosciences 23(12), 625–633. https://doi.org/10.1016/ von Hehn, C. A., Baron, R., & Woolf, C. J. (2012). Deconstructing the neuropathic pain phe-
S0166-2236(00)01674-X. notype to reveal neural mechanisms. Neuron 73(4), 638–652. https://doi.org/10.
Flores-Muñoz, C., Gómez, B., Mery, E., Mujica, P., Gajardo, I., Córdova, C., ... Ardiles, Á. O. 1016/j.neuron.2012.02.008.
(2020). Acute Pannexin 1 blockade mitigates early synaptic plasticity defects in a Hendriksen, E., van Bergeijk, D., Oosting, R. S., & Redegeld, F. A. (2017). Mast cells in neu-
mouse model of Alzheimer’s disease. Frontiers in Cellular Neuroscience 14. https:// roinflammation and brain disorders. Neuroscience and Biobehavioral Reviews 79,
doi.org/10.3389/fncel.2020.00046. 119–133. https://doi.org/10.1016/j.neubiorev.2017.05.001.
Fontainhas, A. M., Wang, M., Liang, K. J., Chen, S., Mettu, P., Damani, M., ... Wong, W. T. Hisaoka-Nakashima, K., Azuma, H., Ishikawa, F., Nakamura, Y., Wang, D., Liu, K., ...
(2011). Microglial morphology and dynamic behavior is regulated by ionotropic glu- Morioka, N. (2020). Corticosterone induces HMGB1 release in primary cultured rat
tamatergic and GABAergic neurotransmission. PLoS One 6(1), Article e15973. https:// cortical astrocytes: Involvement of Pannexin-1 and P2X7 receptor-dependent mech-
doi.org/10.1371/journal.pone.0015973. anisms. Cells 9(5). https://doi.org/10.3390/cells9051068.
Forrest, M. P., Parnell, E., & Penzes, P. (2018). Dendritic structural plasticity and neuropsy- Hnasko, T. S., & Edwards, R. H. (2012). Neurotransmitter corelease: Mechanism and phys-
chiatric disease. Nature Reviews Neuroscience. https://doi.org/10.1038/nrn.2018.16. iological role. Annual Review of Physiology 74, 225–243. https://doi.org/10.1146/
Francistiová, L., Bianchi, C., Di Lauro, C., Sebastián-Serrano, Á., de Diego-García, L., Kobolák, annurev-physiol-020911-153315.
J., & Díaz-Hernández, M. (2020). The role of P2X7 receptor in Alzheimer’s disease. Holland, S. H. (2020). Regulation of neuroblastoma malignant properties by pannexin 1
Frontiers in Molecular Neuroscience 13, 94. https://doi.org/10.3389/fnmol.2020.00094. channels: Role of post-translational modifications and mutations [University of Ottawa]
Franke, H., & Illes, P. (2006). Involvement of P2 receptors in the growth and survival of PhD Thesis . https://doi.org/10.20381/ruor-24324.
neurons in the CNS. Pharmacology and Therapeutics. https://doi.org/10.1016/j. Horton, S. M., Luna Lopez, C., Blevins, E., Howarth, H., Weisberg, J., Shestopalov, V. I., ...
pharmthera.2005.06.002. Shah, S. B. (2017). Pannexin 1 modulates axonal growth in mouse peripheral nerves.
Frederiksen, S. D., Wicki-Stordeur, L. E., Sanchez-Arias, J. C., & Swayne, L. A. (2019). Ex- Frontiers in Cellular Neuroscience 11, 365. https://doi.org/10.3389/fncel.2017.00365.
ploring the Pannexin 1 interactome: In silico cross-analyses with postsynaptic pro- Horváth, G., Otrokocsi, L., Beko, K., Baranyi, M., Kittel, Á., Fritz-Ruenes, P. A., & Sperlágh, B.
teins and neuropsychiatric disorder susceptibility genes. BioRxiv 801563. https://doi. (2019). P2X7 receptors drive poly(I:C) induced autism-like behavior in mice. Journal
org/10.1101/801563. of Neuroscience 39(13), 2542–2561. https://doi.org/10.1523/JNEUROSCI.1895-18.
Fredholm, B. B., Chen, J. -F., Masino, S. A., & Vaugeois, J. -M. (2005). Actions of adenosine at 2019.
its receptors in the CNS: Insights from knockouts and drugs. Annual Review of Huang, Y., Grinspan, J. B., Abrams, C. K., & Scherer, S. S. (2007). Pannexin1 is expressed by
Pharmacology and Toxicology 45(1), 385–412. https://doi.org/10.1146/annurev. neurons and glia but does not form functional gap junctions. Glia 55(1), 46–56.
pharmtox.45.120403.095731.
Hui, K. P. Y., Cheung, M. -C., Perera, R. A. P. M., Ng, K. -C., Bui, C. H. T., Ho, J. C. W., ... Chan, M.
Freitas-Andrade, M., Bechberger, J. F., MacVicar, B. A., Viau, V., & Naus, C. C. (2017).
C. W. (2020). Tropism, replication competence, and innate immune responses of the
Pannexin 1 knockout and blockade reduces ischemic stroke injury in female but
coronavirus SARS-CoV-2 in human respiratory tract and conjunctiva: An analysis in
not in male mice. Oncotarget 8(23), 36973–36983. https://doi.org/10.18632/
ex-vivo and in-vitro cultures. The Lancet Respiratory Medicine 8(7), 687–695.
oncotarget.16937.
https://doi.org/10.1016/S2213-2600(20)30193-4.
Gajardo, I., Salazar, C. S., Lopez-Espíndola, D., Estay, C., Flores-Muñoz, C., Elgueta, C., ...
Hung, S. -C., Choi, C. H., Said-Sadier, N., Johnson, L., Atanasova, K. R., Sellami, H., ... Ojcius,
Ardiles, Á. O. (2018). Lack of Pannexin 1 alters synaptic GluN2 subunit composition
D. M. (2013). P2X4 assembles with P2X7 and Pannexin-1 in gingival epithelial cells
and spatial reversal learning in mice. Frontiers in Molecular Neuroscience 11, 114.
and modulates ATP-induced reactive oxygen species production and Inflammasome
https://doi.org/10.3389/fnmol.2018.00114.
activation. PLoS One 8(7), Article e70210. https://doi.org/10.1371/journal.pone.
Gajardo-Gómez, R., Santibañez, C. A., Labra, V. C., Gómez, G. I., Eugenin, E. A., & Orellana, J. 0070210.
A. (2020). HIV gp120 protein increases the function of Connexin 43 Hemichannels
Iglesias, R., Dahl, G., Qiu, F., Spray, D. C., & Scemes, E. (2009). Pannexin 1: The molecular
and Pannexin-1 channels in astrocytes: Repercussions on Astroglial function.
substrate of astrocyte “hemichannels”. The Journal of Neuroscience 29(21),
International Journal of Molecular Sciences 21(7), 2503. https://doi.org/10.3390/
7092–7097. https://doi.org/10.1523/JNEUROSCI.6062-08.2009.
ijms21072503.
Illes, P., Burnstock, G., & Tang, Y. (2019). Astroglia-derived ATP modulates CNS neuronal
Garg, C., Seo, J. H., Ramachandran, J., Loh, J. M., Calderon, F., & Contreras, J. E. (2018).
circuits. Trends in Neurosciences 42(12), 885–898. https://doi.org/10.1016/j.tins.
Trovafloxacin attenuates neuroinflammation and improves outcome after traumatic
2019.09.006.
brain injury in mice. Journal of Neuroinflammation 15(1), 1–15. https://doi.org/10.
1186/s12974-018-1069-9. Illes, P., Rubini, P., Ulrich, H., Zhao, Y., & Tang, Y. (2020). Regulation of microglial functions
by Purinergic mechanisms in the healthy and diseased CNS. 9. (pp. 1108). Cells, 1108.
