The role of ASICs in cerebral ischemia - Zhi-Gang Xiong1 and Tian-Le Xu2

Page created by Shane Campbell
 
CONTINUE READING
The role of ASICs in cerebral ischemia - Zhi-Gang Xiong1 and Tian-Le Xu2
Focus Article

The role of ASICs in cerebral
ischemia
Zhi-Gang Xiong1∗ and Tian-Le Xu2

                 Cerebral ischemia is a leading cause of death and long-term disabilities worldwide.
                 Excessive intracellular Ca2+ accumulation in neurons has been considered essential
                 for neuronal injury associated with cerebral ischemia. Although the involvement
                 of glutamate receptors in neuronal Ca2+ accumulation and toxicity has been the
                 subject of intensive investigation, inhibitors for these receptors showed little effect
                 in clinical trials. Thus, additional Ca2+ toxicity pathway(s) must be involved.
                 Acidosis is a common feature in cerebral ischemia and was known to cause brain
                 injury. The mechanisms were, however, unclear. The finding that ASIC1a channels
                 are highly enriched in brain neurons, their activation by ischemic acidosis, and
                 their demonstrated Ca2+ permeability suggested a role for these channels in Ca2+
                 accumulation and neuronal injury associated with cerebral ischemia. Indeed, a
                 number of studies have now provided solid evidence supporting the involvement
                 of ASIC1a channel activation in ischemic brain injury. © 2012 WILEY-VCH Verlag GmbH
                 & Co. KGaA, Weinheim.

                                                         How to cite this article:
                              WIREs Membr Transp Signal 2012, 1:655–662. doi: 10.1002/wmts.57

INTRODUCTION                                                                 Although multiple pathways and biochemical
                                                                      changes contribute to ischemic brain injury, exces-
I  schemic stroke, or cerebral ischemia, is the third
   most common cause of death in most industrialized
countries. Although major advances have occurred
                                                                      sive intracellular Ca2+ accumulation and resultant
                                                                      toxicity has been considered essential in the pathol-
                                                                      ogy of cerebral ischemia.2 In the resting conditions,
in the prevention of stroke during the past several
                                                                      free intracellular Ca2+ concentration ([Ca2+ ]i ) in neu-
decades, no effective treatment is now available.
                                                                      rons is maintained at nanomolar range. Following
Current clinical practices for stroke patients utilize
                                                                      cerebral ischemia, however, [Ca2+ ]i can rise to as
thrombolytic agent tissue plasminogen activator (tPA)
                                                                      high as several micromolars. Excessive accumula-
to reopen the clotted vessels.1 This approach,
                                                                      tion of Ca2+ in neurons leads to uncontrolled acti-
however, has very limited success due to a short
                                                                      vation of various enzymes causing breakdown of
therapeutic time window of 3 h and side effect of
                                                                      proteins, lipids, and nucleic acids, and the destruc-
intracranial hemorrhage. On the other hand, cell
                                                                      tion of neurons.3–5 In addition, overloading Ca2+ in
death is prominent following stroke. Therefore, the
                                                                      mitochondria can cause opening of mitochondria per-
need for a continuous search of neuronal damage
                                                                      meability transition pore (PTP), promoting apoptosis
mechanisms and effective therapeutic strategies for
                                                                      through release of cytochrome c and activation of
neuroprotection remains high.
                                                                      caspases.6
                                                                             Ca2+ can enter neurons through various path-
Conflict of interest: Both authors have no conflict of interest to    ways, among which glutamate receptor-gated chan-
declare.                                                              nels have received the most attention. Unfortunately,
∗
    Correspondence to: zxiong@msm.edu                                 clinical trials targeting these channels have shown
1
 Neuroscience Institute, Morehouse School of Medicine, Atlanta,       little effect in improving the outcome of cerebral
GA 30310, USA                                                         ischemia.7 Multiple factors may have contributed
2
 Neuroscience Division, Department of Biochemistry and Molec-         to the failure of the trials. In particular, additional
ular Cell Biology, Institute of Medical Sciences, Shanghai
Jiao Tong University School of Medicine, Shanghai 200025,             glutamate-independent Ca2+ entry and toxicity path-
China                                                                 ways must be considered.

