Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 3.
Published in final edited form as: Clin Exp Neuroimmunol. 2015 Aug 3;6(3):245–263. doi: 10.1111/cen3.12237

Neuron–astrocyte interactions in neurodegenerative diseases: Role of neuroinflammation

Kakulavarapu V Rama Rao 1, Tammy Kielian 1
PMCID: PMC4629520  NIHMSID: NIHMS708301  PMID: 26543505

Abstract

Selective neuron loss in discrete brain regions is a hallmark of various neurodegenerative disorders, although the mechanisms responsible for this regional vulnerability of neurons remain largely unknown. Earlier studies attributed neuron dysfunction and eventual loss during neurodegenerative diseases as exclusively cell autonomous. Although cell-intrinsic factors are one critical aspect in dictating neuron death, recent evidence also supports the involvement of other central nervous system cell types in propagating non-cell autonomous neuronal injury during neurodegenerative diseases. One such example is astrocytes, which support neuronal and synaptic function, but can also contribute to neuroinflammatory processes through robust chemokine secretion. Indeed, aberrations in astrocyte function have been shown to negatively impact neuronal integrity in several neurological diseases. The present review focuses on neuroinflammatory paradigms influenced by neuron–astrocyte cross-talk in the context of select neurodegenerative diseases.

Keywords: Alzheimer’s disease, amyotrophic lateral sclerosis, astrocytes, Batten disease, cell autonomous, neuroinflammation, neurons, non-cell autonomous

Introduction

Many neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and neuronal ceroid lipofuscinosis, a family of inherited lysosomal storage disorders collectively referred to as Batten disease, are associated with neurodegeneration in select brain regions.13 For decades, neuron loss in many neurodegenerative diseases has been traditionally viewed as a cell-autonomous process. However, recent discoveries have shown that other central nervous system (CNS) cells can contribute to neuronal injury/death in these disorders in a non-cell autonomous manner, which has challenged the neuro-centric view of physiopathology in neurodegenerative diseases.49 Accordingly, intercellular communication between neurons and other CNS parenchymal populations, including astrocytes, microglia and oligodendrocytes, have been identified and the significance of these cell–cell communications in the context of neuroprotection or neurodegeneration has been extensively investigated.49 The present review focuses on the role of astrocytes in dictating neuronal health versus pathology, as other contributions in this special issue are dedicated to neuron–microglia and neuron–oligodendrocyte cross-talk.

Astrocytes are indispensable for neuron homeostasis in the CNS, and carry out a variety of functions ranging from bioenergetic and metabolic support of neurons, neurotransmitter clearance from the synaptic cleft after neuronal excitation, spatio-temporal buffering of extracellular K+, scavenging reactive oxygen species (ROS), regulation of cerebral blood flow, and providing trophic support to neurons by regulating axonal guidance and synaptogenesis.1016 Astrocytes are endowed with a complex machinery to execute these diverse functions by expressing neurotransmitter receptors and transporters, gap junctions/hemichannels, cell-specific enzymes for metabolic support (i.e. glutamine synthetase) as well as the capacity to secrete numerous factors into the extracellular milieu that exert paracrine effects on neurons.14,1722 Any perturbation in one or more of these astrocytic functions can profoundly affect neuronal homeostasis, as evidenced in acute and chronic neurodegenerative diseases.8,2328

Astrocyte activation and neuroinflammation

Reactive astrocytosis (astrocyte activation) is observed in nearly all neurodegenerative diseases, and is broadly defined as a defensive reaction by astrocytes in response to a variety of external stimuli, which accompanies either acute (i.e. traumatic brain injury, cerebral ischemia) or chronic neurological conditions (i.e. AD, PD, ALS, Batten disease).27,2933 On activation, astrocytes undergo morphological changes and alter their gene expression profiles. A major hallmark of reactive astrocytosis is increased expression of the intermediate filament proteins glial fibrillary acidic protein (GFAP) and vimentin.34 Although the functional implications of augmenting intermediate filament protein expression are unclear, this likely participates in limiting bystander damage to surrounding healthy parenchyma, as has been shown in stroke and spinal cord injury.3538

A number of signals can induce reactive astrocytosis that are likely context-dependent, including pathogen-associated molecular patterns (PAMP; such as lipopolysaccharide and peptidoglycan)3941 and danger-associated molecular patterns (DAMP), which are typically intracellular molecules that are sensed as danger signals when released from damaged/dying cells.42 In addition, emerging evidence supports the concept that astrocytes can also sense danger from within, triggered by mitochondrial damage or disruptions in autophagic pathways, which elicits a specialized intracellular response mediated by Nod-like receptors (NLR).4346 Typically, intracellular NLR engagement converges on inflammasome activation; however, the functional impact of NLR signaling in astrocytes remains a conundrum, as evidence suggests that highly purified astrocytes do not produce interleukin (IL)-1β,40,47 the best characterized inflammasome substrate.

Astrocytes can also perturb neuron homeostasis in the context of neurodegenerative diseases by exerting inflammatory effector functions.4857 Although astrocytes are not traditionally considered an inflammatory cell type, they are recognized as a robust source of chemokines in response to diverse stimuli,58 and by extension, play a critical role in peripheral leukocyte recruitment into the inflamed/injured CNS. In terms of sensing PAMP and DAMP, astrocytes express pattern recognition receptors, including various Toll-like receptors (TLR; TLR2, TLR3, TLR4 TLR5 and TLR9), although their repertoire is less extensive compared with microglia, the resident innate immune cell in the CNS parenchyma. Among the TLR detected in astrocytes, TLR3 is the most highly induced in response to pro-inflammatory cytokines (tumor necrosis factor-α, interleukin [IL]-1β and interferon-γ) as well as TLR agonists, including poly I:C, LPS, flagellin and CpG oligonucleotides (ODN),59,60 whereas TLR2 plays a pivotal role in recognizing Gram-positive bacteria, such as Staphylococcus aureus, as well as microbial lipoproteins.39,61,62 Astrocytes can also sense bacterial/viral DNA (as modeled by non-methylated CpG ODN) through TLR9, which also triggers an inflammatory cascade. In astrocytes, as in other cell types, all TLR transduce activation signals through the adaptor protein, MyD88, with the exception of TLR3, which utilizes TRIF, and TLR4, which can signal through either a MyD88-dependent or -independent pathway. TLR signaling leads to nuclear factor kappa-B and mitogen activated protein kinase activation, which in astrocytes primarily elicits the production of type I interferon (i.e. interferon-α and interferon-β) as well as a wide array of chemokines,6365 including CCL2, CCL5, CCL20, CXCL1, CXCL2, CXCL10 and CXCL12.59,66 These chemokines serve to promote immune cell recruitment from the systemic circulation and amply immune circuits within the CNS.66 Typically, the desired result is a swift and efficient inflammatory response to neutralize the offending pathogen/insult and promote rapid resolution/healing to restore CNS homeostasis. However, if inflammatory responses are perpetuated, this can lead to heightened or chronic inflammation, which can be detrimental to neuron survival. This highlights the need to achieve a delicate balance with regard to inflammation in the CNS, which can be influenced by astrocytes based on their central role in controlling neuron health and strategic positioning as sentinels at the blood–brain barrier.

While chemokine synthesis by astrocytes is widely accepted, whether they are a major source of pro-inflammatory cytokines remains a subject of debate. Although astrocytes have been shown to produce pro-inflammatory cytokines in vitro, including tumor necrosis factor-α and IL-1β,67,68 it remains possible that microglial contamination, which is a common occurrence in astrocyte cultures, could be influencing these responses. In support of this possibility, FACS-purified astrocytes did not show any evidence of pro-inflammatory cytokine production in response to the Gram-negative pathogen, Citrobacter koseri.40 Likewise, astrocyte responses to TLR2 and TLR3 agonists were greatly enhanced by microglia, and responses to TLR4 agonists were completely dependent on the presence of functional microglia in cultures.47 However, it should be noted that astrocytes are responsive to pro-inflammatory cytokines derived from external sources acting in a paracrine manner, as astrocytes express cell surface receptors for various pro-inflammatory cytokines. These cytokines, in turn, trigger the synthesis of a variety of chemokines and other inflammatory mediators from astrocytes, including prostaglandins, arachidonic acid, and ROS and reactive nitrogen species (RNS).37,48 The impact of reactive astrocytosis in the context of neuroinflammation will be discussed in the following sections in select neurodegenerative diseases, including AD, PD, ALS and forms of neuronal ceroid lipofuscinosis, collectively referred to as Batten disease. The rationale for selecting these neurodegenerative diseases is based on their shared characteristics of aggregate or storage material accumulation in different neural cell types.69

AD

AD represents the most common neurodegenerative disorder, with approximately 44 million people suffering from this disease worldwide and over 5 million people afflicted in the USA.70 Although most cases of AD are sporadic in etiology, familial forms of AD are inherited as an autosomal dominant trait with mutations in amyloid precursor protein, presenilin 1 and presenilin 2 identified as pathogenic factors.7174 Clinical symptoms of AD include short-term memory loss, difficulty in judgment, progressive cognitive decline, tactile agnosia and apraxia.71 AD is typified by the intracellular accumulation of neurofibrillary tangles (NFT) composed of hyperphosphorylated tau in neurons within the entorhinal cortex, hippocampus and neocortex,7578 as well as senile plaques composed of amyloid-β (Aβ).7981 Structural changes occurring in the AD brain include cortical atrophy with shrinkage of gyri and widening of sulci with enlarged ventricles in the frontal and temporal lobes.79,80

Neuronal loss in AD is moderate in cortical structures (26–30%), with up to 45% pyramidal neuron loss reported in AD patients, which correlates with the density of NFT and senile plaques.71,82,83 Neuronal loss is associated with reactive astrocytosis concomitant with a wide range of alterations in normal astrocyte functions (discussed in detail in the following sections).32,8486 The extent to which reactive astrocytes affect neuron function in AD remains unclear. However, Aβ treatment of neuron–astrocyte co-cultures caused a transient increase in astrocyte [Ca2+]i that elicited mitochondrial depolarization and ROS production, resulting in neuronal death.24,87,88 Likewise, Aβ augments glucose uptake in astrocytes, which increases glycogen deposition, H2O2 production and glutathione release. These changes in astrocyte metabolism elicited by Aβ exposure have been shown to impair neuronal viability and reduce synaptic proteins in neuron–astrocyte co-cultures89. Additionally, Aβ augmented glutamate release from astrocytes, leading to neuronal NMDA receptor stimulation and subsequent synaptic loss.90 Aβ has been shown to activate poly(ADP-ribose) polymerase in astrocytes, and cause neuronal death through oxidative stress.91 Furthermore, astrocytes participate in Aβ clearance,92 and Aβ degradation is impaired in reactive astrocytes, which might increase the risk for direct toxic effects of Aβ on neurons,93 although microglia are a major effector cell responsible for Aβ clearance.94,95

PD

PD is the second most common neurodegenerative disorder with an incidence of 120 per 100 000 persons by the age 70 years. Greater than 95% of PD is sporadic, without any genetic linkage or predisposition identified, although α-synuclein mutations have been linked to familial forms of PD.96,97 Clinical symptoms of PD range from tremors, rigidity, absence of voluntary movements, stooped posture and abnormal facial expressions as a result of muscle spasticity.98101

Prominent neuropathological features of PD include intraneuronal deposition of cytoplasmic inclusions called Lewy bodies and nigrostriatal dopaminergic neuron loss102104, which results in dopamine depletion in the brain.105107 Besides striatal dopaminergic neuron loss, PD is also occasionally associated with the death of noradrenergic and serotonergic neurons in the raphe nucleus, as well as cholinergic neurons in the entorhinal cortex.108,109 These changes are typically associated with robust astrocyte activation.27,31,110112 Multiple lines of evidence have shown that reactive astrocytes adversely affect neuronal function in the context of PD. For example, the engraftment of astrocytes expressing human brain derived neurotrophic factor into the striatum of a rat parkinsonism model reduced neurobehavioral deficits.113 Likewise, the astrocyte modulating agent 2-propyloctanoic acid not only reduced striatal dopaminergic neuron loss, but also prevented reactive astrocytosis in a rat model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonism,114 suggesting that astrocyte activation contributes to neuron loss in PD. 2-propyloctanoic acid is known to inhibit S100β expression and augment glutamate transporter expression, the latter of which might be envisioned to limit neurotoxicity by enhancing glutamate uptake. Additionally, 5-lipoxygenase activity was increased in reactive astrocytes after MPTP administration, which was suggested to inhibit dopamine uptake in striatal neurons, thereby rendering these cells sensitive to MPTP-mediated death.115 Likewise, suppression of inducible nitric oxide synthase and associated inflammatory events in astrocytes after MPTP injection protected striatal neurons from apoptosis.116 Collectively, these studies suggest that astrocytes can negatively impact neuronal survival in the context of PD. However, it should be noted that many of these findings were obtained with MPTP injection, which is an aggressive model that elicits rapid and robust dopaminergic neuron loss. Whether astrocytes negatively impact neuron integrity in other PD models where disease progression is more protracted in nature remains to be determined.

ALS

ALS is a progressive neurodegenerative disease that affects motor neurons in the cerebral cortex, brain stem and spinal cord, resulting in fatal paralysis.6,117 Clinical features of ALS include progressive peripheral muscle weakness, atrophy, and spasticity as a result of upper and lower motor neuron degeneration. The majority of ALS cases are sporadic (>90%), whereas a minor fraction are associated with mutations in Cu/Zn superoxide dismutase (SOD1). In ALS, SOD1 mutations result in protein misfolding and aggregate formation, which is thought to cause motor neuron death in both cell-autonomous and non-cell autonomous manners.56 With regard to the latter, neuroinflammation has recently gained traction as a prominent factor in ALS pathogenesis, with both microglia and astrocytes producing inflammatory mediators that impact motor neuron survival. For example, mutant SOD1 has been shown to trigger chemokine and cytokine production by astrocytes that are neurotoxic.118 Interestingly, expression of mutant human SOD1 in mouse astrocytes, but not motor neurons, elicited neuronal death by releasing soluble factors that induced BAX-dependent death.118 Likewise, co-cultures of motor neurons expressing wild-type human SOD1 with astrocytes transduced with mutant human SOD1 also caused significant motor neuron death119 typified by the loss of mitochondrial function and induction of mitochondrial permeability transition.120 Collectively, these studies highlight an important non-cell autonomous role for astrocytes in mediating motor neuron death in ALS.