Garré, J. M., Retamal, M. A., Cassina, P., Barbeito, L., Bukauskas, F. F., Sáez, J. C., ... Abudara,
V. (2010). FGF-1 induces ATP release from spinal astrocytes in culture and opens Illes, P., Xu, G. -Y., & Tang, Y. (2020). Purinergic signaling in the central nervous system in
pannexin and connexin hemichannels. Proceedings of the National Academy of health and disease. Neuroscience Bulletin 36(11), 1239–1241. https://doi.org/10.1007/
Sciences of the United States of America 107(52), 22659–22664. https://doi.org/10. s12264-020-00602-7.
1073/pnas.1013793107. Ito, U., Ohno, K., Nakamura, R., Suganuma, F., & Inaba, Y. (1979). Brain edema during is-
Gawlik, M., Wagner, M., Pfuhlmann, B., & Stöber, G. (2016). The role of Pannexin gene var- chemia and after restoration of blood flow. Measurement of water, sodium, potas-
iants in schizophrenia: Systematic analysis of phenotypes. European Archives of Psy- sium content and plasma protein permeability. Stroke 10(5), 542–547. https://doi.
chiatry and Clinical Neuroscience 266(5), 433–437. https://doi.org/10.1007/s00406- org/10.1161/01.str.10.5.542.
015-0619-8. Jackson, D. G., Wang, J., Keane, R. W., Scemes, E., & Dahl, G. (2014). ATP and potassium
Gessi, S., Merighi, S., Sacchetto, V., Simioni, C., & Borea, P. A. (2011). Adenosine receptors ions: A deadly combination for astrocytes. Scientific Reports 4(1), 1–9. https://doi.
and cancer. Biochimica et Biophysica Acta - Biomembranes 1808(5), 1400–1412. org/10.1038/srep04576.
https://doi.org/10.1016/j.bbamem.2010.09.020. Jankowski, J., Perry, H. M., Medina, C. B., Huang, L., Yao, J., Bajwa, A., ... Okusa, M. D. (2018).
Glass, M., Dragunow, M., & Faull, R. L. M. (2000). The pattern of neurodegeneration in Epithelial and endothelial Pannexin1 channels mediate AKI. Journal of the American
Huntington’s disease: A comparative study of cannabinoid, dopamine, adenosine Society of Nephrology 29(7), 1887–1899. https://doi.org/10.1681/ASN.2017121306.
and GABA(A) receptor alterations in the human basal ganglia in Huntington’s disease. Jayashankar, V., & Edinger, A. L. (2020). Macropinocytosis confers resistance to therapies
Neuroscience 97(3), 505–519. https://doi.org/10.1016/S0306-4522(00)00008-7. targeting cancer anabolism. Nature Communications 11(1), 1121. https://doi.org/10.
Good, M. E., Chiu, Y. H., Poon, I. K. H., Medina, C. B., Butcher, J. T., Mendu, S. K., ... 1038/s41467-020-14928-3.
Ravichandran, K. S. (2018). Pannexin 1 channels as an unexpected new target of Jeong, Y. H., Walsh, M. C., Yu, J., Shen, H., Wherry, E. J., & Choi, Y. (2020). Mice lacking the
the anti-hypertensive drug spironolactone. Circulation Research 122(4), 606–615. purinergic receptor P2X5 exhibit defective inflammasome activation and early sus-
https://doi.org/10.1161/CIRCRESAHA.117.312380. ceptibility to Listeria monocytogenes. The Journal of Immunology, Article ji1901423.
Good, M. E., Eucker, S. A., Li, J., Bacon, H. M., Lang, S. M., Butcher, J. T., ... Isakson, B. E. https://doi.org/10.4049/jimmunol.1901423.
(2018). Endothelial cell Pannexin1 modulates severity of ischemic stroke by regulat- Jessa, S., Blanchet-Cohen, A., Krug, B., Vladoiu, M., Coutelier, M., Faury, D., ... Kleinman, C. L.
ing cerebral inflammation and myogenic tone. JCI Insight 3(6). https://doi.org/10. (2019). Stalled developmental programs at the root of pediatric brain tumors. Nature
1172/jci.insight.96272. Genetics 51(12), 1702–1713. https://doi.org/10.1038/s41588-019-0531-7.
Grassi, F. (2010). Purinergic control of neutrophil activation. Journal of Molecular Cell Jian, Z., Ding, S., Deng, H., Wang, J., Yi, W., Wang, L., ... Xiong, X. (2016). Probenecid pro-
Biology 2(4), 176–177. https://doi.org/10.1093/jmcb/mjq014. tects against oxygen–glucose deprivation injury in primary astrocytes by regulating
Guzman, S. J., & Gerevich, Z. (2016). P2Y receptors in synaptic transmission and plasticity: inflammasome activity. Brain Research 1643, 123–129. https://doi.org/10.1016/j.
Therapeutic potential in cognitive dysfunction. Neural Plasticity 2016. https://doi.org/ brainres.2016.05.002.
10.1155/2016/1207393 Hindawi Limited. Jiang, T., Xu, R. X., Zhang, A. W., Di, W., Xiao, Z. J., Miao, J. Y., ... Fang, Y. N. (2012). Effects of
Hamann, M., & Attwell, D. (1996). Non-synaptic release of ATP by electrical stimulation in transcranial direct current stimulation on hemichannel pannexin-1 and neural plas-
slices of rat hippocampus, cerebellum and Habenula. European Journal of Neuroscience ticity in rat model of cerebral infarction. Neuroscience 226, 421–426. https://doi.org/
8(7), 1510–1515. https://doi.org/10.1111/j.1460-9568.1996.tb01613.x. 10.1016/j.neuroscience.2012.09.035.

13
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

Jin, Q., Zhang, B., Zheng, X., Li, N., Xu, L., Xie, Y., ... Ye, S. (2020). Cryo-EM structures of Malik, S., & Eugenin, E. A. (2019). Role of Connexin and Pannexin containing channels in
human pannexin 1 channel. Cell Research 30(5), 449–451. https://doi.org/10.1038/ HIV infection and NeuroAIDS. Neuroscience Letters 695, 86–90. https://doi.org/10.
s41422-020-0310-0. 1016/j.neulet.2017.09.005.
Jo, S., & Bean, B. P. (2011). Inhibition of neuronal voltage-gated sodium channels by Bril- Mangiafico, R. A., Sarnataro, F., Mangiafico, M., & Fiore, C. E. (2006). Impaired cognitive
liant blue G. Molecular Pharmacology 80(2), 247–257. https://doi.org/10.1124/mol. performance in asymptomatic peripheral arterial disease: Relation to C-reactive pro-
110.070276. tein and D-dimer levels. Age and Ageing 35(1), 60–65. https://doi.org/10.1093/ageing/
Jurkowski, M. P., Bettio, L., Woo, E. K., Patten, A., Yau, S. -Y., & Gil-Mohapel, J. (2020). Be- afi219.
yond the hippocampus and the SVZ: Adult neurogenesis throughout the brain. Martín, A., Domercq, M., & Matute, C. (2018). Inflammation in stroke: The role of cholin-
Frontiers in Cellular Neuroscience 14. https://doi.org/10.3389/fncel.2020.576444. ergic, purinergic and glutamatergic signaling. Therapeutic Advances in Neurological
Kaneko, Y., Tachikawa, M., Akaogi, R., Fujimoto, K., Ishibashi, M., Uchida, Y., ... Terasaki, T. Disorders 11. https://doi.org/10.1177/1756286418774267 SAGE Publications Ltd.
(2015). Contribution of pannexin 1 and connexin 43 hemichannels to extracellular Martin, E., Amar, M., Dalle, C., Youssef, I., Boucher, C., Le Duigou, C., ... Delarasse, C. (2019).
calcium-dependent transport dynamics in human blood-brain barrier endothelial New role of P2X7 receptor in an Alzheimer’s disease mouse model. Molecular
cells. Journal of Pharmacology and Experimental Therapeutics 353(1), 192–200. Psychiatry 24(1), 108–125. https://doi.org/10.1038/s41380-018-0108-3.
https://doi.org/10.1124/jpet.114.220210. Medina, C. B., Mehrotra, P., Arandjelovic, S., Perry, J. S. A., Guo, Y., Morioka, S., ...