Volume 1, September/October 2012 © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.                                        655
Focus Article                                                                                     wires.wiley.com/mts

BRAIN ACIDOSIS IN CEREBRAL                                        To provide a link between ASIC1a activation
ISCHEMIA                                                   and ischemic brain injury, both in vitro neuronal
                                                           injury and in vivo cerebral ischemia models were
Acidosis, a condition characterized by too much acid       employed. A brief (1 h) acid incubation, in the pres-
in the tissue or body fluid, is one of the most common     ence of blockers of glutamate receptors and voltage-
pathophysiological changes in the brain associated         gated Ca2+ channels, was able to induce substantial
with acute neurological conditions such as cerebral        neuronal injury measured at 6 h or 24 h after acid
ischemia.8,9 In the ischemic core, for example, a rapid    treatment. This acid-induced, glutamate-independent
drop of pH to 6.5 or lower is frequently observed.10,11    neuronal injury was inhibited by amiloride or PcTX1,
The lack of oxygen supply promotes anaerobic               supporting the involvement of homomeric ASIC1a
glycolysis which leads to increased production of          channels. Consistent with an essential role for ASIC1a
lactic acid.11 Accumulation of lactic acid, along with     subunit in acid injury, neurons cultured from ASIC1
increased production of H+ from ATP hydrolysis,            knockout mice were resistant to acid incubation.15,18
and release of H+ from presynaptic terminals,12            Reducing the concentration of extracellular Ca2+ ,
contributes to the acid buildup in the brain. Acidosis     which decreases the driving force for Ca2+ entry, also
has long been recognized to aggravate brain injury         ameliorated acid injury. To know whether ASIC1a-
associated with cerebral ischemia.8,9 However, the         mediated injury can also take place in ischemic
detailed mechanism(s) remained elusive, although a         condition, acid-mediated neuronal injury was stud-
number of possibilities have been suggested long           ied in the condition of oxygen glucose deprivation
before the role of acid-sensing ion channels (ASICs)       (OGD). OGD, in the presence of blockers of glu-
was recognized.8,13,14                                     tamate receptors and voltage-gated Ca2+ channels,
                                                           enhanced the acid-induced neuronal injury which
ASIC1a ACTIVATION IS INVOLVED                              was inhibited by amiloride and PcTX1. Thus, in
                                                           vitro studies support a role for ASIC1a activation
IN ACIDOSIS-MEDIATED ISCHEMIC                              in acidosis-mediated, Ca2+ -dependent, ischemic neu-
BRAIN INJURY                                               ronal injury. Since a recent study showed that PcTX1
On the basis of the evidence that ASIC1a subunits          also inhibits heteromeric ASIC1a/ASIC2b channels in
are highly expressed in brain neurons, their activa-       addition to homomeric ASIC1a channels,19 the poten-
tion by pH drops to the level commonly seen in             tial contribution of heteromeric ASIC1a/ASIC2b chan-
cerebral ischemia, and their permeability to Ca2+          nels to acidosis-mediated neuronal injury cannot be
and Na+ , Xiong and colleagues tested the hypoth-          excluded.
esis that activation of ASIC1a channels is involved               Does activation of ASIC1a channels also play a
in neuronal Ca2+ accumulation and injury associated        role in ischemic brain injury in vivo? To answer this
with cerebral ischemia.15 Using patch-clamp recording      question, two sets of experiments were performed.
and fast-perfusion technique, large inward currents        The first experiment examined whether application
were recorded in cultured mouse cortical neurons in        of ASIC inhibitors reduces ischemic brain injury, and
response to rapid perfusion of acidic solutions at pH      the second tested whether knockout of the ASIC1
levels relevant to cerebral ischemia. The acid-activated   gene renders the animal resistant to cerebral ischemia.
currents in cortical neurons were sensitive to non-        Rodent model of focal ischemia, by middle cerebral
specific ASIC blocker amiloride and partially inhibited    artery occlusion (MCAO), was employed for both
by ASIC1a-specific inhibitor PcTX1, suggesting that        experiments. In rats and mice, intracerebroventricu-
the currents were mediated by ASIC1a-containing            lar injection of ASIC1a inhibitor PcTX1 reduced the
channels. Consistent with the presence of func-            infarct volume by up to 60%, measured at 24 h after
tional homomeric ASIC1a channels, which are Ca2+ -         MCAO.15,20 Similar to the pharmacological blockade,
permeable,16 perfusion of acidic solution in these         ASIC1 gene knockout provided a comparable degree
neurons increased intracellular Ca2+ concentration,        of protection against ischemic brain injury.15 Remark-
even in the presence of blockers of voltage-gated Ca2+     ably, in contrast to most glutamate antagonists which
channels and glutamate receptors. As expected, the         have only a short time window of
WIREs Membrane Transport and Signaling                                                                             Role of ASICs in cerebral ischemia