Neuronal ceroid lipofuscinosis

Neuronal ceroid lipofuscinosis (NCL) encompasses a family of inherited disorders caused by mutations in CLN genes collectively referred to as Batten disease.121,122 To date, mutations in 14 different CLN genes have been identified that are broadly classified into infantile, late infantile, juvenile and adult onset forms.122,123 The childhood forms of Batten disease are characterized by blindness, behavioral deficits, seizures, and progressive cognitive and motor impairment that leads to premature death, with the infantile and late infantile forms showing the most aggressive progression.124,125 A histopathological hallmark of all NCL is the lysosomal accumulation of autofluorescent lipopigments and proteins,126 including subunit C of mitochondrial adenosine triphosphate (ATP) synthase (primarily in juvenile NCL) and sphingolipid pigments (in other forms of NCL).127129

Infantile NCL (INCL) is the most aggressive NCL form, with an average life expectancy of 2–6 years.130 INCL is caused by a mutation in CLN1, which encodes for palmitoyl protein thioesterase, an enzyme responsible for the cleavage of long-chain fatty acid residues on proteins containing cysteine moieties.131,132 Clinical symptoms of INCL can manifest as early as 6 months-of-age, which rapidly progress to severe motor and cognitive deficits, seizures, and premature death.125 Late infantile NCL (LINCL) is caused by mutations in CLN2 that encode the lysosomal enzyme tripeptidyl peptidase I, which cleaves tripeptides from the terminal amine groups of partially unfolded proteins.133,134 LINCL onset is typically delayed compared with the infantile form, with affected children generally succumbing to disease between the ages of 8–12 years. Other CLN mutations have been associated with variant late infantile NCL, including CLN5, CLN6 and CLN8, where comparatively less is known regarding gene function or associated pathophysiology. Juvenile NCL (JNCL) results from mutations in the CLN3 gene, for review, see125. The precise function of CLN3 remains unknown; however, based on functional analysis in yeast models and neuronal cell lines, CLN3 has been implicated in lysosomal acidification, endocytic and vesicle trafficking, and proper maintenance of mitochondrial function.135137 Similar to INCL and LINCL, JNCL also presents with visual impairment, seizures, and progressive cognitive and motor decline, but with a later onset, typically between 5–10 years-of-age,125 and the disease course is more protracted, with an average life expectancy between the late teens to early-mid 20s.

NCL are associated with neuronal loss in select brain regions, including the hippocampus, thalamus and various cortical areas.138140 Additionally, astrocyte activation is evident in each NCL form.141 In JNCL mouse models, studies have shown early prominent astrocyte activation, as indicated by increased GFAP expression, that precedes neuronal loss in these mice that does not occur until around 12 months-of-age.142,143 Astrocyte activation is also apparent in INCL, and in this setting astrocytes appear to play a protective role in disease, as CLN1/GFAP/vimentin triple mutant mice experienced an accelerated disease course typified by heightened cytokine/chemokine production.144 It remains unclear why disease progression was exacerbated when GFAP and vimentin were absent, as astrocytes themselves remained intact. This finding suggests that GFAP and vimentin loss alters astrocyte physiology or function, making them less adept at providing what appear to be beneficial roles, as disease severity was increased in CLN1/GFAP/vimentin mutant animals. Reactive astrocytes have also been described in LINCL and variant late infantile NCL forms (i.e. CLN5 and CLN6), which often precedes neurodegeneration.145149

Although a direct effect of dysfunctional astrocytes on neuronal loss in the various NCL forms is not yet established, a recent study from our group has shown a progressive decline in the expression of key molecules involved in glutamate homeostasis (i.e. GLAST and glutamine synthetase) in activated astrocytes in JNCL.143 By extension, this is expected to have a negative impact on neuron homeostasis and survival in the context of CLN3 mutation.

Potential factors that disrupt neuron-astrocyte interactions in neurodegenerative diseases

As aforementioned, astrocytes are indispensable for neuron homeostasis, as they carry out a variety of critical functions, including glutamate clearance at the synaptic cleft, release of gliotransmitters that provide trophic support to neurons, and supplying precursors for neuronal energy metabolism and anti-oxidant defense. Alterations in one or more of these astrocyte functions have been shown in several neurodegenerative diseases, which likely contribute to neuronal loss in these disorders. A few examples are discussed below.

Altered gliotransmission

Astrocytes express numerous neurotransmitter receptors, including glutamate, gamma-aminobutyric acid, acetylcholine, norepinephrine and adenosine,150 and constantly communicate with neighboring neurons through a variety of second messengers, such as Ca2+ and IP3.42 During intense synaptic activity, neurotransmitter release (in particular glutamate) can elicit increased [Ca2+]i in astrocytes that in turn, triggers glutamate release from astrocytes primarily through hemichannels. Hemichannel-mediated glutamate release feeds back to activate neurons to regulate synaptic transmission, thus maintaining a type of neurotransmission “synchrony” through Ca2+-glutamate oscillations between astrocytes and neurons.151153 Some of these events are reportedly altered during neurodegenerative disease. For example, Aβ was shown to specifically augment [Ca2+]i in astrocytes, but not neurons, and this rise in astrocytic [Ca2+]i induced neuronal mitochondrial permeability transition (mPT) by a mechanism involving ROS production.87 Likewise, Aβ was shown to induce glutamate release from astrocytes, leading to neuronal NMDA receptor activation and synapse loss.90

Defects in astrocyte glutamate uptake by either reduced glutamate transporter expression or activity could also cause neuronal death by excitotoxicity. For example, Aβ reduces astrocyte GLAST and GLT-1 levels,154 and decreased EAAT2 glutamate transporter expression has been observed in post-mortem AD brains.155 Astrocyte glutamate clearance was also impaired after MPTP exposure,156,157 as well as in a mouse model of PD.158 Loss of astrocyte glutamate transporters was also shown in ALS.159161 Astrocytes in a mouse model of JNCL show reduced GLAST and glutamine synthetase expression, revealing glutamate recycling defects in this disorder.143 Therefore, although these neurodegenerative diseases have distinct etiologies, impaired glutamate regulatory mechanisms appear to represent a unifying theme that might contribute to neuron loss, although this remains to be definitively determined.

Astrocyte hemichannels

Recent studies suggest that gliotransmitters can also be released by astrocytes through hemichannel (HC) opening.162,163 HC are composed of a hexameric ring of connexin or pannexin proteins; however, only the former are capable of forming gap junction channels when they align with adjacent HC on neighboring cells.164 HC have been reported to open under both physiological and pathological conditions to facilitate the release of various small m.w. metabolites and gliotransmitters from astrocytes, including ATP, glucose, glutathione, glutamate, gamma-aminobutyric acid and D-serine.163 Transient HC opening could be beneficial by maintaining molecular gradients across the astrocyte membrane; however, chronic HC activity can have deleterious effects not only on astrocytes by dissipating chemical gradients, but also on adjacent neurons thorough sustained ATP and D-Serine release, which can cause neuronal death.162,165 Astrocytes in a mouse model of JNCL showed a transient increase in HC opening at an early postnatal age (1 month) that significantly preceded neuronal loss, which occurs around 12 months in this model.143 HC blockade using the novel carbenoxolone-based inhibitor, INI-0602, enhanced gap junction communication, and led to significant reductions in lysosomal storage material in the brains of CLN3 mutant mice.143 Increased astrocytic HC activity has also been observed in a mouse model of AD,166 and exogenous Aβ application in acute brain slices augmented astrocyte HC activity, resulting in increased ATP and glutamate release and neuronal death.167 Furthermore, inflammatory cytokines that are elevated in various neurodegenerative diseases (i.e. IL-1β, tumor necrosis factor-α) significantly increase astrocyte HC activity, again reinforcing the interplay between neuroinflammation and astrocytic dysfunction and downstream implications on neurodegeneration.168,169172

Purinergic receptors

Purinergic receptors play a pivotal role in neuron–astrocyte communication, as ATP and glutamate release mediated by astrocyte HC opening and metabotropic purinergic receptors (P2YR subtypes) trigger neuronal activity, thereby completing the glutamate neurotransmission loop between neurons and astrocytes.173175 However, excessive ATP release from astrocytes could exert autocrine effects by stimulating purinergic receptors to induce prostaglandin and nitric oxide production,176,177 which would disrupt astrocyte–neuron interactions. Alternatively, sustained ATP release can stimulate microglial purinergic receptors to elicit pro-inflammatory responses.178 In the light of these events, purinergic receptor activation has been observed in various neurodegenerative diseases including AD, PD and ALS,179182 and purinergic receptor antagonists have generally showed beneficial effects in counteracting neuroinflammation in these diseases.183 Furthermore, synaptophysin loss and memory dysfunction were significantly reduced in adenosine receptor 2A (A2A) KO mice after intracerebral Aβ administration.184 Likewise, microglia isolated from P2X(7) receptor KO mice produced less IL-1β after Aβ treatment.185 Interestingly, conditional A2A receptor deletion in forebrain neurons improved motor incoordination in a mouse MPTP model of Parkinson’s disease, whereas striatal dopamine levels remained low. Intracerebral administration of KW-6002, an A2A receptor antagonist, significantly attenuated the MPTP-induced loss of striatal dopamine levels as well as microglial and astrocyte activation, suggesting that glial purinergic receptors are critical for exerting toxic effects in PD.186 In contrast to these findings, a recent study reported a somewhat unexpected result in the SOD1-G93A model of ALS, where P2X7 receptor ablation exacerbated reactive gliosis and motor neuron death, suggesting that purinergic receptors can also exert neuroprotective effects in select neurodegenerative disorders, at least during some stages of disease progression.187

D-serine

D-serine, an endogenous NMDA receptor agonist, has been identified as an important gliotransmitter in neuron–astrocyte interactions with respect to glutamatergic neurotransmission.188,189. D-serine is synthesized from L-serine in a reaction mediated by serine racemase (SR), whose activity is highly enriched in neurons.190 However, as neurons are unable to synthesize L-serine from glucose, L-serine is provided by astrocytes, which abundantly synthesize L-serine from glucose by a reaction catalyzed by D-3-phosphoglycerate dehydrogenase.191 Once released from astrocytes, L-serine is internalized by neurons and is converted to D-serine by SR for export back to astrocytes (through the serine shuttle) for storage.192 Therefore, astrocytes have an abundant D-serine pool and regulate its release to activate neuronal NMDA receptors in a controlled manner. While regulated D-serine release from astrocytes plays an important role in long-term potentiation, an important event in learning and memory,193 excessive D-serine release has been implicated in neuronal excitotoxicity through a NMDA-dependent mechanism in neurodegenerative diseases.194,195 An interesting study reported that Aβ-induced NMDA-mediated excitotoxicity was attenuated in SR knockout mice, which highlights the importance of D-serine in neuronal excitotoxic mechanisms under the influence of astrocytes. Furthermore, SR activity and D-serine levels were increased in SOD1 transgenic mice, resulting in enhanced NMDA-mediated excitotoxic death of neurons.196,197

Neuron–astrocyte interactions in bioenergetic/metabolic regulation

The main bioenergetic/metabolic pathways utilized by astrocytes and neurons differ. Specifically, astrocytes rely heavily on glycolysis in response to neuron-derived glutamate and the process of glutamate reuptake is a highly energy demanding process, whereas neuronal energetic demands shift them more towards ATP-generating pathways, such as oxidative phosphorylation.198 The energetic crosstalk between astrocytes and neurons will be discussed in the following section with examples provided from AD, PD, ALS and NCL where available.

Astrocyte–neuron lactate shuttle

Bioenergetic and metabolic communication exists between neurons and astrocytes through glucose/lactate exchange. Lactate is predominantly, if not exclusively, produced in astrocytes, and glutamate plays a central role in this process.199 For example, during neurotransmission, neurons release glutamate into the synaptic cleft, which is rapidly cleared by astrocytes through selective glutamate transporters along with the release of 3 Na+ ions. Na+ release is mediated by Na+/K+ ATPase activity, which consumes 1 ATP to extrude 3 Na+ ions from astrocytes. The majority of glutamate internalized by astrocytes is converted to glutamine, while a portion is diverted to the TCA cycle through conversion to α-ketoglutarate.200 This process of glutamate–glutamine conversion is energy-demanding, and to meet these energy demands, astrocytes enhance glucose uptake and stimulate glycolysis to produce pyruvate, which is later converted to lactate and transported to neurons through neuron-specific monocarboxylate transporter 4.201,187

Although the astrocyte neuron lactate shuttle has been historically accepted as the model whereby astrocyte-derived lactate functions as a major energy fuel for neurons, in recent years, the astrocyte neuron lactate shuttle mechanism has been a subject of debate.202,203 Accordingly, it has been argued that stimulation of glycolysis to produce lactate during intense neuronal activity in response to glutamatergic neurotransmission is not necessarily operative in all types of astrocytes. Furthermore, neurons do possess an inherent capacity to increase glucose uptake and channel glucose to glycolysis to sustain their energy demands during neuronal excitation.202,203 Based on the differential kinetic properties of glucose and lactate transporters in neurons (glucose transport capacity being higher in neurons),204,205 it has been proposed that neurons actively take up glucose and release lactate for astrocytes,202,203 a phenomenon that appears to oppose the astrocyte neuron lactate shuttle.199 This possibility has been counter argued by Magistretti’s group in that the mere examination of glucose transporter capacity being higher in neurons might not translate into neurons having increased glucose utilization rates than astrocytes.206 This has also been supported by studies where depletion of the neuron-selective glucose transporter, GLUT3, in Tg mice did not show changes in glucose utilization,207 whereas, deletion of the astrocytic glucose transporter, GLUT1, resulted in a severe neurological phenotype.208 Perhaps this controversy could be resolved by extending studies integrating in vivo and in vitro metabolomics together with thorough investigations into inherited metabolic disorders associated with glucose metabolism.