Khakh, B. S., & Alan North, R. (2006). P2X receptors as cell-surface ATP sensors in health Ravichandran, K. S. (2020). Metabolites released from apoptotic cells act as tissue
and disease. Nature 442(7102), 527–532 Nature Publishing Group https://doi.org/10. messengers. Nature 580(7801), 130–135. https://doi.org/10.1038/s41586-020-
1038/nature04886. 2121-3.
Kovács, G., Környei, Z., Tóth, K., Baranyi, M., Brunner, J., Neubrandt, M., ... Sperlágh, B. Michalski, K., & Kawate, T. (2016). Carbenoxolone inhibits Pannexin1 channels through
(2018). Modulation of P2X7 purinergic receptor activity by extracellular Zn 2+ in interactions in the first extracellular loop. Journal of General Physiology 147(2),
cultured mouse hippocampal astroglia. Cell Calcium 75, 1–13. https://doi.org/10. 165–174. https://doi.org/10.1085/jgp.201511505.
1016/j.ceca.2018.07.010. Michalski, K., Syrjanen, J. L., Henze, E., Kumpf, J., Furukawa, H., & Kawate, T. (2020). The
Kovalzon, V. M., Moiseenko, L. S., Ambaryan, A. V., Kurtenbach, S., Shestopalov, V. I., & Cryo-EM structure of pannexin 1 reveals unique motifs for ion selection and inhibi-
Panchin, Y. V. (2017). Sleep-wakefulness cycle and behavior in pannexin1 knockout tion. ELife 9, Article e54670. https://doi.org/10.7554/eLife.54670.
mice. Behavioural Brain Research 318, 24–27. https://doi.org/10.1016/j.bbr.2016.10. Miras-Portugal, M. T., Sebastián-Serrano, Á., De Diego García, L., & Díaz-Hernández, M.
015. (2017). Neuronal P2X7 receptor: Involvement in neuronal physiology and. Journal
Kyrargyri, V., Madry, C., Rifat, A., Arancibia-Carcamo, I. L., Jones, S. P., Chan, V. T. T., ... of Neuroscience 37(30), 7063–7072. https://doi.org/10.1523/JNEUROSCI.3104-16.
Attwell, D. (2020). P2Y 13 receptors regulate microglial morphology, surveillance, 2017.
and resting levels of interleukin 1β release. Glia 68(2), 328–344. https://doi.org/10. Molica, F., Morel, S., Meens, M. J., Denis, J. F., Bradfield, P. F., Penuela, S., ... Kwak, B. R.
1002/glia.23719. (2015). Functional role of a polymorphism in the pannexin1 gene in collageninduced
Lagos-Cabré, R., Alvarez, A., Kong, M., Burgos-Bravo, F., Cárdenas, A., Rojas-Mancilla, E., ... platelet aggregation. Thrombosis and Haemostasis 114(2), 325–336. https://doi.org/
Leyton, L. (2017). αVβ3 integrin regulates astrocyte reactivity. Journal of 10.1160/TH14-11-0981.
Neuroinflammation 14(1), 194. https://doi.org/10.1186/s12974-017-0968-5. Mou, L., Ke, M., Song, M., Shan, Y., Xiao, Q., Liu, Q., ... Deng, D. (2020). Structural basis for
Lai, C. P., Bechberger, J. F., Thompson, R. J., MacVicar, B. A., Bruzzone, R., & Naus, C. C. gating mechanism of Pannexin 1 channel. Cell Research 30(5), 452–454. https://doi.
(2007). Tumor-suppressive effects of pannexin 1 in C6 glioma cells. Cancer Research org/10.1038/s41422-020-0313-x.
67(4), 1545–1554. Mousseau, M., Burma, N. E., Lee, K. Y., Leduc-Pessah, H., Kwok, C. H. T., Reid, A. R., ... Trang,
T. (2018). Microglial pannexin-1 channel activation is a spinal determinant of joint
Lalo, U., Palygin, O., Rasooli-Nejad, S., Andrew, J., Haydon, P. G., & Pankratov, Y. (2014).
pain. Science Advances 4(8), Article eaas9846. https://doi.org/10.1126/sciadv.aas9846.
Exocytosis of ATP from astrocytes modulates phasic and tonic inhibition in the neo-
cortex. PLoS Biology 12(1). https://doi.org/10.1371/journal.pbio.1001747. Narahari, A. K., Kreutzberger, A. J., Gaete, P. S., Chiu, Y. -H., Leonhardt, S. A., Medina, C. B., ...
Bayliss, D. A. (2021). ATP and large signaling metabolites flux through caspase-
Lalo, U., Palygin, O., Verkhratsky, A., Grant, S. G. N., & Pankratov, Y. (2016). ATP from syn-
activated Pannexin 1 channels. ELife 10. https://doi.org/10.7554/elife.64787.
aptic terminals and astrocytes regulates NMDA receptors and synaptic plasticity
Naviaux, R. K., Zolkipli, Z., Wang, L., Nakayama, T., Naviaux, J. C., Le, T. P., ... Powell, S. B.
through PSD-95 multi-protein complex. Scientific Reports 6(1), 1–20. https://doi.
(2013). Antipurinergic therapy corrects the autism-like features in the poly(IC)
org/10.1038/srep33609.
mouse model. PLoS One 8(3), Article e57380. https://doi.org/10.1371/journal.pone.
Lambrecht, G., Friebe, T., Grimm, U., Windscheif, U., Bungardt, E., Hildebrandt, C., ...
0057380.
Mutschler, E. (1992). PPADS, a novel functionally selective antagonist of P2
Navis, K. E., Fan, C. Y., Trang, T., Thompson, R. J., & Derksen, D. J. (2020). Pannexin 1 chan-
purinoceptor-mediated responses. European Journal of Pharmacology 217(2–3),
nels as a therapeutic target: structure, inhibition, and outlook. ACS Chemical
217–219.
Neuroscience 11(15), 2163–2172. https://doi.org/10.1021/acschemneuro.0c00333.
Li, S., Zang, Z., He, J., Chen, X., Yu, S., Pei, Y., ... Liu, S. (2017). Expression of pannexin 1 and 2
Nishiyama, J. (2019). Plasticity of dendritic spines: Molecular function and dysfunction in
in cortical lesions from intractable epilepsy patients with focal cortical dysplasia.
neurodevelopmental disorders. Psychiatry and Clinical Neurosciences 73(9), 541–550.
Oncotarget 8(4), 6883–6895. https://doi.org/10.18632/oncotarget.14317.
https://doi.org/10.1111/pcn.12899.
Li, Y. -C., Bai, W. -Z., Hashikawa, T., & Yan-Chao Li, C. (2020). The neuroinvasive potential
Nouri-Nejad, D., O’Donnell, B. L., Patil, C. S., Sanchez-Pupo, R. E., Johnston, D.,
of SARS-CoV2 may play a role in the respiratory failure of COVID-19 patients. Journal
Sayedyahossein, S., ... Penuela, S. (2021). Pannexin 1 mutation found in melanoma tu-
of Medical Virology. https://doi.org/10.1002/jmv.25728.
mour reduces phosphorylation, glycosylation, and trafficking of the channel-forming
Linden, J., Koch-Nolte, F., & Dahl, G. (2019). Purine release, metabolism, and signaling in protein. Molecular Biology of the Cell, Article mbc.E19-10-0585. https://doi.org/10.
the inflammatory response. Annual Review of Immunology 37, 325–347. https://doi. 1091/mbc.e19-10-0585.
org/10.1146/annurev-immunol-051116-052406.
Ohta, A., Gorelik, E., Prasad, S. J., Ronchese, F., Lukashev, D., Wong, M. K. K., ... Sitkovsky, M.
Lohman, A. W., & Isakson, B. E. (2014). Differentiating connexin hemichannels and (2006). A2A adenosine receptor protects tumors from antitumor T cells. Proceedings
pannexin channels in cellular ATP release. FEBS Letters 588(8), 1379–1388. https:// of the National Academy of Sciences of the United States of America 103(35),
doi.org/10.1016/j.febslet.2014.02.004. 13132–13137. https://doi.org/10.1073/pnas.0605251103.