of human brain neurons in culture. Thus, Ca2+ -                               ischemic conditions and whether the effects of
permeable ASIC1a channels represent a promising                               ASIC1a activation (e.g., membrane depolarization
therapeutic target for human cerebral ischemia.                               and intracellular Ca2+ accumulation) could be long-
                                                                              lasting and detrimental are crucial in determining the
                                                                              pathological functions of these channels.
ISCHEMIA-RELATED SIGNALS                                                            To this end, Xiong and colleagues first showed
ENHANCE THE ACTIVATION OF ASICs                                               that OGD treatment can dramatically potentiate
                                                                              the activity of neuronal ASICs—the amplitude of
Even though the results from both in vitro and                                the acid-activated current was enhanced while the
in vivo pharmacological and molecular biological                              decay of the current was reduced following 1 h
interventions clearly supported an important role for                         OGD.15 The overall outcome of these two effects
ASIC1a activation in neuronal injury associated with                          would be a dramatically increased ASIC-mediated
cerebral ischemia, several questions remained to be                           response, which might be translated into enlarged
addressed.                                                                    and longer-lasting intracellular Ca2+ elevation. What
      Under in vitro experimental conditions, currents                        could be the underlying mechanisms responsible for
of most ASIC subtypes, particularly the homomeric                             the changes of electrophysiological property of ASICs
ASIC1a channels, decay rapidly in the continuous                              observed after OGD or ischemia? This question was
presence of acidic pH,23 a phenomena of channel                               not directly answered by the studies of Xiong et al.
desensitization. In addition, pre-exposure of these                           However, later studies by different laboratories have
channels to small pH drops (e.g., from 7.4 to                                 provided important missing links. It is now known
7.2) that are insufficient to activate the channel                            that various ischemia-related signals, for example,
also suppresses the channel activity in response to                           arrachidonic acid, dynorphine, lactate, and spermine,
subsequent, large pH drops, a process termed steady-                          can dramatically enhance the amplitude and/or reduce
state desensitization.24,25 Furthermore, it has been                          the desensitization of ASICs27 (Figure 1).
shown that the activities and/or expression of some
ion channels could be dramatically down-regulated
following the ischemia/hypoxia, as exemplified                                Arachidonic Acid
by N-methyl d-aspartate (NMDA) channels in                                    Arachidonic acid (AA) is a polyunsaturated fatty acid
hypoxic turtle brain.26 Thus, whether a significant                           present in the phospholipids of all cell membranes and
amount of ASIC1a current can be activated in                                  one of the most abundant fatty acids in the brain. In

                                  Non-ischemic condition                                       Ischemic condition
                               Pre synaptic release                                            Pre synaptic release

                             H+                                                  Lactic acid
                             Ca2+
                             Na+                                            Arachidonic acid
                             Glu            Ca2+             Glu            Dynorphines
                                                                                                                      Glu
                                 Na+                         Ca2+           Nitric oxide                  Ca2+
                                             H+                                                 Na+
                                                                            Protease
                                                                                                            H+              Ca2+
                                             ASIC 1a         GluR           Spermine                        ASIC 1a
                                                                                                                            GluR

                                               CaMKII
                                                                                                            CaMKII

                                       Synaptic plasticity                                            Cell death

FIGURE 1 | Activation of ASIC1a channels and outcomes in non-ischemic and ischemic conditions. In non-ischemic condition, activation of
ASIC1a channels and slight increase of cellular Ca2+ is involved in synaptic plasticity. In ischemic condition, ASIC1a activation is dramatically
potentiated by various factors. Increased concentration of H+ per se activates more current. In addition, various ischemia-related signals not only
increased the amplitude but also reduced the desensitization of the ASIC responses. Overload of neurons with Ca2+ leads to cell death.

Volume 1, September/October 2012 © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.                                                                  657
Focus Article                                                                                     wires.wiley.com/mts