Despite controversies with regard to astrocytic and neuronal glucose/lactate metabolism, defective neuron–astrocyte metabolic coupling has been reported in various neurodegenerative diseases.32,209211 For example, a significant reduction in brain glucose uptake and cerebral glucose metabolic rate was reported in AD patients.212 Likewise, cerebral glucose uptake was reduced in an AD transgenic mouse model, which resulted in reduced cerebral lactate release, suggesting impaired astrocyte glycolysis in AD that can have profound effects on neuronal energy metabolism.213 This is further supported by reduced monocarboxylic acid transporter expression in astrocytes214, revealing multiple defects in neuron-astrocytic metabolic coupling in AD. Additionally, several glycolytic and citric acid cycle metabolites were reduced in an AD transgenic rat model,215 and various energy metabolites, including glucose, lactate and other glycolytic intermediates, were also reduced in the CSF of PD patients.216 Interestingly, in vitro exposure of dopamergic neurons to CSF from PD patients resulted in increased tyrosine hydroxylase expression and subsequent neuron death, as shown by increased LDH release,217 which is also consistent with defective neuron–astrocyte metabolic cross-talk in PD. Metabolic dysfunction involving the lactate shuttle has also been reported in a transgenic ALS mouse model, as shown by reduced astrocytic lactate transporter expression and concomitant reduction in lactate levels, as well as a reduction in phosphoglycerate kinase, a rate-limiting enzyme of glycolysis.218 Magnetic resonance spectroscopy in patients with various NCL forms showed a marked elevation of brain myo-inositol (astrocytic marker) and lactate levels,219 the latter suggestive of a neuronal energy metabolic block. Additionally, increased levels of phosphofructo kinase, likely translating to increased glycolysis, was shown in the brains of CLN3Δex7/8 mice, a model of JNCL, suggesting compensatory mechanisms in astrocytic energy metabolism.220 Furthermore, deletion of btn1, the yeast orthologue of CLN3, showed increased glycolysis.221

Astrocytic glycogen metabolism

Astrocytes also exert metabolic crosstalk with neurons through glycogen metabolism. Glycogen is the storage form of glucose and is readily mobilized in astrocytes to produce glucose-6-phosphate, which is further channeled into glycolysis to yield lactate.222 Glycogenolysis is regulated by neuronal vasoactive intestinal peptide and noradrenaline to provide lactate in response to increasing neuron metabolic demands.222 Studies have shown that astrocytic glycogen contributes to learning and memory.223 Alterations in astrocyte glycogen metabolism have been reported in AD and multiple sclerosis, where fluoxetine, which improves neuronal energy metabolism by promoting astrocytic glycogenolysis, has been shown to reduce the number of tremors and significantly enhance cerebral blood flow in PD patients. Fluoxetine also significantly improved memory and cognitive deficits in AD patients.224,225 It is notable that various inflammatory mediators, including cytokines, chemokines, prostaglandins and ROS, are known to alter neuron–astrocyte metabolic coupling.86 Accordingly, astrocytes exposed to inflammatory cytokines failed to deliver anti-inflammatory precursors, such as cysteinylglycine for GSH synthesis in neurons.226 Likewise, cytokines were shown to augment glucose uptake in astrocytes, which was not reflected by enhanced lactate levels, suggesting that cytokines perturb downstream events of glucose metabolism in astrocytes, resulting in less lactate production and release that can negatively impact neuronal survival.227,228 Inflammatory cytokines also enhance glutathione release from astrocytes and concomitantly induce superoxide production, representing a “double whammy” to impair astrocyte anti-oxidant activity.227

Oxidative stress (OS) is another deleterious factor in the setting of neuroinflammation that can compromise neuron–astrocyte metabolic interactions.209,229,230 Exaggerated ROS production during aberrant metabolic reactions in either astrocytes or neurons can elicit DNA, RNA and protein damage.231 Increased protein carbonyls were consistently found in AD patients in both neurons and astrocytes with pronounced accumulation in non-synaptic and synaptic mitochondria.232 Additionally, increased carbonylation of metabolic enzymes in the AD brain, including creatine kinase, pyruvate kinase, enolase, phosphoglycerate mutase, lactate dehydrogenase and ATP synthase, supports the possibility of perturbed glycolysis in astrocytes and concomitant downstream impairments on ATP production.233 Such oxidative inactivation of LDH and other energy generating enzymes, such as glyceraldehyde 3-phosphate dehydrogenase and enolase, has also been observed in PD and ALS.234236 Collectively, these reports strongly support the concept that various inflammatory mechanisms elicited during neurodegenerative diseases can perturb neuron–astrocyte metabolic coupling and contribute to neurodegeneration.

Mitochondrial dysfunction in neurodegenerative diseases

Mitochondrial dysfunction is a characteristic feature of numerous neurodegenerative diseases.209,237,238 For example, Aβ has been shown to selectively enhance [Ca2+]i in astrocytes, but not neurons, which induced mPT opening and activation of the DNA repair enzyme Poly(ADP ribose) polymerase. These astrocytic changes were shown to induce oxidative stress in neurons, and inhibition of astrocytic mPT with cyclosporine A significantly attenuated neuronal death,87 suggesting that abnormal astrocytic function contributes to neuronal loss in AD. Similarly, astrocytes derived from DJ-1 knockout mice (a model of familial PD) showed severe mitochondrial abnormalities, including reduced mitochondrial motility, mPT induction and failure to protect neurons from oxidative injury and death,239241 showing that astrocytic mitochondrial abnormalities impair neuroprotective responses in PD. Likewise, astrocytes derived from mutant SOD1 mice showed mitochondrial dysfunction, which translated to less neuroprotective activity. Interestingly, overexpression of mitochondrial-targeted catalase significantly improved mitochondrial function in SOD1 mutant astrocytes and prevented motor neuron death.242 Furthermore, Nrf2 activation and glutathione induction in astrocytes protected motor neurons from oxidative injury and apoptosis.243 Mitochondrial abnormalities have also been reported in neurons from a mouse model of JNCL (CLN3−/−), reflected by decreased oxygen consumption and mitochondrial electron transport chain enzymes.244 Similarly, enlarged mitochondria were observed in a neuronal cerebellar granular cell line derived from JNCL mice.129 Although astrocytic mitochondrial dysfunction in NCL has not yet been reported, it should be noted that astrocyte activation is observed in all NCL forms, and mitochondrial dysfunction is a common denominator in reactive astrocytes, suggesting a potential link between the two (see review Rama Rao and Kielian, 2015).

Astrocytes and neuronal trophic support

As aforementioned, a main function of astrocytes is to provide trophic support to neurons.245,246 This is achieved, in part, by the synthesis and secretion of various soluble factors by astrocytes, including extracellular matrix proteins (matricellular proteins, such as thrombospondins, hevins and glypicans), cholesterol and tenascin, as well as growth factors (transforming growth factor-β, CNTF, S100B) that bind to neuronal receptors and engage signaling pathways to synthesize/recruit proteins involved in synapse modeling.247249 Although the involvement of astrocyte-derived soluble factors on neuronal synaptic integrity has not been extensively investigated, studies have shown that astrocyte matricellular proteins and growth factor production is impaired in neurodegenerative diseases where astrocytes have been implicated in the loss of synaptic integrity.248,250,251 For example, AD post-mortem brains showed reduced thrombospondin-1 (TSP-1) levels,252 and Aβ treatment of astrocytes in vivo augmented ADAMTS-4, which contains a TSP-1 motif.253 A recent study showed that Aβ1–42 augmented intracellular TSP-1 levels in astrocytes concomitant with reduced extracellular TSP-1 protein, suggesting aberrant TSP-1 trafficking.251 Furthermore, conditioned medium from Aβ-treated astrocytes caused a significant reduction in synaptophysin and PSD95 expression in primary neurons,251 suggesting that impaired astrocytic trophic support in the context of AD might impact neuronal synaptic integrity. Similar to what has been observed after astrocyte exposure to Aβ, astrocytes from STAT3 KO mice released less TSP-1, which led to a reduction in synaptophysin and PSD95 levels in motor neurons, suggesting that astrocytic TSP-1 functions to promote motor neuron survival and synaptic integrity.254 It is interesting that various inflammatory signals, such as oxidative stress, prostaglandins and cytokines, also impair astrocyte TSP-1 synthesis.255257 Collectively, these findings suggest that the loss of critical astrocyte trophic support negatively impacts neuronal survival during neurodegenerative diseases.

Conclusions

Neurodegenerative diseases are debilitating neurological disorders typically accompanied by neuronal loss in select brain regions that can manifest as memory loss, movement disorders, paralysis and/or severe cognitive deficits. Both cell-autonomous and non-cell autonomous pathways have been implicated in mediating neuronal death in these disorders, and emerging evidence strongly supports a role for astrocytic dysfunction affecting neuronal loss in a non-cell autonomous manner by inducing neuroinflammatory responses (Fig. 1). Therefore, targeting astrocyte dysfunction in these diseases might have a therapeutic benefit to counteract neuropathology.

Figure 1.

Figure 1

Potential factors contributing to the disruption of neuron–astrocyte interactions in neurodegenerative diseases. Astrocyte dysfunction likely arising from reactive astrocytosis contributes to neuron death in a non-cell autonomous manner through the release of damaging molecules (glutamate [Glu], adenosine triphosphate [ATP], D-serine [D-ser]) as well as inflammatory mediators, such as chemokines. The majority of these molecules also act on astrocytes in an autocrine manner and exacerbate astrocyte dysfunction. Furthermore, aberrant synthesis/release of matricellular proteins, such as thrombospondins (TSP), as well as neuronal energy precursors (lactate) from astrocytes could severely curtail trophic support to neurons and disrupt energy metabolic coupling between neurons and astrocytes. In addition, accumulation of storage materials within neurons also directly regulates cell death in a cell autonomous manner. Aβ, amyloid-β; α-Syn, α-synuclein; EAAT2, glial excitatory amino acid transporter 2; Gln, glutamine, GS, glutamine synthetase; HC, hemichannels; m-SOD1, mutant superoxide dismutase; MCT1&2, monocarboxylic acid transporters 1 & 2; NFT, neurofibrillary tangles; NMDA, N-methyl-D-aspartate; P2Y & P2X2, purinergic receptors; ROS, reactive oxygen species; TSP-11/2, thrombospondins-1/2.

Acknowledgments

This work was supported by the National Institute of Neurological Disorders and Stroke (NINDS) 1R21NS084392-01A1 to T.K.