Lohman, A. W., Leskov, I. L., Butcher, J. T., Johnstone, S. R., Stokes, T. A., Begandt, D., ... Oliveira, Á., Illes, P., & Ulrich, H. (2016). Purinergic receptors in embryonic and adult
Isakson, B. E. (2015). Pannexin 1 channels regulate leukocyte emigration through neurogenesis. Neuropharmacology 104, 272–281. https://doi.org/10.1016/j.
the venous endothelium during acute inflammation. Nature Communications 6. neuropharm.2015.10.008.
https://doi.org/10.1038/ncomms8965. Oliveira-Giacomelli, Á., Naaldijk, Y., Sardá-Arroyo, L., Gonçalves, M. C. B., Corrêa-Velloso, J.,
López, X., Escamilla, R., Fernández, P., Duarte, Y., González-Nilo, F., Palacios-Prado, N., ... Pillat, M. M., ... Ulrich, H. (2018). Purinergic receptors in neurological diseases with
Sáez, J. C. (2020). Stretch-induced activation of pannexin 1 channels can be motor symptoms: Targets for therapy. Frontiers in Pharmacology 9(APR), 325.
prevented by PKA-dependent phosphorylation. International Journal of Molecular https://doi.org/10.3389/fphar.2018.00325.
Sciences 21(23), 9180. https://doi.org/10.3390/ijms21239180. Orellana, J. A., Froger, N., Ezan, P., Jiang, J. X., Bennett, M. V. L., Naus, C. C., ... Sáez, J. C.
Lovelace, M. D., Gu, B. J., Eamegdool, S. S., Weible, M. W., Wiley, J. S., Allen, D. G., & Chan- (2011). ATP and glutamate released via astroglial connexin 43 hemichannels mediate
Ling, T. (2015). P2X7 receptors mediate innate phagocytosis by human neural pre- neuronal death through activation of pannexin 1 hemichannels. Journal of
cursor cells and neuroblasts. Stem Cells 33(2), 526–541. https://doi.org/10.1002/ Neurochemistry 118(5), 826–840. https://doi.org/10.1111/j.1471-4159.2011.07210.x.
stem.1864. Orellana, J. A., Shoji, K. F., Abudara, V., Ezan, P., Amigou, E., Saez, P. J., & Giaume, C. (2011).
Luo, Z., Wang, J., Tang, S., Zheng, Y., Zhou, X., Tian, F., & Xu, Z. (2020). Dynamic-related Amyloid beta-induced death in neurons involves glial and neuronal hemichannels.
protein 1 inhibitor eases epileptic seizures and can regulate equilibrative nucleoside The Journal of Neuroscience 31(13), 4962–4977. https://doi.org/10.1523/JNEUROSCI.
transporter 1 expression. BMC Neurology 20(1). https://doi.org/10.1186/s12883-020- 6417-10.2011.
01921-y. Orellana, J. A., Velasquez, S., Williams, D. W., Sáez, J. C., Berman, J. W., & Eugenin, E. A.
Ma, W., Compan, V., Zheng, W., Martin, E., North, R. A., Verkhratsky, A., & Surprenant, A. (2013). Pannexin1 hemichannels are critical for HIV infection of human primary
(2012). Pannexin 1 forms an anion-selective channel. Pflugers Archiv European Journal CD4 + T lymphocytes. Journal of Leukocyte Biology 94(3), 399–407. https://doi.org/
of Physiology 463(4), 585–592. https://doi.org/10.1007/s00424-012-1077-z. 10.1189/jlb.0512249.
Madry, C., Kyrargyri, V., Arancibia-Cárcamo, I. L., Jolivet, R., Kohsaka, S., Bryan, R. M., & Orellana, J. A., Moraga-Amaro, R., DÃaz-Galarce, R., Rojas, S., Maturana, C. J., Stehberg, J., &
Attwell, D. (2018). Microglial ramification, surveillance, and interleukin-1β release Sáez, J. C. (2015). Restraint stress increases hemichannel activity in hippocampal glial
are regulated by the two-pore domain K+ channel THIK-1. Neuron 97(2), 299–312. cells and neurons. Frontiers in Cellular Neuroscience 9(APR), 1–12. https://doi.org/10.
e6. https://doi.org/10.1016/j.neuron.2017.12.002. 3389/fncel.2015.00102.
Makarenkova, H. P., Shah, S. B., & Shestopalov, V. I. (2018). The two faces of pannexins: Pan, H. C., Chou, Y. C., & Sun, S. H. (2015). P2X7R-mediated Ca2+-independent d-serine
New roles in inflammation and repair. Journal of Inflammation Research 11, release via pannexin-1 of the P2X7R-pannexin-1 complex in astrocytes. GLIA 63(5),
273–288. https://doi.org/10.2147/JIR.S128401. 877–893. https://doi.org/10.1002/glia.22790.

14
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

Pankratov, Y., Lalo, U., Krishtal, O. A., & Verkhratsky, A. (2009). P2X receptors and synaptic Ruan, Z., Orozco, I. J., Du, J., & Lü, W. (2020). Structures of human pannexin 1 reveal ion
plasticity. Neuroscience 158(1), 137–148. https://doi.org/10.1016/j.neuroscience. pathways and mechanism of gating. Nature, 1–6. https://doi.org/10.1038/s41586-
2008.03.076. 020-2357-y.
Pankratov, Y., Lalo, U., Verkhratsky, A., & North, R. A. (2006). Vesicular release of ATP at Sandilos, J. K., Chiu, Y. H., Chekeni, F. B., Armstrong, A. J., Walk, S. F., Ravichandran, K. S., &
central synapses. Pflügers Archiv/European Journal of Physiology 452(5), 589–597. Bayliss, D. A. (2012). Pannexin 1, an ATP release channel, is activated by caspase
https://doi.org/10.1007/s00424-006-0061-x. cleavage of its pore-associated C terminal autoinhibitory region. J Biol Chem..
Parkinson, F. E., Damaraju, V. L., Graham, K., Yao, S. Y. M., Baldwin, S. A., Cass, C. E., & https://doi.org/10.1074/jbc.M111.323378.
Young, J. D. (2011). Molecular biology of nucleoside transporters and their distribu- Sáez, P. J., Shoji, K. F., Retamal, M. A., Harcha, P. A., Ramírez, G., Jiang, J. X., ... Sáez, J. C.
tions and functions in the brain. Current Topics in Medicinal Chemistry 11(8), (2013). ATP is required and advances cytokine-induced gap junction formation in
948–972. https://doi.org/10.2174/156802611795347582. microglia in vitro. Mediators of Inflammation 2013, 16. https://doi.org/10.1155/2013/
Pascual, O., Casper, K. B., Kubera, C., Zhang, J., Revilla-Sanchez, R., Sul, J. Y., ... Haydon, P. G. 216402.
(2005). Neurobiology: Astrocytic purinergic signaling coordinates synaptic networks. Safarian, N., Whyte-Fagundes, P., Zoidl, C., Grigull, J., & Zoidl, G. (2020). Visuomotor defi-
Science 310(5745), 113–116. https://doi.org/10.1126/science.1116916. ciency in panx1a knockout zebrafish is linked to dopaminergic signaling. Scientific
Patel, D., Zhang, X., & Veenstra, R. D. (2014). Connexin hemichannel and pannexin chan- Reports 10(1). https://doi.org/10.1038/s41598-020-66378-y.
nel electrophysiology: How do they differ? FEBS Letters 588(8), 1372–1378. https:// Sanchez-Arias, J. C., Candlish, R. C., van der Slagt, E., & Swayne, L. A. (2020). Pannexin 1
doi.org/10.1016/j.febslet.2013.12.023. regulates dendritic protrusion dynamics in immature cortical neurons. Eneuro, Article
Pelegrin, P., & Surprenant, A. (2006). Pannexin-1 mediates large pore formation and ENEURO.0079-20.2020. https://doi.org/10.1523/ENEURO.0079-20.2020.
interleukin-1β release by the ATP-gated P2X7 receptor. The EMBO Journal 25(21), Sanchez Arias, J. C., Wicki-Stordeur, L. E., Candlish, R. C., van der Slagt, E., Paci, I., Rao, P. P.