addition to being involved in cellular signaling as a      CaMKII phosphorylation of ASIC1a with KN-93, or
lipid second messenger,28 AA plays important roles in      mutation of ASIC1a at Ser478 and Ser479, produced
pathological conditions including brain ischemia.28,29     neuroprotection. Thus, phosphorylation of ASIC1a by
Following brain ischemia, the rise of [Ca2+ ]i leads       CaMKII deteriorates ASIC-mediated neuronal injury
to activation of phospholipase A2, resulting in            in ischemia.
increased production of lipid mediators including
AA. Although the exact mechanisms are unclear, high
concentrations of lipid mediators are known to cause       Dynorphins
neurotoxicity.28                                           Dynorphins are endogenous neuropeptides abun-
      On the basis of demonstrated effects of AA           dantly expressed in the central nervous system (CNS).
on a variety of voltage-gated and ligand-gated             They are involved in a variety of physiologic func-
ion channels, for example, potentiation of NMDA            tions. Under pathophysiological conditions where
channel currents,30 Allen and Attwell tested the effect    their levels are substantially elevated, these peptides
of AA on ASICs. In rat cerebellar Purkinje cells, bath     can be neurotoxic, partially mediated through gluta-
perfusion of 5 or 10 μM AA produced a large increase       mate receptors.35 Recently, Sherwood and Askwith
in the amplitude of the ASIC current. In addition          reported that, at high concentrations dynorphins such
to potentiating the peak amplitude, AA enhanced            as big dynorphin potentiate acid-activated currents in
or induced an additional sustained component of            mouse cortical neurons and in CHO cells expressing
the current.31 In heterologous expression systems,         homomeric ASIC1a channels.36 The potentiation of
AA potentiates both homomeric ASIC1a and ASIC2a            the ASIC1a activity was mediated through a reduction
channels.32 Thus, promoting the activation of ASIC1a       of the steady-state desensitization of these channels.
channels could be one of the mechanisms mediating          In the absence of big dynorphine, pre-exposing neu-
the neurotoxicity of AA.                                   rons to conditioning pH of 7.0 completely desensitizes
                                                           the channels, resulting in no responses to subsequent
                                                           larger decrease in pH (e.g., to 5.0). In the presence of
                                                           big dynorphine, however, ASIC1a currents were read-
CaMKII
                                                           ily activated. As expected, big dynorphine enhanced
Ca2+ /calmodulin (CaM)-dependent protein kinase II
                                                           ASIC1a-mediated neuronal injury during prolonged
(CaMKII) is the most abundant kinase isoform in
                                                           acidosis.
the brain and a major mediator of the function of
excitatory glutamate receptors. Influx of intracellular
Ca2+ through glutamate receptors triggers an               Lactate
autophosphorylation of CaMKII and activation of            Back in 2001, Immke and McCleskey demonstrated
the enzyme. Although activation of CaMKII plays            that, in sensory neurons that innervate the heart and
a prominent role in synaptic plasticity, increased         COS-7 cells transfected with AIC1a channels, addition
CaMKII activity has been implicated in the regulation      of lactate, at the level seen in ischemia, dramatically
of neuronal cell death after cerebral ischemia.33          increased the amplitude of the ASIC current activated
      Studies by Gao et al. demonstrated a link            by a moderate pH drop to ∼7.0.37 The increase of the
between CaMKII activation and acidotoxic neuronal          current amplitude was accompanied by a reduced
death.34 They showed that global ischemia in rats          current desensitization. Applications of lactate at
results in an increased phosphorylation of ASIC1a          pH values that do not activate ASICs caused no
at Ser478 and Ser479 by CaMKII. This phosphory-            response. Thus, lactate acted by potentiating but not
lation sensitizes the channel to low pH, exacerbat-        activating the ASICs. The effect of lactate persisted
ing cell death by allowing prolonged and increased         in excised membrane patches indicating the lack of
Ca2+ entry.                                                second messenger involvement. As lactate has the
      Consistent with the report by Xiong and              ability to chelate the divalent cations, which have
colleagues,15 Gao and colleagues observed an               a modulatory role for various membrane receptors
enhancement of ASIC currents by OGD. Inhibition            and ion channels,38–40 it was logical to hypothesize
of CaMKII with KN-93 or CaMKIINtide abolished              that potentiation of the ASIC currents could be due
the enhancement of ASIC currents, indicating an            to a chelation of Ca2+ and Mg2+ in the solution.
involvement of CaMKII. They further demonstrated           Indeed, adjusting the concentrations of Ca2+ and
an increased phosphorylation of ASIC1a protein             Mg2+ eliminated the potentiating effect of lactate.37
after transient global ischemia, which was blocked         Similar to the cardiac sensory neurons, potentiation
by intracerebroventricular administration of KN-93         of the ASIC current by lactate has been reported in
or CaMKIINtide. Pharmacological inhibition of              other neurons.31

658                             © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. Volume 1, September/October 2012
WIREs Membrane Transport and Signaling                                                              Role of ASICs in cerebral ischemia