References

  • 1.Whitehouse PJ, Struble RG, Hedreen JC, Clark AW, Price DL. Alzheimer’s disease and related dementias: selective involvement of specific neuronal systems. CRC Crit Rev Clin Neurobiol. 1985;1:319–339. [PubMed] [Google Scholar]
  • 2.Graybiel AM, Hirsch EC, Agid Y. The nigrostriatal system in Parkinson’s disease. Adv Neurol. 1990;53:17–29. [PubMed] [Google Scholar]
  • 3.Cleveland DW, Bruijn LI, Wong PC, Marszalek JR, Vechio JD, Lee MK, et al. Mechanisms of selective motor neuron death in transgenic mouse models of motor neuron disease. Neurology. 1996;47:S54–61. doi: 10.1212/wnl.47.4_suppl_2.54s. discussion S61–52. [DOI] [PubMed] [Google Scholar]
  • 4.Ilieva H, Polymenidou M, Cleveland DW. Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J Cell Biol. 2009;187:761–772. doi: 10.1083/jcb.200908164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wallis N, Zagami CJ, Beart PM, O’Shea RD. Combined excitotoxic-oxidative stress and the concept of non-cell autonomous pathology of ALS: insights into motoneuron axonopathy and astrogliosis. Neurochem Int. 2012;61:523–530. doi: 10.1016/j.neuint.2012.02.026. [DOI] [PubMed] [Google Scholar]
  • 6.Zhao W, Xie W, Le W, Beers DR, He Y, Henkel JS, et al. Activated microglia initiate motor neuron injury by a nitric oxide and glutamate-mediated mechanism. J Neuropathol Exp Neurol. 2004;63:964–977. doi: 10.1093/jnen/63.9.964. [DOI] [PubMed] [Google Scholar]
  • 7.Di Malta C, Fryer JD, Settembre C, Ballabio A. Astrocyte dysfunction triggers neurodegeneration in a lysosomal storage disorder. Proc Natl Acad Sci U S A. 2012;109:E2334–2342. doi: 10.1073/pnas.1209577109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Van Den Bosch L, Robberecht W. Crosstalk between astrocytes and motor neurons: what is the message? Exp Neurol. 2008;211:1–6. doi: 10.1016/j.expneurol.2008.01.008. [DOI] [PubMed] [Google Scholar]
  • 9.Marchetti B, L’Episcopo F, Morale MC, Tirolo C, Testa N, Caniglia S, et al. Uncovering novel actors in astrocyte-neuron crosstalk in Parkinson’s disease: the Wnt/beta-catenin signaling cascade as the common final pathway for neuroprotection and self-repair. Eur J Neurosci. 2013;37:1550–1563. doi: 10.1111/ejn.12166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hertz L. Functional interactions between neurons and astrocytes I. Turnover and metabolism of putative amino acid transmitters. Prog Neurobiol. 1979;13:277–323. doi: 10.1016/0301-0082(79)90018-2. [DOI] [PubMed] [Google Scholar]
  • 11.Magistretti PJ, Sorg O, Yu N, Martin JL, Pellerin L. Neurotransmitters regulate energy metabolism in astrocytes: implications for the metabolic trafficking between neural cells. Dev Neurosci. 1993;15:306–312. doi: 10.1159/000111349. [DOI] [PubMed] [Google Scholar]
  • 12.Bezzi P, Vesce S, Panzarasa P, Volterra A. Astrocytes as active participants of glutamatergic function and regulators of its homeostasis. Adv Exp Med Biol. 1999;468:69–80. doi: 10.1007/978-1-4615-4685-6_6. [DOI] [PubMed] [Google Scholar]
  • 13.Vesce S, Bezzi P, Volterra A. The highly integrated dialogue between neurons and astrocytes in brain function. Sci Prog. 1999;82(Pt 3):251–270. doi: 10.1177/003685049908200304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Newman EA. New roles for astrocytes: regulation of synaptic transmission. Trends Neurosci. 2003;26:536–542. doi: 10.1016/S0166-2236(03)00237-6. [DOI] [PubMed] [Google Scholar]
  • 15.Ehlers MD. Synapse formation: astrocytes spout off. Curr Biol. 2005;15:R134–R137. doi: 10.1016/j.cub.2005.02.013. [DOI] [PubMed] [Google Scholar]
  • 16.Ransom BR, Ransom CB. Astrocytes: multitalented stars of the central nervous system. Methods Mol Biol. 2012;814:3–7. doi: 10.1007/978-1-61779-452-0_1. [DOI] [PubMed] [Google Scholar]
  • 17.Vesce S, Rossi D, Brambilla L, Volterra A. Glutamate release from astrocytes in physiological conditions and in neurodegenerative disorders characterized by neuroinflammation. Int Rev Neurobiol. 2007;82:57–71. doi: 10.1016/S0074-7742(07)82003-4. [DOI] [PubMed] [Google Scholar]
  • 18.Dodla MC, Mumaw J, Stice SL. Role of astrocytes, soluble factors, cells adhesion molecules and neurotrophins in functional synapse formation: implications for human embryonic stem cell derived neurons. Curr Stem Cell Res Ther. 2010;5:251–260. doi: 10.2174/157488810791824520. [DOI] [PubMed] [Google Scholar]
  • 19.Hamilton NB, Attwell D. Do astrocytes really exocytose neurotransmitters? Nat Rev Neurosci. 2010;11:227–238. doi: 10.1038/nrn2803. [DOI] [PubMed] [Google Scholar]
  • 20.Gimsa U, Mitchison NA, Brunner-Weinzierl MC. Immune privilege as an intrinsic CNS property: astrocytes protect the CNS against T-cell-mediated neuroinflammation. Mediators Inflamm. 2013;2013:320519. doi: 10.1155/2013/320519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chung WS, Allen NJ, Eroglu C. Astrocytes Control Synapse, Formation, Function and Elimination. Cold Spring Harb Perspect Biol. 2015 doi: 10.1101/cshperspect.a020370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Volterra A, Meldolesi J. Astrocytes, from brain glue to communication elements: the revolution continues. Nat Rev Neurosci. 2005;6:626–640. doi: 10.1038/nrn1722. [DOI] [PubMed] [Google Scholar]
  • 23.Avila-Munoz E, Arias C. When astrocytes become harmful: functional and inflammatory responses that contribute to Alzheimer’s disease. Ageing Res Rev. 2014;18:29–40. doi: 10.1016/j.arr.2014.07.004. [DOI] [PubMed] [Google Scholar]
  • 24.Abramov AY, Canevari L, Duchen MR. Calcium signals induced by amyloid beta peptide and their consequences in neurons and astrocytes in culture. Biochim Biophys Acta. 2004;1742:81–87. doi: 10.1016/j.bbamcr.2004.09.006. [DOI] [PubMed] [Google Scholar]
  • 25.Sidoryk-Wegrzynowicz M, Wegrzynowicz M, Lee E, Bowman AB, Aschner M. Role of astrocytes in brain function and disease. Toxicol Pathol. 2011;39:115–123. doi: 10.1177/0192623310385254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rappold PM, Tieu K. Astrocytes and therapeutics for Parkinson’s disease. Neurotherapeutics. 2010;7:413–423. doi: 10.1016/j.nurt.2010.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Episcopo FL, Tirolo C, Testa N, Caniglia S, Morale MC, Marchetti B. Reactive astrocytes are key players in nigrostriatal dopaminergic neurorepair in the MPTP mouse model of Parkinson’s disease: focus on endogenous neurorestoration. Curr Aging Sci. 2013;6:45–55. doi: 10.2174/1874609811306010007. [DOI] [PubMed] [Google Scholar]
  • 28.Barbeito LH, Pehar M, Cassina P, Vargas MR, Peluffo H, Viera L, et al. A role for astrocytes in motor neuron loss in amyotrophic lateral sclerosis. Brain Res Brain Res Rev. 2004;47:263–274. doi: 10.1016/j.brainresrev.2004.05.003. [DOI] [PubMed] [Google Scholar]
  • 29.Pike CJ, Cummings BJ, Monzavi R, Cotman CW. Beta-amyloid-induced changes in cultured astrocytes parallel reactive astrocytosis associated with senile plaques in Alzheimer’s disease. Neuroscience. 1994;63:517–531. doi: 10.1016/0306-4522(94)90547-9. [DOI] [PubMed] [Google Scholar]
  • 30.Feeney SJ, McKelvie PA, Austin L, Jean-Francois MJ, Kapsa R, Tombs SM, et al. Presymptomatic motor neuron loss and reactive astrocytosis in the SOD1 mouse model of amyotrophic lateral sclerosis. Muscle Nerve. 2001;24:1510–1519. doi: 10.1002/mus.1176. [DOI] [PubMed] [Google Scholar]
  • 31.Carbone M, Duty S, Rattray M. Riluzole neuroprotection in a Parkinson’s disease model involves suppression of reactive astrocytosis but not GLT-1 regulation. BMC Neurosci. 2012;13:38. doi: 10.1186/1471-2202-13-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Steele ML, Robinson SR. Reactive astrocytes give neurons less support: implications for Alzheimer’s disease. Neurobiol Aging. 2012;33:423 e421–413. doi: 10.1016/j.neurobiolaging.2010.09.018. [DOI] [PubMed] [Google Scholar]
  • 33.Charron G, Doudnikoff E, Canron MH, Li Q, Vega C, Marais S, et al. Astrocytosis in parkinsonism: considering tripartite striatal synapses in physiopathology? Front Aging Neurosci. 2014;6:258. doi: 10.3389/fnagi.2014.00258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hol EM, Pekny M. Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system. Curr Opin Cell Biol. 2015;32C:121–130. doi: 10.1016/j.ceb.2015.02.004. [DOI] [PubMed] [Google Scholar]
  • 35.Pekny M, Eliasson C, Siushansian R, Ding M, Dixon SJ, Pekna M, et al. The impact of genetic removal of GFAP and/or vimentin on glutamine levels and transport of glucose and ascorbate in astrocytes. Neurochem Res. 1999;24:1357–1362. doi: 10.1023/a:1022572304626. [DOI] [PubMed] [Google Scholar]
  • 36.Li L, Lundkvist A, Andersson D, Wilhelmsson U, Nagai N, Pardo AC, et al. Protective role of reactive astrocytes in brain ischemia. J Cereb Blood Flow Metab. 2008;28:468–481. doi: 10.1038/sj.jcbfm.9600546. [DOI] [PubMed] [Google Scholar]
  • 37.Burda JE, Sofroniew MV. Reactive gliosis and the multicellular response to CNS damage and disease. Neuron. 2014;81:229–248. doi: 10.1016/j.neuron.2013.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Pekny M, Pekna M. Astrocyte reactivity and reactive astrogliosis: costs and benefits. Physiol Rev. 2014;94:1077–1098. doi: 10.1152/physrev.00041.2013. [DOI] [PubMed] [Google Scholar]
  • 39.Esen N, Tanga FY, DeLeo JA, Kielian T. Toll-like receptor 2 (TLR2) mediates astrocyte activation in response to the Gram-positive bacterium Staphylococcus aureus. J Neurochem. 2004;88:746–758. doi: 10.1046/j.1471-4159.2003.02202.x. [DOI] [PubMed] [Google Scholar]
  • 40.Liu S, Kielian T. MyD88 is pivotal for immune recognition of Citrobacter koseri and astrocyte activation during CNS infection. J Neuroinflammation. 2011;8:35. doi: 10.1186/1742-2094-8-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Vincent AJ, Choi-Lundberg DL, Harris JA, West AK, Chuah MI. Bacteria and PAMPs activate nuclear factor kappaB and Gro production in a subset of olfactory ensheathing cells and astrocytes but not in Schwann cells. Glia. 2007;55:905–916. doi: 10.1002/glia.20512. [DOI] [PubMed] [Google Scholar]
  • 42.Rossi D, Volterra A. Astrocytic dysfunction: insights on the role in neurodegeneration. Brain Res Bull. 2009;80:224–232. doi: 10.1016/j.brainresbull.2009.07.012. [DOI] [PubMed] [Google Scholar]
  • 43.Silverman WR, de Rivero Vaccari JP, Locovei S, Qiu F, Carlsson SK, Scemes E, et al. The pannexin 1 channel activates the inflammasome in neurons and astrocytes. J Biol Chem. 2009;284:18143–18151. doi: 10.1074/jbc.M109.004804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Minkiewicz J, de Rivero Vaccari JP, Keane RW. Human astrocytes express a novel NLRP2 inflammasome. Glia. 2013;61:1113–1121. doi: 10.1002/glia.22499. [DOI] [PubMed] [Google Scholar]
  • 45.Liu L, Chan C. IPAF inflammasome is involved in interleukin-1beta production from astrocytes, induced by palmitate; implications for Alzheimer’s Disease. Neurobiol Aging. 2014;35:309–321. doi: 10.1016/j.neurobiolaging.2013.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, et al. NLRP3 Inflammasome Is Expressed and Functional in Mouse Brain Microglia but Not in Astrocytes. PLoS One. 2015;10:e0130624. doi: 10.1371/journal.pone.0130624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Holm TH, Draeby D, Owens T. Microglia are required for astroglial Toll-like receptor 4 response and for optimal TLR2 and TLR3 response. Glia. 2012;60:630–638. doi: 10.1002/glia.22296. [DOI] [PubMed] [Google Scholar]
  • 48.Sofroniew MV. Multiple roles for astrocytes as effectors of cytokines and inflammatory mediators. Neuroscientist. 2014;20:160–172. doi: 10.1177/1073858413504466. [DOI] [PubMed] [Google Scholar]
  • 49.Liu C, Cui G, Zhu M, Kang X, Guo H. Neuroinflammation in Alzheimer’s disease: chemokines produced by astrocytes and chemokine receptors. Int J Clin Exp Pathol. 2014;7:8342–8355. [PMC free article] [PubMed] [Google Scholar]
  • 50.Franceschi C, Valensin S, Lescai F, Olivieri F, Licastro F, Grimaldi LM, et al. Neuroinflammation and the genetics of Alzheimer’s disease: the search for a pro-inflammatory phenotype. Aging (Milano) 2001;13:163–170. doi: 10.1007/BF03351475. [DOI] [PubMed] [Google Scholar]
  • 51.Eikelenboom P, Bate C, Van Gool WA, Hoozemans JJ, Rozemuller JM, Veerhuis R, et al. Neuroinflammation in Alzheimer’s disease and prion disease. Glia. 2002;40:232–239. doi: 10.1002/glia.10146. [DOI] [PubMed] [Google Scholar]