5071–5082. https://doi.org/10.1038/sj.emboj.7601378. N., ... Swayne, L. A. (2020). PANX1 in inflammation heats up: New mechanistic in-
Pellegatti, P., Raffaghello, L., Bianchi, G., Piccardi, F., Pistoia, V., & Di Virgilio, F. (2008). In- sights with implications for injury and infection. Cell Calcium 90, 102253. https://
creased level of extracellular ATP at tumor sites: In vivo imaging with plasma mem- doi.org/10.1016/j.ceca.2020.102253.
brane luciferase. PLoS One 3(7), Article e2599. https://doi.org/10.1371/journal.pone. Sanchez-Arias, J. C., Liu, M., Choi, C. S. W., Ebert, S. N., Brown, C. E., & Swayne, L. A. (2019).
0002599. Pannexin 1 regulates network ensembles and dendritic spine development in cortical
Penuela, S., Bhalla, R., Gong, X. -Q., Cowan, K. N., Celetti, S. J., Cowan, B. J., ... Laird, D. W. neurons. ENeuro 6(3). https://doi.org/10.1523/ENEURO.0503-18.2019.
(2007). Pannexin 1 and pannexin 3 are glycoproteins that exhibit many distinct char- Sandilos, J. K., Chiu, Y. -H., Chekeni, F. B., Armstrong, A. J., Walk, S. F., Ravichandran, K. S., &
acteristics from the connexin family of gap junction proteins. J Cell Sci 120(21), Bayliss, D. A. (2012). Pannexin 1, an ATP Release Channel, is activated by caspase
3772–3783. https://doi.org/10.1242/jcs.009514. cleavage of its pore-associated C-terminal autoinhibitory region. Journal of Biological
Penuela, S., Gehi, R., & Laird, D. W. (2013). The biochemistry and function of pannexin Chemistry 287(14), 11303–11311. https://doi.org/10.1074/jbc.M111.323378.
channels. Biochimica et Biophysica Acta - Biomembranes 1828(1), 15–22. https://doi. Sang, Q., Zhang, Z., Shi, J., Sun, X., Li, B., Yan, Z., Xue, S., Ai, A., Lyu, Q., Li, W., Zhang, J., Wu, L.,
org/10.1016/j.bbamem.2012.01.017. Mao, X., Chen, B., Mu, J., Li, Q., Du, J., Sun, Q., Jin, L., ... Wang, L. (2019). A pannexin 1
Penzes, P., Cahill, M. E., Jones, K. A., VanLeeuwen, J. E., & Woolfrey, K. M. (2011). Dendritic channelopathy causes human oocyte death. Science Translational Medicine 11(485).
spine pathology in neuropsychiatric disorders. Nature Neuroscience 14(3), 285–293. https://doi.org/10.1126/scitranslmed.aav8731.
https://doi.org/10.1038/nn.2741. Santiago, M. F., Veliskova, J., Patel, N. K., Lutz, S. E., Caille, D., Charollais, A., ... Scemes, E.
Poon, I. K. H., Chiu, Y. H., Armstrong, A. J., Kinchen, J. M., Juncadella, I. J., Bayliss, D. A., & (2011). Targeting Pannexin1 improves seizure outcome. PLoS One 6(9), Article
Ravichandran, K. S. (2014). Unexpected link between an antibiotic, pannexin chan- e25178. https://doi.org/10.1371/journal.pone.0025178.
nels and apoptosis. Nature 507(7492), 329–334. https://doi.org/10.1038/ Scemes, E., & Spray, D. C. (2012). Extracellular K+ and astrocyte signaling via connexin
nature13147. and pannexin channels. Neurochemical Research 37(11), 2310–2316. https://doi.org/
Phillips, N. A., & Mate-Kole, C. C. (1997). Cognitive deficits in peripheral vascular disease. 10.1007/s11064-012-0759-4.
Stroke 28(4), 777–784. https://doi.org/10.1161/01.STR.28.4.777. Scemes, E., Suadicani, S. O., Dahl, G., & Spray, D. C. (2007). Connexin and pannexin medi-
Prochnow, N., Abdulazim, A., Kurtenbach, S., Wildförster, V., Dvoriantchikova, G., Hanske, ated cell-cell communication. Neuron Glia Biology 3(3), 199–208. https://doi.org/10.
J., ... Zoidl, G. (2012). Pannexin1 stabilizes synaptic plasticity and is needed for learn- 1017/S1740925X08000069.
ing. PLoS One 7(12), Article e51767. https://doi.org/10.1371/journal.pone.0051767. Scemes, E., Velíšek, L., & Velíšková, J. (2019). Astrocyte and neuronal Pannexin1 contrib-
Pronin, A., Pham, D., An, W., Dvoriantchikova, G., Reshetnikova, G., Qiao, J., ... Shestopalov, ute distinctly to seizures. ASN Neuro 11, Article 175909141983350. https://doi.org/
V. I. (2019). Inflammasome activation induces pyroptosis in the retina exposed to oc- 10.1177/1759091419833502.
ular hypertension injury. Frontiers in Molecular Neuroscience 12, 36. https://doi.org/ Schlaggar, B. L., Fox, K., & O’Leary, D. M. (1993). Postsynaptic control of plasticity in devel-
10.3389/fnmol.2019.00036. oping somatosensory cortex. Nature.. https://doi.org/10.1038/364623a0.
Qian, Y., Wang, X., Liu, Y., Li, Y., Colvin, R. A., Tong, L., ... Chen, X. (2014). Extracellular ATP Sebastião, A. M., & Ribeiro, J. A. (2015). Neuromodulation and metamodulation by aden-
is internalized by macropinocytosis and induces intracellular ATP increase and drug osine: Impact and subtleties upon synaptic plasticity regulation. Brain Research 1621,
resistance in cancer cells. Cancer Letters 351(2), 242–251. https://doi.org/10.1016/j. 102–113. https://doi.org/10.1016/j.brainres.2014.11.008.
canlet.2014.06.008. Seminario-Vidal, L., Okada, S. F., Sesma, J. I., Kreda, S. M., Van Heusden, C. A., Zhu, Y., ...
Qu, R., Dong, L., Zhang, J., Yu, X., Wang, L., & Zhu, S. (2020). Cryo-EM structure of human Lazarowski, E. R. (2011). Rho signaling regulates pannexin 1-mediated ATP release
heptameric Pannexin 1 channel. Cell Research. https://doi.org/10.1038/s41422-020- from airway epithelia. Journal of Biological Chemistry 286(30), 26277–26286.
0298-5. https://doi.org/10.1074/jbc.M111.260562.
Qu, Y., Misaghi, S., Newton, K., Gilmour, L. L., Louie, S., Cupp, J. E., ... Dixit, V. M. (2011). Seo, J. H., Dalal, M. S., Calderon, F., & Contreras, J. E. (2020). Myeloid Pannexin-1 mediates
Pannexin-1 is required for ATP release during apoptosis but not for Inflammasome acute leukocyte infiltration and leads to worse outcomes after brain trauma. Journal
activation. The Journal of Immunology 186(11), 6553–6561. https://doi.org/10.4049/ of Neuroinflammation 17(1), 1–13. https://doi.org/10.1186/s12974-020-01917-y.
jimmunol.1100478. Séror, C., Melki, M. T., Subra, F., Raza, S. Q., Bras, M., Saïdi, H., ... Perfettini, J. L. (2011). Ex-
Rafael, A., Cairus, A., Tizzoni, M., Abudara, V., & Vitureira, N. (2020). Glial ATP and large tracellular ATP acts on P2Y2 purinergic receptors to facilitate HIV-1 infection. Journal
pore channels modulate synaptic strength in response to chronic inactivity. of Experimental Medicine 208(9), 1823–1834. https://doi.org/10.1084/jem.20101805.
Molecular Neurobiology. https://doi.org/10.1007/s12035-020-01919-0. Shao, Q., Lindstrom, K., Shi, R., Kelly, J., Schroeder, A., Juusola, J., ... Laird, D. W. (2016). A
Ray, A., Zoidl, G., Weickert, S., Wahle, P., & Dermietzel, R. (2005). Site-specific and devel- germline variant in the PANX1 gene has reduced channel function and is associated
opmental expression of pannexin1 in the mouse nervous system. The European Jour- with multisystem dysfunction. Journal of Biological Chemistry. https://doi.org/10.
nal of Neuroscience 21(12), 3277–3290. 1074/jbc.M116.717934.