TABLE 1 Ischemia-related Endogenous Modulators Known to Potentiate ASIC-mediated Responses
                      Neurons or ASIC subunit
 Modulator                tested effective          Modulation site(s)       Functional outcome                              References
 Arachidonic acid     Cerebellar Purkinje cells     Unknown                  Increased amplitude                               32, 60
                      Sensory neuron                                         Reduced desensitization
                      ASIC1a
                      ASIC3
 CaMK II              Hippocampal neurons           Intracellular            Increased amplitude                                 34
                      ASIC1a                        S478 and S479            Increased injury
 Dynorphine           Cortical neurons              Extracellular            Reduced steady-state desensitization                36
                      Hippocampal neurons           PcTX1 site               Increased acid-injury
                      ASIC1a
                      ASIC1b
 Lactate              Sensory neuron,               Extracellular            Increased amplitude                               37, 60
                      Cerebellar Purkinje neurons                            Reduced desensitization
                      ASIC1a
                      ASIC3
 Nitric oxide         DRG neurons                   Extracellular            Increased current amplitude                       45, 61
                      Neuro2A cells                                          Increased acid-injury
                      ASIC1a
                      ASIC1b
                      ASIC2a
                      ASIC3
 Protease             Hippocampal neurons           Extracellular            Enhanced current amplitude activated                48
                      ASIC1a                        PcTX1 site                  from a baseline pH of 7.0
                                                                             Increased recovery from desensitization
 Spermine             Cortical neurons              Extracellular            Increased amplitude                               56, 57
                      Hippocampal neurons           PcTX1 site               Reduced desensitization
                      ASIC1a                        E219 and E242            Reduced steady-state desensitization
                                                                             Increased recovery from desensitization
                                                                             Increased acid-injury

Nitric oxide                                                         ASICs, probably through oxidization of cysteine
Nitric oxide (NO) is an important reactive                           residues.45
oxygen/nitrogen species which has a variety of
physiological and pathological functions.41 During
ischemia, intracellular Ca2+ overload leads to                       Proteases
activation of the Ca2+ -dependent neuronal form of                   Brain ischemia is accompanied by increased protease
nitric oxide synthase (nNOS), resulting in an increased              activity.46 Following ischemia, blood-derived pro-
production of NO.42,43 NO can also be released by                    teases have access to the interstitial space from a com-
activated microglia.44 Excessive NO production is                    promised blood–brain barrier.46,47 Studies by Poirot
known to increase neuronal injury.44 Although the                    and colleagues demonstrated an ASIC1a-specific mod-
formation of a strong oxidant of peroxynitrite is                    ulation of the ASIC activity by serine proteases.48
likely involved in cell injury, other mechanisms cannot              Exposure of cells to trypsin, for example, leads to a
be excluded. Cadiou and colleagues reported that                     decreased ASIC1a current if the channel is activated
NO donor S-nitroso-N-acetylpenicillamine (SNAP)                      by a pH drop from pH 7.4. However, if acidification
potentiates ASIC currents in DRG neurons and in                      occurs from a lower basal pH (e.g., 7.0), a con-
CHO cells expressing ASIC subunits. Modulators                       dition pertinent to brain ischemia, protease exposure
of the cGMP/PKG pathway had no effect on the                         increases, rather than decreases, the ASIC1a activity.48
potentiation, but in excised patches from CHO                        Further studies demonstrate that trypsin modulates
cells expressing ASIC2a, the potentiation could be                   the ASIC1a function by cleaving this subunit at Arg-
reversed by externally applying reducing agents.                     145, which is in the N-terminus of the extracellular
NO, therefore, has a direct external effect on                       loop overlapping with the PcTX1 binding site.49

Volume 1, September/October 2012 © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.                                                   659
Focus Article                                                                                           wires.wiley.com/mts

Spermine                                                     CONCLUSION
Spermine is a polyvalent cation involved in var-             Stroke or cerebral ischemia is a leading health problem
ious physiological processes. High concentration             worldwide. Unfortunately, there is still no effec-
of spermine can induce neuronal depolarization
                                                             tive treatment for stroke patients. Searching for
and cytoplasmic Ca2+ overload, which may lead
                                                             new brain injury mechanisms and effective thera-
to neuronal damage.50 Following ischemia, the
                                                             peutic strategies is a major challenge and priority.
activity of ornithine decarboxylase (ODC), a rate-
                                                             Acidosis is a primary feature associated with cere-
limiting enzyme responsible for polyamine synthesis is
                                                             bral ischemia and is known to cause brain injury.
enhanced, leading to elevated level of spermine.51
                                                             The finding that activation of ASICs contributes
Although the modulation of NMDA receptor
                                                             to acidosis- and ischemia-induced intracellular Ca2+
function52,53 might explain its neurotoxicity, several
                                                             accumulation and neuronal injury has suggested that
studies have yielded inconsistent results.54,55
                                                             these channels may represent potential new thera-
      Babini and colleagues demonstrated that sper-
                                                             peutic targets for stroke intervention. In additional
mine potentiates the activities of ASICs.56 More
                                                             to developing pharmacological agents directly target-
recently, Duan and colleagues showed that extra-
                                                             ing ASICs, alternative strategies can be considered
cellular spermine exacerbated ischemic neuronal
                                                             by targeting ischemia-related signals known to poten-
injury through sensitization of ASIC1a channels to
                                                             tiate the activity of these channels (Table 1). Alter-
acidosis.57 In addition to increasing channel activa-
                                                             native neuroprotective strategies may also consider
tion, spermine reduced channel desensitization and
                                                             targeting the mechanisms and pathways that control
accelerated recovery from desensitization in response
                                                             the expression level of total protein and/or surface
to repeated acid stimulation. Thus, extracellular
                                                             ASIC1a.58,59
spermine contributes to ischemic neuronal injury,
at least in part, by enhancing the activity of ASIC1a
channels.57