  • 52.McGeer PL, McGeer EG. Local neuroinflammation and the progression of Alzheimer’s disease. J Neurovirol. 2002;8:529–538. doi: 10.1080/13550280290100969. [DOI] [PubMed] [Google Scholar]
  • 53.Tilleux S, Hermans E. Neuroinflammation and regulation of glial glutamate uptake in neurological disorders. J Neurosci Res. 2007;85:2059–2070. doi: 10.1002/jnr.21325. [DOI] [PubMed] [Google Scholar]
  • 54.Mosley RL, Benner EJ, Kadiu I, Thomas M, Boska MD, Hasan K, et al. Neuroinflammation, Oxidative Stress and the Pathogenesis of Parkinson’s Disease. Clin Neurosci Res. 2006;6:261–281. doi: 10.1016/j.cnr.2006.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Przedborski S. Neuroinflammation and Parkinson’s disease. Handb Clin Neurol. 2007;83:535–551. doi: 10.1016/S0072-9752(07)83026-0. [DOI] [PubMed] [Google Scholar]
  • 56.Hooten KG, Beers DR, Zhao W, Appel SH. Protective and Toxic Neuroinflammation in Amyotrophic Lateral Sclerosis. Neurotherapeutics. 2015;12:364–375. doi: 10.1007/s13311-014-0329-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Rizzo F, Riboldi G, Salani S, Nizzardo M, Simone C, Corti S, et al. Cellular therapy to target neuroinflammation in amyotrophic lateral sclerosis. Cell Mol Life Sci. 2014;71:999–1015. doi: 10.1007/s00018-013-1480-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ransohoff RM, Brown MA. Innate immunity in the central nervous system. J Clin Invest. 2012;122:1164–1171. doi: 10.1172/JCI58644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Farina C, Krumbholz M, Giese T, Hartmann G, Aloisi F, Meinl E. Preferential expression and function of Toll-like receptor 3 in human astrocytes. J Neuroimmunol. 2005;159:12–19. doi: 10.1016/j.jneuroim.2004.09.009. [DOI] [PubMed] [Google Scholar]
  • 60.Bsibsi M, Persoon-Deen C, Verwer RW, Meeuwsen S, Ravid R, Van Noort JM. Toll-like receptor 3 on adult human astrocytes triggers production of neuroprotective mediators. Glia. 2006;53:688–695. doi: 10.1002/glia.20328. [DOI] [PubMed] [Google Scholar]
  • 61.Yoshimura A, Lien E, Ingalls RR, Tuomanen E, Dziarski R, Golenbock D. Cutting edge: recognition of Gram-positive bacterial cell wall components by the innate immune system occurs via Toll-like receptor 2. J Immunol. 1999;163:1–5. [PubMed] [Google Scholar]
  • 62.Laflamme N, Soucy G, Rivest S. Circulating cell wall components derived from gram-negative, not gram-positive, bacteria cause a profound induction of the gene-encoding Toll-like receptor 2 in the CNS. J Neurochem. 2001;79:648–657. doi: 10.1046/j.1471-4159.2001.00603.x. [DOI] [PubMed] [Google Scholar]
  • 63.Hanke ML, Kielian T. Toll-like receptors in health and disease in the brain: mechanisms and therapeutic potential. Clin Sci (Lond) 2011;121:367–387. doi: 10.1042/CS20110164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Costello DA, Lynch MA. Toll-like receptor 3 activation modulates hippocampal network excitability, via glial production of interferon-beta. Hippocampus. 2013;23:696–707. doi: 10.1002/hipo.22129. [DOI] [PubMed] [Google Scholar]
  • 65.Gesuete R, Packard AE, Vartanian KB, Conrad VK, Stevens SL, Bahjat FR, et al. Poly-ICLC preconditioning protects the blood-brain barrier against ischemic injury in vitro through type I interferon signaling. J Neurochem. 2012;123(Suppl 2):75–85. doi: 10.1111/j.1471-4159.2012.07946.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol. 2007;28:138–145. doi: 10.1016/j.it.2007.01.005. [DOI] [PubMed] [Google Scholar]
  • 67.Sriram K, Benkovic SA, Hebert MA, Miller DB, O’Callaghan JP. Induction of gp130-related cytokines and activation of JAK2/STAT3 pathway in astrocytes precedes up-regulation of glial fibrillary acidic protein in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of neurodegeneration: key signaling pathway for astrogliosis in vivo? J Biol Chem. 2004;279:19936–19947. doi: 10.1074/jbc.M309304200. [DOI] [PubMed] [Google Scholar]
  • 68.Wiese S, Karus M, Faissner A. Astrocytes as a source for extracellular matrix molecules and cytokines. Front Pharmacol. 2012;3:120. doi: 10.3389/fphar.2012.00120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Boland B, Platt FM. Bridging the age spectrum of neurodegenerative storage diseases. Best Pract Res Clin Endocrinol Metab. 2015;29:127–143. doi: 10.1016/j.beem.2014.08.009. [DOI] [PubMed] [Google Scholar]
  • 70.Thompson PM, Vinters HV. Pathologic lesions in neurodegenerative diseases. Prog Mol Biol Transl Sci. 2012;107:1–40. doi: 10.1016/B978-0-12-385883-2.00009-6. [DOI] [PubMed] [Google Scholar]
  • 71.Terry RD, Davies P. Dementia of the Alzheimer type. Annu Rev Neurosci. 1980;3:77–95. doi: 10.1146/annurev.ne.03.030180.000453. [DOI] [PubMed] [Google Scholar]
  • 72.Cummings JL, Vinters HV, Cole GM, Khachaturian ZS. Alzheimer’s disease: etiologies, pathophysiology, cognitive reserve, and treatment opportunities. Neurology. 1998;51:S2–17. doi: 10.1212/wnl.51.1_suppl_1.s2. ; discussion S65–17. [DOI] [PubMed] [Google Scholar]
  • 73.Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C, et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature. 2012;482:216–220. doi: 10.1038/nature10821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Vinters HV. Emerging concepts in Alzheimer’s disease. Annu Rev Pathol. 2015;10:291–319. doi: 10.1146/annurev-pathol-020712-163927. [DOI] [PubMed] [Google Scholar]
  • 75.Iqbal K, Wisniewski HM, Grundke-Iqbal I, Korthals JK, Terry RD. Chemical pathology of neurofibrils. Neurofibrillary tangles of Alzheimer’s presenile-senile dementia. J Histochem Cytochem. 1975;23:563–569. doi: 10.1177/23.7.1141687. [DOI] [PubMed] [Google Scholar]
  • 76.Terry RD. The Fine Structure of Neurofibrillary Tangles in Alzheimer’s Disease. J Neuropathol Exp Neurol. 1963;22:629–642. doi: 10.1097/00005072-196310000-00005. [DOI] [PubMed] [Google Scholar]
  • 77.Lewis DA, Campbell MJ, Terry RD, Morrison JH. Laminar and regional distributions of neurofibrillary tangles and neuritic plaques in Alzheimer’s disease: a quantitative study of visual and auditory cortices. J Neurosci. 1987;7:1799–1808. doi: 10.1523/JNEUROSCI.07-06-01799.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Vinters HV, Secor DL, Read SL, Frazee JG, Tomiyasu U, Stanley TM, et al. Microvasculature in brain biopsy specimens from patients with Alzheimer’s disease: an immunohistochemical and ultrastructural study. Ultrastruct Pathol. 1994;18:333–348. doi: 10.3109/01913129409023202. [DOI] [PubMed] [Google Scholar]
  • 79.Terry RD, Gonatas NK, Weiss M. Ultrastructural Studies in Alzheimer’s Presenile Dementia. Am J Pathol. 1964;44:269–297. [PMC free article] [PubMed] [Google Scholar]
  • 80.Terry RD. Morphological changes in Alzheimer’s disease-senile dementia: ultrastructural changes and quantitative studies. Res Publ Assoc Res Nerv Ment Dis. 1979;57:99–105. [PubMed] [Google Scholar]
  • 81.Praprotnik D, Smith MA, Richey PL, Vinters HV, Perry G. Filament heterogeneity within the dystrophic neurites of senile plaques suggests blockage of fast axonal transport in Alzheimer’s disease. Acta Neuropathol. 1996;91:226–235. doi: 10.1007/s004010050420. [DOI] [PubMed] [Google Scholar]
  • 82.Mann DM. The neuropathology of Alzheimer’s disease: a review with pathogenetic, aetiological and therapeutic considerations. Mech Ageing Dev. 1985;31:213–255. doi: 10.1016/0047-6374(85)90092-2. [DOI] [PubMed] [Google Scholar]
  • 83.Mann DM, Yates PO, Marcyniuk B. Correlation between senile plaque and neurofibrillary tangle counts in cerebral cortex and neuronal counts in cortex and subcortical structures in Alzheimer’s disease. Neurosci Lett. 1985;56:51–55. doi: 10.1016/0304-3940(85)90439-2. [DOI] [PubMed] [Google Scholar]
  • 84.Pike CJ, Cummings BJ, Cotman CW. Early association of reactive astrocytes with senile plaques in Alzheimer’s disease. Exp Neurol. 1995;132:172–179. doi: 10.1016/0014-4886(95)90022-5. [DOI] [PubMed] [Google Scholar]
  • 85.Abraham CR. Reactive astrocytes and alpha1-antichymotrypsin in Alzheimer’s disease. Neurobiol Aging. 2001;22:931–936. doi: 10.1016/s0197-4580(01)00302-5. [DOI] [PubMed] [Google Scholar]
  • 86.Fuller S, Munch G, Steele M. Activated astrocytes: a therapeutic target in Alzheimer’s disease? Expert Rev Neurother. 2009;9:1585–1594. doi: 10.1586/ern.09.111. [DOI] [PubMed] [Google Scholar]
  • 87.Angelova PR, Abramov AY. Interaction of neurons and astrocytes underlies the mechanism of Abeta-induced neurotoxicity. Biochem Soc Trans. 2014;42:1286–1290. doi: 10.1042/BST20140153. [DOI] [PubMed] [Google Scholar]
  • 88.Abeti R, Abramov AY. Mitochondrial Ca in neurodegenerative disorders. Pharmacol Res. 2015 doi: 10.1016/j.phrs.2015.05.007. [DOI] [PubMed] [Google Scholar]
  • 89.Allaman I, Gavillet M, Belanger M, Laroche T, Viertl D, Lashuel HA, et al. Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability. J Neurosci. 2010;30:3326–3338. doi: 10.1523/JNEUROSCI.5098-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Talantova M, Sanz-Blasco S, Zhang X, Xia P, Akhtar MW, Okamoto S, et al. Abeta induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proc Natl Acad Sci U S A. 2013;110:E2518–2527. doi: 10.1073/pnas.1306832110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Abeti R, Abramov AY, Duchen MR. Beta-amyloid activates PARP causing astrocytic metabolic failure and neuronal death. Brain. 2011;134:1658–1672. doi: 10.1093/brain/awr104. [DOI] [PubMed] [Google Scholar]
  • 92.Wyss-Coray T, Loike JD, Brionne TC, Lu E, Anankov R, Yan F, et al. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med. 2003;9:453–457. doi: 10.1038/nm838. [DOI] [PubMed] [Google Scholar]
  • 93.Kimura N, Okabayashi S, Ono F. Dynein dysfunction disrupts beta-amyloid clearance in astrocytes through endocytic disturbances. Neuroreport. 2014;25:514–520. doi: 10.1097/WNR.0000000000000124. [DOI] [PubMed] [Google Scholar]
  • 94.Rogers J, Strohmeyer R, Kovelowski CJ, Li R. Microglia and inflammatory mechanisms in the clearance of amyloid beta peptide. Glia. 2002;40:260–269. doi: 10.1002/glia.10153. [DOI] [PubMed] [Google Scholar]
  • 95.Lai AY, McLaurin J. Clearance of amyloid-beta peptides by microglia and macrophages: the issue of what, when and where. Future Neurol. 2012;7:165–176. doi: 10.2217/fnl.12.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Przedborski S, Vila M. The last decade in Parkinson’s disease research. Basic sciences. Adv Neurol. 2001;86:177–186. [PubMed] [Google Scholar]
  • 97.Dauer W, Przedborski S. Parkinson’s disease: mechanisms and models. Neuron. 2003;39:889–909. doi: 10.1016/s0896-6273(03)00568-3. [DOI] [PubMed] [Google Scholar]
  • 98.Mitchell SL, Kiely DK, Kiel DP, Lipsitz LA. The epidemiology, clinical characteristics, and natural history of older nursing home residents with a diagnosis of Parkinson’s disease. J Am Geriatr Soc. 1996;44:394–399. doi: 10.1111/j.1532-5415.1996.tb06408.x. [DOI] [PubMed] [Google Scholar]
  • 99.Lapane KL, Fernandez HH, Friedman JH. Prevalence, clinical characteristics, and pharmacologic treatment of Parkinson’s disease in residents in long-term care facilities. SAGE Study Group. Pharmacotherapy. 1999;19:1321–1327. doi: 10.1592/phco.19.16.1321.30877. [DOI] [PubMed] [Google Scholar]
  • 100.Locascio JJ, Corkin S, Growdon JH. Relation between clinical characteristics of Parkinson’s disease and cognitive decline. J Clin Exp Neuropsychol. 2003;25:94–109. doi: 10.1076/jcen.25.1.94.13624. [DOI] [PubMed] [Google Scholar]
  • 101.Hirsch EC, Jenner P, Przedborski S. Pathogenesis of Parkinson’s disease. Mov Disord. 2013;28:24–30. doi: 10.1002/mds.25032. [DOI] [PubMed] [Google Scholar]
  • 102.Yoshimura N, Hayashi S, Fukushima Y. Diffuse Mallory bodies in the liver, diffuse Lewy bodies in the brain and diffuse fat replacement (lipomatous pseudohypertrophy) of the pancreas in a patient with juvenile Parkinson’s disease. Acta Pathol Jpn. 1992;42:826–831. doi: 10.1111/j.1440-1827.1992.tb01884.x. [DOI] [PubMed] [Google Scholar]
  • 103.Owen AD, Schapira AH, Jenner P, Marsden CD. Indices of oxidative stress in Parkinson’s disease, Alzheimer’s disease and dementia with Lewy bodies. J Neural Transm Suppl. 1997;51:167–173. doi: 10.1007/978-3-7091-6846-2_14. [DOI] [PubMed] [Google Scholar]
  • 104.Spillantini MG, Goedert M. The alpha-synucleinopathies: Parkinson’s disease, dementia with Lewy bodies, and multiple system atrophy. Ann N Y Acad Sci. 2000;920:16–27. doi: 10.1111/j.1749-6632.2000.tb06900.x. [DOI] [PubMed] [Google Scholar]