Recouvreux, M. V., & Commisso, C. (2017). Macropinocytosis: A metabolic adaptation to Sharma, A. K., Charles, E. J., Zhao, Y., Narahari, A. K., Baderdinni, P. K., Good, M. E., ...
nutrient stress in cancer. Frontiers in Endocrinology 8, 261. https://doi.org/10.3389/ Laubach, V. E. (2018). Pannexin-1 channels on endothelial cells mediate vascular in-
fendo.2017.00261. flammation during lung ischemia-reperfusion injury. American Journal of Physiology -
Reh, R. K., Dias, B. G., Nelson, C. A., Kaufer, D., Werker, J. F., Kolb, B., ... Hensch, T. K. (2020). Lung Cellular and Molecular Physiology 315(2), L301–L312. https://doi.org/10.1152/
Critical period regulation across multiple timescales. Proceedings of the National ajplung.00004.2018.
Academy of Sciences of the United States of America 117(38), 23242–23251. https:// Shea, T. B., Fischer, I., & Sapirstein, V. S. (1985). Effect of retinoic acid on growth and mor-
doi.org/10.1073/pnas.1820836117. phological differentiation of mouse NB2a neuroblastoma cells in culture.
Reisinger, S., Khan, D., Kong, E., Berger, A., Pollak, A., & Pollak, D. D. (2015). The poly(I:C)- Developmental Brain Research 21(2), 307–314. https://doi.org/10.1016/0165-3806
induced maternal immune activation model in preclinical neuropsychiatric drug dis- (85)90220-2.
covery. Pharmacology & Therapeutics 149, 213–226. https://doi.org/10.1016/j. Silverman, W., Locovei, S., & Dahl, G. (2008). Probenecid, a gout remedy, inhibits
pharmthera.2015.01.001. pannexin 1 channels. American Journal of Physiology - Cell Physiology 295(3),
Reynolds, C. P., Matthay, K. K., Villablanca, J. G., & Maurer, B. J. (2003). Retinoid therapy of C761–C767. https://doi.org/10.1152/ajpcell.00227.2008.
high-risk neuroblastoma. Cancer Letters 197(1–2), 185–192. https://doi.org/10.1016/ Silverman, W. R., de Rivero Vaccari, J. P., Locovei, S., Qiu, F., Carlsson, S. K., Scemes, E., ...
S0304-3835(03)00108-3. Dahl, G. (2009). The Pannexin 1 channel activates the Inflammasome in neurons
Rigato, C., Swinnen, N., Buckinx, R., Couillin, I., Mangin, J. M., Rigo, J. M., ... Le Corronc, H. and astrocytes. Journal of Biological Chemistry 284(27), 18143–18151. https://doi.
(2012). Microglia proliferation is controlled by P2X7 receptors in a pannexin-1- org/10.1074/jbc.M109.004804.
independent manner during early embryonic spinal cord invasion. Journal of Sipe, G. O., Lowery, R. L., Tremblay, M.-È., Kelly, E. A., Lamantia, C. E., & Majewska, A. K.
Neuroscience 32(34), 11559–11573. https://doi.org/10.1523/JNEUROSCI.1042-12. (2016). Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex.
2012. Nature Communications 7, 10905. https://doi.org/10.1038/ncomms10905.
Rodrigues, R. J., Marques, J. M., & Cunha, R. A. (2019). Purinergic signalling and brain de- Sosinsky, G. E., Boassa, D., Dermietzel, R., Duffy, H. S., Laird, D. W., MacVicar, B., ... Dahl, G.
velopment. Seminars in Cell and Developmental Biology 95, 34–41. https://doi.org/10. (2011). Pannexin channels are not gap junction hemichannels. Channels 5(3),
1016/j.semcdb.2018.12.001. 193–197. https://doi.org/10.4161/chan.5.3.15765.

15
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

Srinivasan, R., Lu, T. -Y., Chai, H., Golshani, P., Coppola, G., Khakh Correspondence, B. S., ... Wei, L., Yang, X., Shi, X., & Chen, Y. (2015). Pannexin-1 silencing inhibits the proliferation
Khakh, B. S. (2016). New transgenic mouse lines for selectively targeting astrocytes and of U87-MG cells. Molecular Medicine Reports 11(5), 3487–3492. https://doi.org/10.
studying calcium signals in astrocyte processes in situ and in vivo NeuroResource new 3892/mmr.2015.3169.
transgenic mouse lines for selectively targeting astrocytes and studying calcium signals Wei, R., Wang, J., Xu, Y., Yin, B., He, F., Du, Y., ... Luo, B. (2015). Probenecid protects against
in astrocyte processes in situ and in vivo. https://doi.org/10.1016/j.neuron.2016.11.030. cerebral ischemia/reperfusion injury by inhibiting lysosomal and inflammatory dam-
Stefanis, L. (2012). α-Synuclein in Parkinson’s disease. Cold Spring Harbor Perspectives in age in rats. Neuroscience.. https://doi.org/10.1016/j.neuroscience.2015.05.070.
Medicine 2(2). https://doi.org/10.1101/cshperspect.a009399. Wei, Z. -Y., Qu, H. -L., Dai, Y. -J., Wang, Q., Ling, Z. -M., Su, W. -F., ... Chen, G. (2021).
Swartz, T. H., Esposito, A. M., Durham, N. D., Hartmann, B. M., & Chen, B. K. (2014). P2X- Pannexin 1, a large-pore membrane channel, contributes to hypotonicity-induced
selective purinergic antagonists are strong inhibitors of HIV-1 fusion during both cell- ATP release in Schwann cells. Neural Regeneration Research 16(5), 899. https://doi.
to-cell and cell-free infection. Journal of Virology 88(19), 11504–11515. https://doi. org/10.4103/1673-5374.290911.
org/10.1128/jvi.01158-14. Weilinger, N. L., Lohman, A. W., Rakai, B. D., Ma, E. M. M., Bialecki, J., Maslieieva, V., ...
Swayne, L. A., & Bennett, S. A. L. (2016). Connexins and pannexins in neuronal develop- Thompson, R. J. (2016). Metabotropic NMDA receptor signaling couples Src family ki-
ment and adult neurogenesis. BMC Cell Biology 17(1). https://doi.org/10.1186/ nases to pannexin-1 during excitotoxicity. Nature Neuroscience 19(3), 432–442.
s12860-016-0089-5. https://doi.org/10.1038/nn.4236.
Swayne, L. A., & Boyce, A. K. J. (2017). Regulation of Pannexin 1 surface expression by ex- Weilinger, N. L., Tang, P. L., & Thompson, R. J. (2012). Anoxia-induced NMDA receptor ac-
tracellular ATP: Potential implications for nervous system function in health and dis- tivation opens Pannexin channels via Src family kinases. Journal of Neuroscience 32
ease. Frontiers in Cellular Neuroscience. https://doi.org/10.3389/fncel.2017.00230. (36), 12579–12588. https://doi.org/10.1523/JNEUROSCI.1267-12.2012.
Swayne, L. A., Johnstone, S. R., Ng, C. S., Sanchez-Arias, J. C., Good, M. E., Penuela, S., & Wellmann, M., Álvarez-Ferradas, C., Maturana, C. J., Sáez, J. C., & Bonansco, C. (2018).
Isakson, B. E. (2020). Consideration of pannexin 1 channels in covid-19 pathology Astroglial ca 2+ −dependent hyperexcitability requires p2y 1 purinergic receptors
and treatment. American Journal of Physiology - Lung Cellular and Molecular Physiology and pannexin-1 channel activation in a chronic model of epilepsy. Frontiers in
319(1), L121–L125. https://doi.org/10.1152/ajplung.00146.2020. Cellular Neuroscience 12, 446. https://doi.org/10.3389/fncel.2018.00446.
Sweeney, M. D., Kisler, K., Montagne, A., Toga, A. W., & Zlokovic, B. V. (2018). The role of Whyte-Fagundes, P., & Zoidl, G. (2018). Mechanisms of pannexin1 channel gating and
brain vasculature in neurodegenerative disorders. Nature Neuroscience 21(10), regulation. Biochimica et Biophysica Acta - Biomembranes 1860(1), 65–71 Elsevier B.