ACKNOWLEDGMENT
The work in Z.G.X.’s lab is supported in part by NIH R01NS047506, R01NS066027, UL1 RR025008, U54
RR026137, AHA 0840132N, and ALZ IIRG-10-173350.

REFERENCES
 1. Weintraub MI. Thrombolysis (tissue plasminogen acti-         traumatic brain injury? Lancet Neurol 2002, 1:
    vator) in stroke: a medicolegal quagmire. Stroke 2006,       383–386.
    37:1917–1922.                                             8. Siesjo BK, Katsura K, Kristian T. Acidosis-related dam-
 2. Choi DW. Calcium-mediated neurotoxicity: relation-           age. Adv Neurol 1996, 71:209–233.
    ship to specific channel types and role in ischemic       9. Tombaugh GC, Sapolsky RM. Evolving concepts
    damage. Trends Neurosci 1988, 11:465–469.                    about the role of acidosis in ischemic neuropathology.
 3. Lee JM, Zipfel GJ, Choi DW. The changing landscape           J Neurochem 1993, 61:793–803.
    of ischaemic brain injury mechanisms. Nature 1999,       10. Nedergaard M, Kraig RP, Tanabe J, Pulsinelli WA.
    399:A7–A14.                                                  Dynamics of interstitial and intracellular pH in evolving
 4. Simonian NA, Coyle JT. Oxidative stress in neurode-          brain infarct. Am J Physiol 1991, 260:R581–R588.
    generative diseases. Annu Rev Pharmacol Toxicol 1996,    11. Rehncrona S. Brain acidosis. Ann Emerg Med 1985,
    36:83–106.                                                   14:770–776.
 5. Coyle JT, Puttfarcken P. Oxidative stress, glutamate,    12. Wemmie JA, Price MP, Welsh MJ. Acid-sensing ion
    and neurodegenerative disorders. Science 1993, 262:          channels: advances, questions and therapeutic opportu-
    689–695.                                                     nities. Trends Neurosci 2006, 29:578–586.
 6. Polster BM, Fiskum G. Mitochondrial mechanisms           13. Swanson RA, Farrell K, Simon RP. Acidosis causes
    of neural cell apoptosis. J Neurochem 2004, 90:              failure of astrocyte glutamate uptake during hypoxia.
    1281–1289.                                                   J Cereb Blood Flow Metab 1995, 15:417–424.
 7. Ikonomidou C, Turski L. Why did NMDA recep-              14. McDonald JW, Bhattacharyya T, Sensi SL, Lobner D,
    tor antagonists fail clinical trials for stroke and          Ying HS, Canzoniero LMT, Choi DW. Extracellular

660                              © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. Volume 1, September/October 2012
WIREs Membrane Transport and Signaling                                                       Role of ASICs in cerebral ischemia