  • 105.Bennett JP., Jr Striatal dopamine depletion, dopamine receptor stimulation, and GABA metabolism: implications for the therapy of Parkinson’s disease. Ann Neurol. 1986;19:194–197. doi: 10.1002/ana.410190215. [DOI] [PubMed] [Google Scholar]
  • 106.Dubois B, Pillon B. Do cognitive changes of Parkinson’s disease result from dopamine depletion? J Neural Transm Suppl. 1995;45:27–34. [PubMed] [Google Scholar]
  • 107.Prell GD, Khandelwal JK, Burns RS, Blandina P, Morrishow AM, Green JP. Levels of pros-methylimidazoleacetic acid: correlation with severity of Parkinson’s disease in CSF of patients and with the depletion of striatal dopamine and its metabolites in MPTP-treated mice. J Neural Transm Park Dis Dement Sect. 1991;3:109–125. doi: 10.1007/BF02260886. [DOI] [PubMed] [Google Scholar]
  • 108.Kish SJ, Shannak KS, Rajput AH, Gilbert JJ, Hornykiewicz O. Cerebellar norepinephrine in patients with Parkinson’s disease and control subjects. Arch Neurol. 1984;41:612–614. doi: 10.1001/archneur.1984.04210080020007. [DOI] [PubMed] [Google Scholar]
  • 109.Hornykiewicz O, Kish SJ. Biochemical pathophysiology of Parkinson’s disease. Adv Neurol. 1987;45:19–34. [PubMed] [Google Scholar]
  • 110.Nakagawa T, Yabe T, Schwartz JP. Gene expression profiles of reactive astrocytes cultured from dopamine-depleted striatum. Neurobiol Dis. 2005;20:275–282. doi: 10.1016/j.nbd.2005.03.009. [DOI] [PubMed] [Google Scholar]
  • 111.L’Episcopo F, Tirolo C, Caniglia S, Testa N, Morale MC, Serapide MF, et al. Targeting Wnt signaling at the neuroimmune interface for dopaminergic neuroprotection/repair in Parkinson’s disease. J Mol Cell Biol. 2014;6:13–26. doi: 10.1093/jmcb/mjt053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Sun YY, Zhang Y, Sun XP, Liu TY, Liu ZH, Chen G, et al. Fasciculation and elongation protein zeta-1 (FEZ1) expression in reactive astrocytes in a rat model of Parkinson’s disease. Neuropathol Appl Neurobiol. 2014;40:164–176. doi: 10.1111/nan.12077. [DOI] [PubMed] [Google Scholar]
  • 113.Yoshimoto Y, Lin Q, Collier TJ, Frim DM, Breakefield XO, Bohn MC. Astrocytes retrovirally transduced with BDNF elicit behavioral improvement in a rat model of Parkinson’s disease. Brain Res. 1995;691:25–36. doi: 10.1016/0006-8993(95)00596-i. [DOI] [PubMed] [Google Scholar]
  • 114.Kato H, Araki T, Imai Y, Takahashi A, Itoyama Y. Protection of dopaminergic neurons with a novel astrocyte modulating agent (R)-(−)-2-propyloctanoic acid (ONO-2506) in an MPTP-mouse model of Parkinson’s disease. J Neurol Sci. 2003;208:9–15. doi: 10.1016/s0022-510x(02)00411-2. [DOI] [PubMed] [Google Scholar]
  • 115.Kang KH, Liou HH, Hour MJ, Liou HC, Fu WM. Protection of dopaminergic neurons by 5-lipoxygenase inhibitor. Neuropharmacology. 2013;73:380–387. doi: 10.1016/j.neuropharm.2013.06.014. [DOI] [PubMed] [Google Scholar]
  • 116.Carbone DL, Popichak KA, Moreno JA, Safe S, Tjalkens RB. Suppression of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced nitric-oxide synthase 2 expression in astrocytes by a novel diindolylmethane analog protects striatal neurons against apoptosis. Mol Pharmacol. 2009;75:35–43. doi: 10.1124/mol.108.050781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Boillee S, Vande Velde C, Cleveland DW. ALS: a disease of motor neurons and their nonneuronal neighbors. Neuron. 2006;52:39–59. doi: 10.1016/j.neuron.2006.09.018. [DOI] [PubMed] [Google Scholar]
  • 118.Nagai M, Re DB, Nagata T, Chalazonitis A, Jessell TM, Wichterle H, et al. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat Neurosci. 2007;10:615–622. doi: 10.1038/nn1876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Di Giorgio FP, Carrasco MA, Siao MC, Maniatis T, Eggan K. Non-cell autonomous effect of glia on motor neurons in an embryonic stem cell-based ALS model. Nat Neurosci. 2007;10:608–614. doi: 10.1038/nn1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bilsland LG, Nirmalananthan N, Yip J, Greensmith L, Duchen MR. Expression of mutant SOD1 in astrocytes induces functional deficits in motoneuron mitochondria. J Neurochem. 2008;107:1271–1283. doi: 10.1111/j.1471-4159.2008.05699.x. [DOI] [PubMed] [Google Scholar]
  • 121.Getty AL, Rothberg PG, Pearce DA. Diagnosis of neuronal ceroid lipofuscinosis: mutation detection strategies. Expert Opin Med Diagn. 2007;1:351–362. doi: 10.1517/17530059.1.3.351. [DOI] [PubMed] [Google Scholar]
  • 122.Aberg L, Lauronen L, Hamalainen J, Mole SE, Autti T. A 30-year follow-up of a neuronal ceroid lipofuscinosis patient with mutations in CLN3 and protracted disease course. Pediatr Neurol. 2009;40:134–137. doi: 10.1016/j.pediatrneurol.2008.10.012. [DOI] [PubMed] [Google Scholar]
  • 123.Lebrun AH, Moll-Khosrawi P, Pohl S, Makrypidi G, Storch S, Kilian D, et al. Analysis of potential biomarkers and modifier genes affecting the clinical course of CLN3 disease. Mol Med. 2011;17:1253–1261. doi: 10.2119/molmed.2010.00241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sinha S, Satishchandra P, Santosh V, Gayatri N, Shankar SK. Neuronal ceroid lipofuscinosis: a clinicopathological study. Seizure. 2004;13:235–240. doi: 10.1016/S1059-1311(03)00163-8. [DOI] [PubMed] [Google Scholar]
  • 125.Cotman SL, Karaa A, Staropoli JF, Sims KB. Neuronal ceroid lipofuscinosis: impact of recent genetic advances and expansion of the clinicopathologic spectrum. Curr Neurol Neurosci Rep. 2013;13:366. doi: 10.1007/s11910-013-0366-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Palmer DN, Barns G, Husbands DR, Jolly RD. Ceroid lipofuscinosis in sheep. II. The major component of the lipopigment in liver, kidney, pancreas, and brain is low molecular weight protein. J Biol Chem. 1986;261:1773–1777. [PubMed] [Google Scholar]
  • 127.Pardo CA, Rabin BA, Palmer DN, Price DL. Accumulation of the adenosine triphosphate synthase subunit C in the mnd mutant mouse. A model for neuronal ceroid lipofuscinosis. Am J Pathol. 1994;144:829–835. [PMC free article] [PubMed] [Google Scholar]
  • 128.Goebel HH, Schochet SS, Jaynes M, Bruck W, Kohlschutter A, Hentati F. Progress in neuropathology of the neuronal ceroid lipofuscinoses. Mol Genet Metab. 1999;66:367–372. doi: 10.1006/mgme.1999.2808. [DOI] [PubMed] [Google Scholar]
  • 129.Fossale E, Wolf P, Espinola JA, Lubicz-Nawrocka T, Teed AM, Gao H, et al. Membrane trafficking and mitochondrial abnormalities precede subunit c deposition in a cerebellar cell model of juvenile neuronal ceroid lipofuscinosis. BMC Neurosci. 2004;5:57. doi: 10.1186/1471-2202-5-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Hawkins-Salsbury JA, Cooper JD, Sands MS. Pathogenesis and therapies for infantile neuronal ceroid lipofuscinosis (infantile CLN1 disease) Biochim Biophys Acta. 2013;1832:1906–1909. doi: 10.1016/j.bbadis.2013.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Vesa J, Hellsten E, Verkruyse LA, Camp LA, Rapola J, Santavuori P, et al. Mutations in the palmitoyl protein thioesterase gene causing infantile neuronal ceroid lipofuscinosis. Nature. 1995;376:584–587. doi: 10.1038/376584a0. [DOI] [PubMed] [Google Scholar]
  • 132.Hofmann SL, Lee LA, Lu JY, Verkruyse LA. Palmitoyl-protein thioesterase and the molecular pathogenesis of infantile neuronal ceroid lipofuscinosis. Neuropediatrics. 1997;28:27–30. doi: 10.1055/s-2007-973661. [DOI] [PubMed] [Google Scholar]
  • 133.Sleat DE, Donnelly RJ, Lackland H, Liu CG, Sohar I, Pullarkat RK, et al. Association of mutations in a lysosomal protein with classical late-infantile neuronal ceroid lipofuscinosis. Science. 1997;277:1802–1805. doi: 10.1126/science.277.5333.1802. [DOI] [PubMed] [Google Scholar]
  • 134.Sleat DE, Gin RM, Sohar I, Wisniewski K, Sklower-Brooks S, Pullarkat RK, et al. Mutational analysis of the defective protease in classic late-infantile neuronal ceroid lipofuscinosis, a neurodegenerative lysosomal storage disorder. Am J Hum Genet. 1999;64:1511–1523. doi: 10.1086/302427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Pearce DA, Sherman F. Investigation of Batten disease with the yeast Saccharomyces cerevisiae. Mol Genet Metab. 1999;66:314–319. doi: 10.1006/mgme.1999.2820. [DOI] [PubMed] [Google Scholar]
  • 136.Kim Y, Ramirez-Montealegre D, Pearce DA. A role in vacuolar arginine transport for yeast Btn1p and for human CLN3, the protein defective in Batten disease. Proc Natl Acad Sci U S A. 2003;100:15458–15462. doi: 10.1073/pnas.2136651100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Luiro K, Yliannala K, Ahtiainen L, Maunu H, Jarvela I, Kyttala A, et al. Interconnections of CLN3, Hook1 and Rab proteins link Batten disease to defects in the endocytic pathway. Hum Mol Genet. 2004;13:3017–3027. doi: 10.1093/hmg/ddh321. [DOI] [PubMed] [Google Scholar]
  • 138.Anderson GW, Goebel HH, Simonati A. Human pathology in NCL. Biochim Biophys Acta. 2013;1832:1807–1826. doi: 10.1016/j.bbadis.2012.11.014. [DOI] [PubMed] [Google Scholar]
  • 139.Radke J, Stenzel W, Goebel HH. Human NCL Neuropathology. Biochim Biophys Acta. 2015 doi: 10.1016/j.bbadis.2015.05.007. [DOI] [PubMed] [Google Scholar]
  • 140.Cooper JD, Tarczyluk MA, Nelvagal HR. Towards a new understanding of NCL pathogenesis. Biochim Biophys Acta. 2015 doi: 10.1016/j.bbadis.2015.05.014. [DOI] [PubMed] [Google Scholar]
  • 141.Rama Rao KV, Kielian T. Astrocytes and lysosomal storage diseases. Neuroscience. 2015 doi: 10.1016/j.neuroscience.2015.05.061. in Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Pontikis CC, Cella CV, Parihar N, Lim MJ, Chakrabarti S, Mitchison HM, et al. Late onset neurodegeneration in the Cln3−/− mouse model of juvenile neuronal ceroid lipofuscinosis is preceded by low level glial activation. Brain Res. 2004;1023:231–242. doi: 10.1016/j.brainres.2004.07.030. [DOI] [PubMed] [Google Scholar]
  • 143.Burkovetskaya M, Karpuk N, Xiong J, Bosch M, Boska MD, Takeuchi H, et al. Evidence for aberrant astrocyte hemichannel activity in Juvenile Neuronal Ceroid Lipofuscinosis (JNCL) PLoS One. 2014;9:e95023. doi: 10.1371/journal.pone.0095023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Macauley SL, Pekny M, Sands MS. The role of attenuated astrocyte activation in infantile neuronal ceroid lipofuscinosis. J Neurosci. 2011;31:15575–15585. doi: 10.1523/JNEUROSCI.3579-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bronson RT, Donahue LR, Johnson KR, Tanner A, Lane PW, Faust JR. Neuronal ceroid lipofuscinosis (nclf), a new disorder of the mouse linked to chromosome 9. Am J Med Genet. 1998;77:289–297. doi: 10.1002/(sici)1096-8628(19980526)77:4<289::aid-ajmg8>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
  • 146.Wheeler RB, Sharp JD, Schultz RA, Joslin JM, Williams RE, Mole SE. The gene mutated in variant late-infantile neuronal ceroid lipofuscinosis (CLN6) and in nclf mutant mice encodes a novel predicted transmembrane protein. Am J Hum Genet. 2002;70:537–542. doi: 10.1086/338708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Oswald MJ, Palmer DN, Kay GW, Shemilt SJ, Rezaie P, Cooper JD. Glial activation spreads from specific cerebral foci and precedes neurodegeneration in presymptomatic ovine neuronal ceroid lipofuscinosis (CLN6) Neurobiol Dis. 2005;20:49–63. doi: 10.1016/j.nbd.2005.01.025. [DOI] [PubMed] [Google Scholar]
  • 148.Kay GW, Palmer DN, Rezaie P, Cooper JD. Activation of non-neuronal cells within the prenatal developing brain of sheep with neuronal ceroid lipofuscinosis. Brain Pathol. 2006;16:110–116. doi: 10.1111/j.1750-3639.2006.00002.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Morgan JP, Magee H, Wong A, Nelson T, Koch B, Cooper JD, et al. A murine model of variant late infantile ceroid lipofuscinosis recapitulates behavioral and pathological phenotypes of human disease. PLoS One. 2013;8:e78694. doi: 10.1371/journal.pone.0078694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Verkhratsky A, Kettenmann H. Calcium signalling in glial cells. Trends Neurosci. 1996;19:346–352. doi: 10.1016/0166-2236(96)10048-5. [DOI] [PubMed] [Google Scholar]