1318–1331. https://doi.org/10.1038/s41593-018-0234-x. V https://doi.org/10.1016/j.bbamem.2017.07.009.
Takano, T., Xu, C., Funahashi, Y., Namba, T., & Kaibuchi, K. (2015). Neuronal polarization. Wicki-Stordeur, L. E., Dzugalo, A. D., Swansburg, R. M., Suits, J. M., & Swayne, L. A. (2012).
Development (Cambridge) 142(12), 2088–2093. https://doi.org/10.1242/dev.114454. Pannexin 1 regulates postnatal neural stem and progenitor cell proliferation. Neural
Takeuchi, T., Duszkiewicz, A. J., & Morris, R. G. M. (2014). The synaptic plasticity and Development 7(1), 11. https://doi.org/10.1186/1749-8104-7-11.
memory hypothesis: Encoding, storage and persistence. Philosophical Transactions of Willebrords, J., Maes, M., Crespo Yanguas, S., & Vinken, M. (2017). Inhibitors of connexin
the Royal Society, B: Biological Sciences 369(1633). https://doi.org/10.1098/rstb.2013. and pannexin channels as potential therapeutics. Pharmacology and Therapeutics 180,
0288 Royal Society. 144–160 Elsevier Inc https://doi.org/10.1016/j.pharmthera.2017.07.001.
Tan, Z., Liu, Y., Xi, W., Lou, H. F., Zhu, L., Guo, Z., ... Duan, S. (2017). Glia-derived ATP in- Whyte-Fagundes, P., & Zoidl, G. (2016). Inducible targeting of CNS astrocytes in Aldh1l1-
versely regulates excitability of pyramidal and CCK-positive neurons. Nature CreERT2 BAC transgenic mice. F1000Res. 5, 2934. https://doi.org/10.12688/
Communications 8(1), 1–14. https://doi.org/10.1038/ncomms13772. f1000research.10509.1.
Tasci, I., Safer, U., Naharci, M. I., Gezer, M., Demir, O., Bozoglu, E., & Doruk, H. (2018). Un- Wicki-Stordeur, L. E., Sanchez-Arias, J. C., Dhaliwal, J., Carmona-Wagner, E. O.,
detected peripheral arterial disease among older adults with Alzheimer’s disease and Shestopalov, V. I., Lagace, D. C., & Swayne, L. A. (2016). Pannexin 1 differentially af-
other dementias. American Journal of Alzheimer’s Disease and Other Dementias 33(1), fects neural precursor cell maintenance in the ventricular zone and Peri-infarct cor-
5–11. https://doi.org/10.1177/1533317517724000. tex. Journal of Neuroscience 36(4), 1203–1210. https://doi.org/10.1523/JNEUROSCI.
Thompson, R. J. (2006). Ischemia opens neuronal gap junction Hemichannels. Science 312 0436-15.2016.
(5775), 924–927. https://doi.org/10.1126/science.1126241. Wicki-Stordeur, L. E., & Swayne, L. A. (2013). Panx1 regulates neural stem and progenitor
Thompson, R. J., Jackson, M. F., Olah, M. E., Rungta, R. L., Hines, D. J., Beazely, M. A., ... cell behaviours associated with cytoskeletal dynamics and interacts with multiple cy-
MacVicar, B. A. (2008). Activation of pannexin-1 hemichannels augments aberrant toskeletal elements. Cell Communication and Signaling: CCS 11(1), 62. https://doi.org/
bursting in the hippocampus. Science 322(5907), 1555–1559. 10.1186/1478-811X-11-62.
Tremblay, M.-È., Lowery, R. L., & Majewska, A. K. (2010). Microglial interactions with syn- Wicki-Stordeur, L. E., & Swayne, L. A. (2014). The emerging Pannexin 1 signalome: A new
apses are modulated by visual experience. PLoS Biology 8(11), Article e1000527. nexus revealed? Frontiers in Cellular Neuroscience 7(JAN). https://doi.org/10.3389/
https://doi.org/10.1371/journal.pbio.1000527. fncel.2013.00287.
Tremblay, M.-È., Stevens, B., Sierra, A., Wake, H., Bessis, A., & Nimmerjahn, A. (2011). The Wieraszko, A., Goldsmith, G., & Seyfried, T. N. (1989). Stimulation-dependent release of
role of microglia in the healthy brain. The Journal of neuroscience: the official journal of adenosine triphosphate from hippocampal slices. Brain Research 485(2), 244–250.
the Society for Neuroscience 31(45), 16064–16069. https://doi.org/10.1523/ https://doi.org/10.1016/0006-8993(89)90567-2.
JNEUROSCI.4158-11.2011. Woolf, C. J. (2011). Central sensitization: Implications for the diagnosis and treatment of
Truding, R., Shelanski, M. L., Daniels, M. P., & Morell, P. (1974). Comparison of surface pain. Pain 152(Supplement), S2–S15. https://doi.org/10.1016/j.pain.2010.09.030.
membranes isolated from cultured murine neuroblastoma cells in the differentiated Wu, K. C., Lee, C. Y., Chou, F. Y., Chern, Y., & Lin, C. J. (2020). Deletion of equilibrative nu-
or undifferentiated state. Journal of Biological Chemistry 249(12), 3973–3982. cleoside transporter-2 protects against lipopolysaccharide-induced neuroinflamma-
Ulrich, H., & Illes, P. (2014). P2X receptors in maintenance and differentiation of neural tion and blood-brain barrier dysfunction in mice. Brain, Behavior, and Immunity 84,
progenitor cells. Neural Regeneration Research 9(23), 2040–2041. https://doi.org/10. 59–71. https://doi.org/10.1016/j.bbi.2019.11.008.
4103/1673-5374.147925. Wu, P. Y., Lin, Y. C., Chang, C. L., Lu, H. T., Chin, C. H., Hsu, T. T., ... Sun, S. H. (2009). Func-
Varani, K., Rigamonti, D., Sipione, S., Camurri, A., Borea, P. A., Cattabeni, F., ... Cattaneo, E. tional decreases in P2X7 receptors are associated with retinoic acid-induced neuronal
(2001). Aberrant amplification of A 2A receptor signaling in striatal cells expressing differentiation of neuro-2a neuroblastoma cells. Cellular Signalling 21(6), 881–891.
mutant huntingtin. The FASEB Journal 15(7), 1245–1247. https://doi.org/10.1096/fj. Xiong, X. X., Gu, L. J., Shen, J., Kang, X. H., Zheng, Y. Y., Yue, S. B., & Zhu, S. M. (2014). Pro-
00-0730fje. benecid protects against transient focal cerebral ischemic injury by inhibiting HMGB1
Velasquez, S., Prevedel, L., Valdebenito, S., Gorska, A. M., Golovko, M., Khan, N., ... Eugenin, release and attenuating AQP4 expression in mice. Neurochemical Research 39(1),
E. A. (2020). Circulating levels of ATP is a biomarker of HIV cognitive impairment. 216–224. https://doi.org/10.1007/s11064-013-1212-z.
EBioMedicine 51. https://doi.org/10.1016/j.ebiom.2019.10.029. Xu, X., Wicki-Stordeur, L. E., Sanchez-Arias, J. C., Liu, M., Weaver, M. S., Choi, C. S. W., &
Vogt, A., Hormuzdi, S. G., & Monyer, H. (2005). Pannexin1 and Pannexin2 expression in Swayne, L. A. (2018). Probenecid disrupts a novel Pannexin 1-Collapsin response me-
the developing and mature rat brain. Brain Research. Molecular Brain Research 141 diator protein 2 interaction and increases microtubule stability. Frontiers in Cellular
(1), 113–120. Neuroscience 12. https://doi.org/10.3389/fncel.2018.00124.
Vultaggio-Poma, V., Sarti, A. C., & Di Virgilio, F. (2020). Extracellular ATP: A feasible target Xu, Z., Xu, P., Chen, Y., Liu, J., Zhang, Y., Lv, Y., ... Chen, G. (2015). ENT1 inhibition attenu-
for cancer therapy. Cells 9(11). https://doi.org/10.3390/cells9112496. ates epileptic seizure severity via regulation of glutamatergic neurotransmission.