    acidity potentiates AMPA receptor-mediated cortical         28. Farooqui AA, Horrocks LA. Phospholipase A2-
    neuronal death. J Neurosci 1998, 18:6290–6299.                  generated lipid mediators in the brain: the good, the
15. Xiong ZG, Zhu XM, Chu XP, Minami M, Hey J,                      bad, and the ugly. Neuroscientist 2006, 12:245–260.
    Wei WL, MacDonald JF, Wemmie JA, Price MP,                  29. Siesjo BK, Katsura K. Ischemic brain damage: focus on
    Welsh MJ, et al. Neuroprotection in ischemia: block-            lipids and lipid mediators. Adv Exp Med Biol 1992,
    ing calcium-permeable Acid-sensing ion channels. Cell           318:41–56.
    2004, 118:687–698.                                          30. Miller B, Sarantis M, Traynelis SF, Attwell D. Potenti-
16. Waldmann R, Champigny G, Bassilana F, Heurteaux C,              ation of NMDA receptor currents by arachidonic acid.
    Lazdunski M. A proton-gated cation channel involved             Nature 1992, 355:722–725.
    in acid-sensing. Nature 1997, 386:173–177.                  31. Allen NJ, Attwell D. Modulation of ASIC channels
17. Mari Y, Katnik C, Cuevas J. ASIC1a channels are                 in rat cerebellar Purkinje neurons by ischemia-related
    activated by endogenous protons during ischemia and             signals. J Physiol (Lond) 2002, 543:521–529.
    contribute to synergistic potentiation of intracellular     32. Smith ES, Cadiou H, McNaughton PA. Arachidonic
    Ca(2+) overload during ischemia and acidosis. Cell              acid potentiates acid-sensing ion channels in rat sen-
    Calcium 2010, 48:70–82.                                         sory neurons by a direct action. Neuroscience 2007,
18. Yermolaieva O, Leonard AS, Schnizler MK, Abboud                 145:686–698.
    FM, Welsh MJ. Extracellular acidosis increases neu-         33. Coultrap SJ, Vest RS, Ashpole NM, Hudmon A, Bayer
    ronal cell calcium by activating acid-sensing ion channel       KU. CaMKII in cerebral ischemia. Acta Pharmacol Sin
    1a. Proc Natl Acad Sci U S A 2004, 101:6752–6757.               2011, 32:861–872.
19. Sherwood TW, Lee KG, Gormley MG, Askwith CC.                34. Gao J, Duan B, Wang D-G, Deng X-H, Zhang
    Heteromeric acid-sensing ion channels (ASICs) com-              G-Y, Xu L, Xu T-L. Coupling between NMDA recep-
    posed of ASIC2b and ASIC1a display novel channel                tor and acid-sensing ion channel contributes to ischemic
    properties and contribute to acidosis-induced neuronal          neuronal death. Neuron 2005, 48:635–646.
    death. J Neurosci 2011, 31:9723–9734.                       35. Hauser KF, Foldes JK, Turbek CS. Dynorphin A (1-13)
20. Pignataro G, Simon RP, Xiong ZG. Prolonged activa-              neurotoxicity in vitro: opioid and non-opioid mecha-
    tion of ASIC1a and the time window for neuroprotec-             nisms in mouse spinal cord neurons. Exp Neurol 1999,
    tion in cerebral ischaemia. Brain 2007, 130:151–158.            160:361–375.
21. Biegon A, Fry PA, Paden CM, Alexandrovich A, Tsen-          36. Sherwood TW, Askwith CC. Dynorphin opioid pep-
    ter J, Shohami E. Dynamic changes in N-methyl-d-                tides enhance acid-sensing ion channel 1a activity
    aspartate receptors after closed head injury in mice:           and acidosis-induced neuronal death. J Neurosci 2009,
    implications for treatment of neurological and cogni-           29:14371–14380.
    tive deficits. Proc Natl Acad Sci U S A 2004, 101:          37. Immke DC, McCleskey EW. Lactate enhances the acid-
    5117–5122.                                                      sensing Na+ channel on ischemia-sensing neurons. Nat
22. Li M, Inoue K, Branigan D, Kratzer E, Hansen JC, Chen           Neurosci 2001, 4:869–870.
    JW, Simon RP, Xiong Z-G. Acid-sensing ion channels          38. Xiong ZG, MacDonald JF. Sensing of extracellular
    in acidosis-induced injury of human brain neurons.              calcium by neurones. Can J Physiol Pharmacol 1999,
    J Cereb Blood Flow Metab 2010, 30:1247–1260.                    77:715–721.
23. Hesselager M, Timmermann DB, Ahring PK. pH depen-           39. Hess P, Lansman JB, Tsien RW. Calcium channel selec-
    dency and desensitization kinetics of heterologously            tivity for divalent and monovalent cations. Voltage
    expressed combinations of acid-sensing ion channel              and concentration dependence of single channel cur-
    subunits. J Biol Chem 2004, 279:11006–11015.                    rent in ventricular heart cells. J Gen Physiol 1986, 88:
24. Krishtal O. The ASICs: signaling molecules? Modula-             293–319.
    tors? Trends Neurosci 2003, 26:477–483.                     40. Zhou W, Jones SW. Surface charge and calcium chan-
25. Grunder S, Chen X. Structure, function, and pharma-             nel saturation in bullfrog sympathetic neurons. J Gen
    cology of acid-sensing ion channels (ASICs): focus on           Physiol 1995, 105:441–462.
    ASIC1a. Int J Physiol Pathophysiol Pharmacol 2010,          41. Star RA. Nitric oxide. Am J Med Sci 1993, 306:
    2:73–94.                                                        348–358.
26. Bickler PE, Buck LT. Adaptations of vertebrate neu-         42. Schulz JB, Matthews RT, Klockgether T, Dichgans J,
    rons to hypoxia and anoxia: maintaining critical Ca2+           Beal MF. The role of mitochondrial dysfunction and
    concentrations. J Exp Biol 1998, 201:1141–1152.                 neuronal nitric oxide in animal models of neurodegen-
27. Chu XP, Papasian CJ, Wang JQ, Xiong ZG. Modulation              erative diseases. Mol Cell Biochem 1997, 174:193–197.
    of acid-sensing ion channels: molecular mechanisms          43. Bolanos JP, Almeida A. Roles of nitric oxide in brain
    and therapeutic potential. Int J Physiol Pathophysiol           hypoxia-ischemia. Biochim Biophys Acta 1999, 1411:
    Pharmacol 2011, 3:288–309.                                      415–436.