  • 151.Angulo MC, Kozlov AS, Charpak S, Audinat E. Glutamate released from glial cells synchronizes neuronal activity in the hippocampus. J Neurosci. 2004;24:6920–6927. doi: 10.1523/JNEUROSCI.0473-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Araque A, Carmignoto G, Haydon PG, Oliet SH, Robitaille R, Volterra A. Gliotransmitters travel in time and space. Neuron. 2014;81:728–739. doi: 10.1016/j.neuron.2014.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Fellin T, Sul JY, D’Ascenzo M, Takano H, Pascual O, Haydon PG. Bidirectional astrocyte-neuron communication: the many roles of glutamate and ATP. Novartis Found Symp. 2006;276:208–217. doi: 10.1002/9780470032244.ch16. discussion 217–221, 233–207, 275–281. [DOI] [PubMed] [Google Scholar]
  • 154.Matos M, Augusto E, Oliveira CR, Agostinho P. Amyloid-beta peptide decreases glutamate uptake in cultured astrocytes: involvement of oxidative stress and mitogen-activated protein kinase cascades. Neuroscience. 2008;156:898–910. doi: 10.1016/j.neuroscience.2008.08.022. [DOI] [PubMed] [Google Scholar]
  • 155.Tian G, Kong Q, Lai L, Ray-Chaudhury A, Lin CL. Increased expression of cholesterol 24S-hydroxylase results in disruption of glial glutamate transporter EAAT2 association with lipid rafts: a potential role in Alzheimer’s disease. J Neurochem. 2010;113:978–989. doi: 10.1111/j.1471-4159.2010.06661.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Di Monte DA, Tokar I, Langston JW. Impaired glutamate clearance as a consequence of energy failure caused by MPP(+) in astrocytic cultures. Toxicol Appl Pharmacol. 1999;158:296–302. doi: 10.1006/taap.1999.8717. [DOI] [PubMed] [Google Scholar]
  • 157.Hazell AS, Itzhak Y, Liu H, Norenberg MD. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) decreases glutamate uptake in cultured astrocytes. J Neurochem. 1997;68:2216–2219. doi: 10.1046/j.1471-4159.1997.68052216.x. [DOI] [PubMed] [Google Scholar]
  • 158.Hsu CY, Hung CS, Chang HM, Liao WC, Ho SC, Ho YJ. Ceftriaxone prevents and reverses behavioral and neuronal deficits in an MPTP-induced animal model of Parkinson’s disease dementia. Neuropharmacology. 2015;91:43–56. doi: 10.1016/j.neuropharm.2014.11.023. [DOI] [PubMed] [Google Scholar]
  • 159.Pardo AC, Wong V, Benson LM, Dykes M, Tanaka K, Rothstein JD, et al. Loss of the astrocyte glutamate transporter GLT1 modifies disease in SOD1(G93A) mice. Exp Neurol. 2006;201:120–130. doi: 10.1016/j.expneurol.2006.03.028. [DOI] [PubMed] [Google Scholar]
  • 160.Tortarolo M, Crossthwaite AJ, Conforti L, Spencer JP, Williams RJ, Bendotti C, et al. Expression of SOD1 G93A or wild-type SOD1 in primary cultures of astrocytes down-regulates the glutamate transporter GLT-1: lack of involvement of oxidative stress. J Neurochem. 2004;88:481–493. doi: 10.1046/j.1471-4159.2003.02208.x. [DOI] [PubMed] [Google Scholar]
  • 161.Gu R, Hou X, Pang R, Li L, Chen F, Geng J, et al. Human adipose-derived stem cells enhance the glutamate uptake function of GLT1 in SOD1(G93A)-bearing astrocytes. Biochem Biophys Res Commun. 2010;393:481–486. doi: 10.1016/j.bbrc.2010.02.029. [DOI] [PubMed] [Google Scholar]
  • 162.Bosch M, Kielian T. Hemichannels in neurodegenerative diseases: is there a link to pathology? Front Cell Neurosci. 2014;8:242. doi: 10.3389/fncel.2014.00242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Montero TD, Orellana JA. Hemichannels: new pathways for gliotransmitter release. Neuroscience. 2015;286:45–59. doi: 10.1016/j.neuroscience.2014.11.048. [DOI] [PubMed] [Google Scholar]
  • 164.Saez JC, Leybaert L. Hunting for connexin hemichannels. FEBS Lett. 2014;588:1205–1211. doi: 10.1016/j.febslet.2014.03.004. [DOI] [PubMed] [Google Scholar]
  • 165.Saez PJ, Orellana JA, Vega-Riveros N, Figueroa VA, Hernandez DE, Castro JF, et al. Disruption in connexin-based communication is associated with intracellular Ca(2)(+) signal alterations in astrocytes from Niemann-Pick type C mice. PLoS One. 2013;8:e71361. doi: 10.1371/journal.pone.0071361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Delekate A, Fuchtemeier M, Schumacher T, Ulbrich C, Foddis M, Petzold GC. Metabotropic P2Y1 receptor signalling mediates astrocytic hyperactivity in vivo in an Alzheimer’s disease mouse model. Nat Commun. 2014;5:5422. doi: 10.1038/ncomms6422. [DOI] [PubMed] [Google Scholar]
  • 167.Orellana JA, Shoji KF, Abudara V, Ezan P, Amigou E, Saez PJ, et al. Amyloid beta-induced death in neurons involves glial and neuronal hemichannels. J Neurosci. 2011;31:4962–4977. doi: 10.1523/JNEUROSCI.6417-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kielian T. Glial connexins and gap junctions in CNS inflammation and disease. J Neurochem. 2008;106:1000–1016. doi: 10.1111/j.1471-4159.2008.05405.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Orellana JA, Froger N, Ezan P, Jiang JX, Bennett MV, Naus CC, et al. ATP and glutamate released via astroglial connexin 43 hemichannels mediate neuronal death through activation of pannexin 1 hemichannels. J Neurochem. 2011;118:826–840. doi: 10.1111/j.1471-4159.2011.07210.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, et al. Tumor necrosis factor-alpha induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner. J Biol Chem. 2006;281:21362–21368. doi: 10.1074/jbc.M600504200. [DOI] [PubMed] [Google Scholar]
  • 171.Saez PJ, Shoji KF, Aguirre A, Saez JC. Regulation of hemichannels and gap junction channels by cytokines in antigen-presenting cells. Mediators Inflamm. 2014;2014:742734. doi: 10.1155/2014/742734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Abudara V, Roux L, Dallerac G, Matias I, Dulong J, Mothet JP, et al. Activated microglia impairs neuroglial interaction by opening Cx43 hemichannels in hippocampal astrocytes. Glia. 2015;63:795–811. doi: 10.1002/glia.22785. [DOI] [PubMed] [Google Scholar]
  • 173.Franke H, Illes P. Involvement of P2 receptors in the growth and survival of neurons in the CNS. Pharmacol Ther. 2006;109:297–324. doi: 10.1016/j.pharmthera.2005.06.002. [DOI] [PubMed] [Google Scholar]
  • 174.Burnstock G, Krugel U, Abbracchio MP, Illes P. Purinergic signalling: from normal behaviour to pathological brain function. Prog Neurobiol. 2011;95:229–274. doi: 10.1016/j.pneurobio.2011.08.006. [DOI] [PubMed] [Google Scholar]
  • 175.Illes P, Verkhratsky A, Burnstock G, Franke H. P2X receptors and their roles in astroglia in the central and peripheral nervous system. Neuroscientist. 2012;18:422–438. doi: 10.1177/1073858411418524. [DOI] [PubMed] [Google Scholar]
  • 176.Brambilla R, Ceruti S, Malorni W, Cattabeni F, Abbracchio MP. A novel gliotic P2 receptor mediating cyclooxygenase-2 induction in rat and human astrocytes. J Auton Nerv Syst. 2000;81:3–9. doi: 10.1016/s0165-1838(00)00152-1. [DOI] [PubMed] [Google Scholar]
  • 177.Florenzano F, Viscomi MT, Amadio S, D’Ambrosi N, Volonte C, Molinari M. Do ATP and NO interact in the CNS? Prog Neurobiol. 2008;84:40–56. doi: 10.1016/j.pneurobio.2007.10.004. [DOI] [PubMed] [Google Scholar]
  • 178.Farber K, Kettenmann H. Purinergic signaling and microglia. Pflugers Arch. 2006;452:615–621. doi: 10.1007/s00424-006-0064-7. [DOI] [PubMed] [Google Scholar]
  • 179.Jenner P, Mori A, Hauser R, Morelli M, Fredholm BB, Chen JF. Adenosine, adenosine A 2A antagonists, and Parkinson’s disease. Parkinsonism Relat Disord. 2009;15:406–413. doi: 10.1016/j.parkreldis.2008.12.006. [DOI] [PubMed] [Google Scholar]
  • 180.Gyoneva S, Shapiro L, Lazo C, Garnier-Amblard E, Smith Y, Miller GW, et al. Adenosine A2A receptor antagonism reverses inflammation-induced impairment of microglial process extension in a model of Parkinson’s disease. Neurobiol Dis. 2014;67:191–202. doi: 10.1016/j.nbd.2014.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Erb L, Cao C, Ajit D, Weisman GA. P2Y receptors in Alzheimer’s disease. Biol Cell. 2015;107:1–21. doi: 10.1111/boc.201400043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Amadio S, Apolloni S, D’Ambrosi N, Volonte C. Purinergic signalling at the plasma membrane: a multipurpose and multidirectional mode to deal with amyotrophic lateral sclerosis and multiple sclerosis. J Neurochem. 2011;116:796–805. doi: 10.1111/j.1471-4159.2010.07025.x. [DOI] [PubMed] [Google Scholar]
  • 183.Sperlagh B, Illes P. P2X7 receptor: an emerging target in central nervous system diseases. Trends Pharmacol Sci. 2014;35:537–547. doi: 10.1016/j.tips.2014.08.002. [DOI] [PubMed] [Google Scholar]
  • 184.Canas PM, Porciuncula LO, Cunha GM, Silva CG, Machado NJ, Oliveira JM, et al. Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway. J Neurosci. 2009;29:14741–14751. doi: 10.1523/JNEUROSCI.3728-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Sanz JM, Chiozzi P, Ferrari D, Colaianna M, Idzko M, Falzoni S, et al. Activation of microglia by amyloid {beta} requires P2X7 receptor expression. J Immunol. 2009;182:4378–4385. doi: 10.4049/jimmunol.0803612. [DOI] [PubMed] [Google Scholar]
  • 186.Yu L, Shen HY, Coelho JE, Araujo IM, Huang QY, Day YJ, et al. Adenosine A2A receptor antagonists exert motor and neuroprotective effects by distinct cellular mechanisms. Ann Neurol. 2008;63:338–346. doi: 10.1002/ana.21313. [DOI] [PubMed] [Google Scholar]
  • 187.Apolloni S, Amadio S, Montilli C, Volonte C, D’Ambrosi N. Ablation of P2X7 receptor exacerbates gliosis and motoneuron death in the SOD1-G93A mouse model of amyotrophic lateral sclerosis. Hum Mol Genet. 2013;22:4102–4116. doi: 10.1093/hmg/ddt259. [DOI] [PubMed] [Google Scholar]
  • 188.Schell MJ, Molliver ME, Snyder SH. D-serine, an endogenous synaptic modulator: localization to astrocytes and glutamate-stimulated release. Proc Natl Acad Sci U S A. 1995;92:3948–3952. doi: 10.1073/pnas.92.9.3948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Wolosker H, Panizzutti R, De Miranda J. Neurobiology through the looking-glass: D-serine as a new glial-derived transmitter. Neurochem Int. 2002;41:327–332. doi: 10.1016/s0197-0186(02)00055-4. [DOI] [PubMed] [Google Scholar]
  • 190.De Miranda J, Santoro A, Engelender S, Wolosker H. Human serine racemase: moleular cloning, genomic organization and functional analysis. Gene. 2000;256:183–188. doi: 10.1016/s0378-1119(00)00356-5. [DOI] [PubMed] [Google Scholar]
  • 191.Yang JH, Wada A, Yoshida K, Miyoshi Y, Sayano T, Esaki K, et al. Brain-specific Phgdh deletion reveals a pivotal role for L-serine biosynthesis in controlling the level of D-serine, an N-methyl-D-aspartate receptor co-agonist, in adult brain. J Biol Chem. 2010;285:41380–41390. doi: 10.1074/jbc.M110.187443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Wolosker H. Serine racemase and the serine shuttle between neurons and astrocytes. Biochim Biophys Acta. 2011;1814:1558–1566. doi: 10.1016/j.bbapap.2011.01.001. [DOI] [PubMed] [Google Scholar]
  • 193.Yang Y, Ge W, Chen Y, Zhang Z, Shen W, Wu C, et al. Contribution of astrocytes to hippocampal long-term potentiation through release of D-serine. Proc Natl Acad Sci U S A. 2003;100:15194–15199. doi: 10.1073/pnas.2431073100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Fuchs SA, Berger R, de Koning TJ. D-serine: the right or wrong isoform? Brain Res. 2011;1401:104–117. doi: 10.1016/j.brainres.2011.05.039. [DOI] [PubMed] [Google Scholar]
  • 195.Paula-Lima AC, Brito-Moreira J, Ferreira ST. Deregulation of excitatory neurotransmission underlying synapse failure in Alzheimer’s disease. J Neurochem. 2013;126:191–202. doi: 10.1111/jnc.12304. [DOI] [PubMed] [Google Scholar]
  • 196.Sasabe J, Chiba T, Yamada M, Okamoto K, Nishimoto I, Matsuoka M, et al. D-serine is a key determinant of glutamate toxicity in amyotrophic lateral sclerosis. EMBO J. 2007;26:4149–4159. doi: 10.1038/sj.emboj.7601840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Sasabe J, Aiso S. Aberrant control of motoneuronal excitability in amyotrophic lateral sclerosis: excitatory glutamate/D-serine vs. inhibitory glycine/gamma-aminobutanoic acid (GABA) Chem Biodivers. 2010;7:1479–1490. doi: 10.1002/cbdv.200900306. [DOI] [PubMed] [Google Scholar]
  • 198.Magistretti PJ. Neuron-glia metabolic coupling and plasticity. J Exp Biol. 2006;209:2304–2311. doi: 10.1242/jeb.02208. [DOI] [PubMed] [Google Scholar]
  • 199.Pellerin L, Magistretti PJ. Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci U S A. 1994;91:10625–10629. doi: 10.1073/pnas.91.22.10625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Berl S, Clarke DD, Nicklas WJ. Compartmentation of citric acid cycle metabolism in brain: effect of aminooxyacetic acid, ouabain and Ca2+ on the labelling of glutamate, glutamine, aspartate and gaba by [1-14C]acetate, [U-14C]glutamate and [U-14C]asparate. J Neurochem. 1970;17:999–1007. doi: 10.1111/j.1471-4159.1970.tb02253.x. [DOI] [PubMed] [Google Scholar]