Walker, D. S., & Schafer, W. R. (2020). Distinct roles for innexin gap junctions and hemi- Neuromolecular Medicine 17(1), 1–11. https://doi.org/10.1007/s12017-014-8338-2.
channels in mechanosensation. ELife 9. https://doi.org/10.7554/eLife.50597. Yang, D., He, Y., Muñoz-Planillo, R., Liu, Q., & Núñez, G. (2015). Caspase-11 requires the
Walz, W., Ilschner, S., Ohlemeyer, C., Banati, R., & Kettenmann, H. (1993). Extracellular Pannexin-1 channel and the purinergic P2X7 pore to mediate Pyroptosis and Endo-
ATP activates a cation conductance and a K+ conductance in cultured microglial toxic shock. Immunity 43(5), 923–932. https://doi.org/10.1016/j.immuni.2015.10.
cells from mouse brain. Journal of Neuroscience 13(10). https://doi.org/10.1523/ 009.
jneurosci.13-10-04403.1993. Yang, Y., Delalio, L., Best, A. K., Macal, E., Milstein, J., Donnelly, I., ... Johnstone, S. R. (2020).
Wang, J., Ma, M., Locovei, S., Keane, R. W., & Dahl, G. (2007). Modulation of membrane Endothelial pannexin 1 channels control inflammation by regulating intracellular cal-
channel currents by gap junction protein mimetic peptides: Size matters. American cium 1. Journal of Immunology. https://doi.org/10.1101/750323.
Journal of Physiology - Cell Physiology 293(3). https://doi.org/10.1152/ajpcell.00097. Yegutkin, G. G. (2014). Enzymes involved in metabolism of extracellular nucleotides and
2007. nucleosides: Functional implications and measurement of activities. Critical Reviews
Wang, J., Jackson, D. G., & Dahl, G. (2013). The food dye FD&C Blue No. 1 is a selective in- in Biochemistry and Molecular Biology 49(6), 473–497. https://doi.org/10.3109/
hibitor of the ATP release channel Panx1. Journal of General Physiology 141(5), 10409238.2014.953627.
649–656. https://doi.org/10.1085/jgp.201310966. Yeung, A. K., Patil, C. S., & Jackson, M. F. (2020). Pannexin-1 in the CNS: Emerging concepts
Wang, W., Qu, R., Dou, Q., Wu, F., Wang, W., Chen, B., ... Sang, Q. (2021). Homozygous var- in health and disease. Journal of Neurochemistry 154(December 2019), 1–18. https://
iants in PANX1 cause human oocyte death and female infertility. European Journal of doi.org/10.1111/jnc.15004.
Human Genetics. https://doi.org/10.1038/s41431-020-00807-4. Yi, C., Mei, X., Ezan, P., Mato, S., Matias, I., Giaume, C., & Koulakoff, A. (2016). Astroglial
Weaver, J. L., Arandjelovic, S., Brown, G., Mendu, S. K., Schappe, M. S., Buckley, M. W., ... connexin43 contributes to neuronal suffering in a mouse model of Alzheimer’s dis-
Bayliss, D. A. (2017). Hematopoietic pannexin 1 function is critical for neuropathic ease. Cell Death and Differentiation 23(10), 1691–1701. https://doi.org/10.1038/cdd.
pain. Scientific Reports 7(1), 1–15. https://doi.org/10.1038/srep42550. 2016.63.

16
J.C. Sanchez-Arias, E. van der Slagt, H.A. Vecchiarelli et al. Pharmacology & Therapeutics 225 (2021) 107840

Young, J. D., Yao, S. Y. M., Sun, L., Cass, C. E., & Baldwin, S. A. (2008). Human equilibrative vascular cells of the cerebral cortex. Journal of Neuroscience 34(36), 11929–11947.
nucleoside transporter (ENT) family of nucleoside and nucleobase transporter pro- https://doi.org/10.1523/JNEUROSCI.1860-14.2014.
teins. Xenobiotica 38(7–8), 995–1021. https://doi.org/10.1080/00498250801927427. Zhang, Y., Laumet, G., Chen, S. R., Hittelman, W. N., & Pan, H. L. (2015). Pannexin-1 up-
Zappala, A., Cicero, D., Serapide, M. F., Paz, C., Catania, M. V., Falchi, M., ... Cicirata, F. regulation in the dorsal root ganglion contributes to neuropathic pain development.
(2006). Expression of pannexin1 in the CNS of adult mouse: Cellular localization Journal of Biological Chemistry 290(23), 14647–14655. https://doi.org/10.1074/jbc.
and effect of 4-aminopyridine-induced seizures. Neuroscience 141(1), 167–178. M115.650218.
Zhang, D., Jin, W., Liu, H., Liang, T., Peng, Y., Zhang, J., & Zhang, Y. (2020). ENT1 inhibition Zheng, D., Liwinski, T., & Elinav, E. (2020). Inflammasome activation and regulation: To-
attenuates apoptosis by activation of cAMP/pCREB/Bcl2 pathway after MCAO in rats. ward a better understanding of complex mechanisms. Cell Discovery 6(1). https://
Experimental Neurology 331. https://doi.org/10.1016/j.expneurol.2020.113362. doi.org/10.1038/s41421-020-0167-x.
Zhang, H., Kang, E., Wang, Y., Yang, C., Yu, H., Wang, Q., ... Xu, Z. (2016). Brain-specific Zhou, K. Q., Green, C. R., Bennet, L., Gunn, A. J., & Davidson, J. O. (2019). The role of
Crmp2 deletion leads to neuronal development deficits and behavioural impairments connexin and pannexin channels in perinatal brain injury and inflammation.
in mice. Nature Communications 7. https://doi.org/10.1038/ncomms11773. Frontiers in Physiology 10(FEB). https://doi.org/10.3389/fphys.2019.00141 Frontiers
Zhang, J. M., Wang, H. K., Ye, C. Q., Ge, W., Chen, Y., Jiang, Z. L., ... Duan, S. (2003). ATP re- Media S.A..
leased by astrocytes mediates glutamatergic activity-dependent Heterosynaptic sup- Zhou, L., Liu, C., Wang, Z., Shen, H., Wen, Z., Chen, D., ... Chen, G. (2018). Pannexin-1 is in-
pression. Neuron 40(5), 971–982. https://doi.org/10.1016/S0896-6273(03)00717-7. volved in neuronal apoptosis and degeneration in experimental intracerebral hemor-
Zhang, M., Piskuric, N. A., Vollmer, C., & Nurse, C. A. (2012). P2Y2 receptor activation rhage in rats. Molecular Medicine Reports 17(4), 5684–5691. https://doi.org/10.3892/
opens pannexin-1 channels in rat carotid body type II cells: Potential role in amplify- mmr.2018.8624.
ing the neurotransmitter ATP. Journal of Physiology 590(17), 4335–4350. https://doi. Zhou, X., Chen, Q., Huang, H., Zhang, J., Wang, J., Chen, Y., ... Xu, Z. (2020). Inhibition of p38
org/10.1113/jphysiol.2012.236265. MAPK regulates epileptic severity by decreasing expression levels of A1R and ENT1.
Molecular Medicine Reports 22(6), 5348–5357. https://doi.org/10.3892/mmr.2020.
Zhang, X., Chen, Y., Wang, C., & Huang, L. Y. M. (2007). Neuronal somatic ATP release trig-
11614.
gers neuron-satellite glial cell communication in dorsal root ganglia. Proceedings of
Zimmermann, H. (2016). Extracellular ATP and other nucleotides—Ubiquitous triggers of
the National Academy of Sciences of the United States of America 104(23),
intercellular messenger release. Purinergic Signalling 12(1). https://doi.org/10.1007/
9864–9869. https://doi.org/10.1073/pnas.0611048104.
s11302-015-9483-2.
Zhang, Y., Chen, K., Sloan, S. A., Bennett, M. L., Scholze, A. R., O’Keeffe, S., ... Wu, J. Q. (2014).
An RNA-sequencing transcriptome and splicing database of glia, neurons, and

17

You might also like