Volume 1, September/October 2012 © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.                                           661
Focus Article                                                                                               wires.wiley.com/mts

44. Boje KM, Arora PK. Microglial-produced nitric oxide         53. Rock DM, Macdonald RL. Polyamine regulation of
    and reactive nitrogen oxides mediate neuronal cell              N-methyl-d-aspartate receptor channels. Annu Rev
    death. Brain Res 1992, 587:250–256.                             Pharmacol Toxicol 1995, 35:463–482.
45. Cadiou H, Studer M, Jones NG, Smith E StJ, Ballard A,       54. Johnson TD. Polyamines and cerebral ischemia. Prog
    McMahan SB, McNaughton PA. Modulation of acid-                  Drug Res 1998, 50:193–258.
    sensing ion channel activity by nitric oxide. J Neurosci    55. Li J, Doyle KM, Tatlisumak T. Polyamines in the brain:
    2007, 27:13251–13260.                                           distribution, biological interactions, and their potential
46. Gingrich MB, Traynelis SF. Serine proteases and brain           therapeutic role in brain ischaemia. Curr Med Chem
    damage - is there a link? Trends Neurosci 2000,                 2007, 14:1807–1813.
    23:399–407.                                                 56. Babini E, Paukert M, Geisler HS, Grunder S. Alternative
47. Vivien D, Buisson A. Serine protease inhibitors: novel          splicing and interaction with di- and polyvalent cations
    therapeutic targets for stroke? J Cereb Blood Flow              control the dynamic range of acid-sensing ion channel
    Metab 2000, 20:755–764.                                         1 (ASIC1). J Biol Chem 2002, 277:41597–41603.
                                                                57. Duan B, Wang Y-Z, Yang T, Chu X-P, Yu Y, Huang
48. Poirot O, Vukicevic M, Boesch A, Kellenberger S. Selec-
                                                                    Y, Cao H, Hansen J, Simon RP, et al. Extracellular
    tive regulation of acid-sensing ion channel 1 by serine
                                                                    spermine exacerbates ischemic neuronal injury through
    proteases. J Biol Chem 2004, 279:38448–38457.
                                                                    sensitization of ASIC1a channels to extracellular acido-
49. Vukicevic M, Weder G, Boillat A, Boesch A, Kellen-              sis. J Neurosci 2011, 31:2101–2112.
    berger S. Trypsin cleaves acid-sensing ion channel 1a in    58. Pignataro G, Cuomo O, Esposito E, Sirabell R, Renzo
    a domain that is critical for channel gating. J Biol Chem       Di, Annunziato GL. ASIC1a contributes to neuro-
    2006, 281:714–722.                                              protection elicited by ischemic preconditioning and
50. Toninello A, Salvi M, Mondovi B. Interaction of bio-            postconditioning. Int J Physiol Pathophysiol Pharmacol
    logically active amines with mitochondria and their role        2011, 3:1–8.
    in the mitochondrial-mediated pathway of apoptosis.         59. Chai S, Li M, Branigan D, Xiong ZG, Simon RP. Activa-
    Curr Med Chem 2004, 11:2349–2374.                               tion of acid-sensing ion channel 1a (ASIC1a) by surface
51. Kindy MS, Hu Y, Dempsey RJ. Blockade of ornithine               trafficking. J Biol Chem 2010, 285:13002–13011.
    decarboxylase enzyme protects against ischemic brain        60. Allen NJ, Attwell D. Modulation of ASIC channels
    damage. J Cereb Blood Flow Metab 1994, 14:                      in rat cerebellar Purkinje neurons by ischaemia-related
    1040–1045.                                                      signals. J Physiol 2002, 543:521–529.
52. Benveniste M, Mayer ML. Multiple effects of sper-           61. Jetti SK, Swain SM, Majumder S, Chatterjee S, Poorn-
    mine on N-methyl-d-aspartic acid receptor responses             ima V , Bera AK. Evaluation of the role of nitric oxide
    of rat cultured hippocampal neurones. J Physiol 1993,           in acid sensing ion channel mediated cell death. Nitric
    464:131–163.                                                    Oxide 2010, 22:213–219.

662                                © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. Volume 1, September/October 2012
You can also read