  • 201.Pierre K, Magistretti PJ, Pellerin L. MCT2 is a major neuronal monocarboxylate transporter in the adult mouse brain. J Cereb Blood Flow Metab. 2002;22:586–595. doi: 10.1097/00004647-200205000-00010. [DOI] [PubMed] [Google Scholar]
  • 202.Hertz L, Peng L, Dienel GA. Energy metabolism in astrocytes: high rate of oxidative metabolism and spatiotemporal dependence on glycolysis/glycogenolysis. J Cereb Blood Flow Metab. 2007;27:219–249. doi: 10.1038/sj.jcbfm.9600343. [DOI] [PubMed] [Google Scholar]
  • 203.Dienel GA. Brain lactate metabolism: the discoveries and the controversies. J Cereb Blood Flow Metab. 2012;32:1107–1138. doi: 10.1038/jcbfm.2011.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Simpson IA, Carruthers A, Vannucci SJ. Supply and demand in cerebral energy metabolism: the role of nutrient transporters. J Cereb Blood Flow Metab. 2007;27:1766–1791. doi: 10.1038/sj.jcbfm.9600521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Mangia S, DiNuzzo M, Giove F, Carruthers A, Simpson IA, Vannucci SJ. Response to ‘comment on recent modeling studies of astrocyte-neuron metabolic interactions’: much ado about nothing. J Cereb Blood Flow Metab. 2011;31:1346–1353. doi: 10.1038/jcbfm.2011.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Magistretti PJ, Allaman I. A Cellular Perspective on Brain Energy Metabolism and Functional Imaging. Neuron. 2015;86:883–901. doi: 10.1016/j.neuron.2015.03.035. [DOI] [PubMed] [Google Scholar]
  • 207.Schmidt S, Richter M, Montag D, Sartorius T, Gawlik V, Hennige AM, et al. Neuronal functions, feeding behavior, and energy balance in Slc2a3+/− mice. Am J Physiol Endocrinol Metab. 2008;295:E1084–1094. doi: 10.1152/ajpendo.90491.2008. [DOI] [PubMed] [Google Scholar]
  • 208.Wang D, Pascual JM, Yang H, Engelstad K, Mao X, Cheng J, et al. A mouse model for Glut-1 haploinsufficiency. Hum Mol Genet. 2006;15:1169–1179. doi: 10.1093/hmg/ddl032. [DOI] [PubMed] [Google Scholar]
  • 209.Bowling AC, Beal MF. Bioenergetic and oxidative stress in neurodegenerative diseases. Life Sci. 1995;56:1151–1171. doi: 10.1016/0024-3205(95)00055-b. [DOI] [PubMed] [Google Scholar]
  • 210.Slosman DO, Ludwig C, Zerarka S, Pellerin L, Chicherio C, de Ribaupierre A, et al. Brain energy metabolism in Alzheimer’s disease: 99mTc-HMPAO SPECT imaging during verbal fluency and role of astrocytes in the cellular mechanism of 99mTc-HMPAO retention. Brain Res Brain Res Rev. 2001;36:230–240. doi: 10.1016/s0165-0173(01)00099-6. [DOI] [PubMed] [Google Scholar]
  • 211.Finsterer J. Parkinson’s syndrome and Parkinson’s disease in mitochondrial disorders. Mov Disord. 2011;26:784–791. doi: 10.1002/mds.23651. [DOI] [PubMed] [Google Scholar]
  • 212.Kennedy AM, Frackowiak RS, Newman SK, Bloomfield PM, Seaward J, Roques P, et al. Deficits in cerebral glucose metabolism demonstrated by positron emission tomography in individuals at risk of familial Alzheimer’s disease. Neurosci Lett. 1995;186:17–20. doi: 10.1016/0304-3940(95)11270-7. [DOI] [PubMed] [Google Scholar]
  • 213.Merlini M, Meyer EP, Ulmann-Schuler A, Nitsch RM. Vascular beta-amyloid and early astrocyte alterations impair cerebrovascular function and cerebral metabolism in transgenic arcAbeta mice. Acta Neuropathol. 2011;122:293–311. doi: 10.1007/s00401-011-0834-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Ding F, Yao J, Rettberg JR, Chen S, Brinton RD. Early decline in glucose transport and metabolism precedes shift to ketogenic system in female aging and Alzheimer’s mouse brain: implication for bioenergetic intervention. PLoS One. 2013;8:e79977. doi: 10.1371/journal.pone.0079977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Nilsen LH, Witter MP, Sonnewald U. Neuronal and astrocytic metabolism in a transgenic rat model of Alzheimer’s disease. J Cereb Blood Flow Metab. 2014;34:906–914. doi: 10.1038/jcbfm.2014.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Ohman A, Forsgren L. NMR metabonomics of cerebrospinal fluid distinguishes between Parkinson’s disease and controls. Neurosci Lett. 2015;594:36–39. doi: 10.1016/j.neulet.2015.03.051. [DOI] [PubMed] [Google Scholar]
  • 217.Kong P, Zhang BS, Lei P, Kong XD, Zhang SS, Li D, et al. Neurotoxicity of cerebro-spinal fluid from patients with Parkinson’s disease on mesencephalic primary cultures as an in vitro model of dopaminergic neurons. Mol Med Rep. 2015;12:2217–2224. doi: 10.3892/mmr.2015.3575. [DOI] [PubMed] [Google Scholar]
  • 218.Ferraiuolo L, Higginbottom A, Heath PR, Barber S, Greenald D, Kirby J, et al. Dysregulation of astrocyte-motoneuron cross-talk in mutant superoxide dismutase 1-related amyotrophic lateral sclerosis. Brain. 2011;134:2627–2641. doi: 10.1093/brain/awr193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Brockmann K, Pouwels PJ, Christen HJ, Frahm J, Hanefeld F. Localized proton magnetic resonance spectroscopy of cerebral metabolic disturbances in children with neuronal ceroid lipofuscinosis. Neuropediatrics. 1996;27:242–248. doi: 10.1055/s-2007-973772. [DOI] [PubMed] [Google Scholar]
  • 220.Herrmann P, Druckrey-Fiskaaen C, Kouznetsova E, Heinitz K, Bigl M, Cotman SL, et al. Developmental impairments of select neurotransmitter systems in brains of Cln3(Deltaex7/8) knock-in mice, an animal model of juvenile neuronal ceroid lipofuscinosis. J Neurosci Res. 2008;86:1857–1870. doi: 10.1002/jnr.21630. [DOI] [PubMed] [Google Scholar]
  • 221.Pears MR, Codlin S, Haines RL, White IJ, Mortishire-Smith RJ, Mole SE, et al. Deletion of btn1, an orthologue of CLN3, increases glycolysis and perturbs amino acid metabolism in the fission yeast model of Batten disease. Mol Biosyst. 2010;6:1093–1102. doi: 10.1039/b915670d. [DOI] [PubMed] [Google Scholar]
  • 222.Sorg O, Magistretti PJ. Characterization of the glycogenolysis elicited by vasoactive intestinal peptide, noradrenaline and adenosine in primary cultures of mouse cerebral cortical astrocytes. Brain Res. 1991;563:227–233. doi: 10.1016/0006-8993(91)91538-c. [DOI] [PubMed] [Google Scholar]
  • 223.Dienel GA. The metabolic trinity, glucose-glycogen-lactate, links astrocytes and neurons in brain energetics, signaling, memory, and gene expression. Neurosci Lett. 2015 doi: 10.1016/j.neulet.2015.02.052. [DOI] [PubMed] [Google Scholar]
  • 224.Mostert JP, Sijens PE, Oudkerk M, De Keyser J. 1H Magnetic resonance spectroscopy of the internal capsule in human brain: a feasibility study to detect lactate following contralateral motor activity. Eur Radiol. 2005;15:1349–1352. doi: 10.1007/s00330-004-2607-7. [DOI] [PubMed] [Google Scholar]
  • 225.Mostert JP, Koch MW, Heerings M, Heersema DJ, De Keyser J. Therapeutic potential of fluoxetine in neurological disorders. CNS Neurosci Ther. 2008;14:153–164. doi: 10.1111/j.1527-3458.2008.00040.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Steele ML, Fuller S, Maczurek AE, Kersaitis C, Ooi L, Munch G. Chronic inflammation alters production and release of glutathione and related thiols in human U373 astroglial cells. Cell Mol Neurobiol. 2013;33:19–30. doi: 10.1007/s10571-012-9867-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Gavillet M, Allaman I, Magistretti PJ. Modulation of astrocytic metabolic phenotype by proinflammatory cytokines. Glia. 2008;56:975–989. doi: 10.1002/glia.20671. [DOI] [PubMed] [Google Scholar]
  • 228.Yu N, Maciejewski-Lenoir D, Bloom FE, Magistretti PJ. Tumor necrosis factor-alpha and interleukin-1 alpha enhance glucose utilization by astrocytes: involvement of phospholipase A2. Mol Pharmacol. 1995;48:550–558. [PubMed] [Google Scholar]
  • 229.Beal MF. Aging, energy, and oxidative stress in neurodegenerative diseases. Ann Neurol. 1995;38:357–366. doi: 10.1002/ana.410380304. [DOI] [PubMed] [Google Scholar]
  • 230.Beal MF. Mitochondria, NO and neurodegeneration. Biochem Soc Symp. 1999;66:43–54. doi: 10.1042/bss0660043. [DOI] [PubMed] [Google Scholar]
  • 231.Butterfield DA, Perluigi M, Reed T, Muharib T, Hughes CP, Robinson RA, et al. Redox proteomics in selected neurodegenerative disorders: from its infancy to future applications. Antioxid Redox Signal. 2012;17:1610–1655. doi: 10.1089/ars.2011.4109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Ansari MA, Scheff SW. Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J Neuropathol Exp Neurol. 2010;69:155–167. doi: 10.1097/NEN.0b013e3181cb5af4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Lauderback CM, Kanski J, Hackett JM, Maeda N, Kindy MS, Butterfield DA. Apolipoprotein E modulates Alzheimer’s Abeta(1–42)-induced oxidative damage to synaptosomes in an allele-specific manner. Brain Res. 2002;924:90–97. doi: 10.1016/s0006-8993(01)03228-0. [DOI] [PubMed] [Google Scholar]
  • 234.Jenner P. Oxidative stress in Parkinson’s disease. Ann Neurol. 2003;53(Suppl 3):S26–36. doi: 10.1002/ana.10483. discussion S36–28. [DOI] [PubMed] [Google Scholar]
  • 235.Poon HF, Frasier M, Shreve N, Calabrese V, Wolozin B, Butterfield DA. Mitochondrial associated metabolic proteins are selectively oxidized in A30P alpha-synuclein transgenic mice–a model of familial Parkinson’s disease. Neurobiol Dis. 2005;18:492–498. doi: 10.1016/j.nbd.2004.12.009. [DOI] [PubMed] [Google Scholar]
  • 236.Casoni F, Basso M, Massignan T, Gianazza E, Cheroni C, Salmona M, et al. Protein nitration in a mouse model of familial amyotrophic lateral sclerosis: possible multifunctional role in the pathogenesis. J Biol Chem. 2005;280:16295–16304. doi: 10.1074/jbc.M413111200. [DOI] [PubMed] [Google Scholar]
  • 237.Cassarino DS, Bennett JP., Jr An evaluation of the role of mitochondria in neurodegenerative diseases: mitochondrial mutations and oxidative pathology, protective nuclear responses, and cell death in neurodegeneration. Brain Res Brain Res Rev. 1999;29:1–25. doi: 10.1016/s0165-0173(98)00046-0. [DOI] [PubMed] [Google Scholar]
  • 238.Rego AC, Oliveira CR. Mitochondrial dysfunction and reactive oxygen species in excitotoxicity and apoptosis: implications for the pathogenesis of neurodegenerative diseases. Neurochem Res. 2003;28:1563–1574. doi: 10.1023/a:1025682611389. [DOI] [PubMed] [Google Scholar]
  • 239.Mullett SJ, Hinkle DA. DJ-1 deficiency in astrocytes selectively enhances mitochondrial Complex I inhibitor-induced neurotoxicity. J Neurochem. 2011;117:375–387. doi: 10.1111/j.1471-4159.2011.07175.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Lev N, Barhum Y, Ben-Zur T, Melamed E, Steiner I, Offen D. Knocking out DJ-1 attenuates astrocytes neuroprotection against 6-hydroxydopamine toxicity. J Mol Neurosci. 2013;50:542–550. doi: 10.1007/s12031-013-9984-9. [DOI] [PubMed] [Google Scholar]
  • 241.Larsen NJ, Ambrosi G, Mullett SJ, Berman SB, Hinkle DA. DJ-1 knock-down impairs astrocyte mitochondrial function. Neuroscience. 2011;196:251–264. doi: 10.1016/j.neuroscience.2011.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Pehar M, Beeson G, Beeson CC, Johnson JA, Vargas MR. Mitochondria-targeted catalase reverts the neurotoxicity of hSOD1G(9)(3)A astrocytes without extending the survival of ALS-linked mutant hSOD1 mice. PLoS One. 2014;9:e103438. doi: 10.1371/journal.pone.0103438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Vargas MR, Pehar M, Cassina P, Beckman JS, Barbeito L. Increased glutathione biosynthesis by Nrf2 activation in astrocytes prevents p75NTR-dependent motor neuron apoptosis. J Neurochem. 2006;97:687–696. doi: 10.1111/j.1471-4159.2006.03742.x. [DOI] [PubMed] [Google Scholar]
  • 244.Luiro K, Kopra O, Blom T, Gentile M, Mitchison HM, Hovatta I, et al. Batten disease (JNCL) is linked to disturbances in mitochondrial, cytoskeletal, and synaptic compartments. J Neurosci Res. 2006;84:1124–1138. doi: 10.1002/jnr.21015. [DOI] [PubMed] [Google Scholar]
  • 245.Allen NJ. Role of glia in developmental synapse formation. Curr Opin Neurobiol. 2013;23:1027–1033. doi: 10.1016/j.conb.2013.06.004. [DOI] [PubMed] [Google Scholar]
  • 246.Ullian EM, Christopherson KS, Barres BA. Role for glia in synaptogenesis. Glia. 2004;47:209–216. doi: 10.1002/glia.20082. [DOI] [PubMed] [Google Scholar]
  • 247.Stipursky J, Romao L, Tortelli V, Neto VM, Gomes FC. Neuron-glia signaling: Implications for astrocyte differentiation and synapse formation. Life Sci. 2011;89:524–531. doi: 10.1016/j.lfs.2011.04.005. [DOI] [PubMed] [Google Scholar]
  • 248.Eroglu C. The role of astrocyte-secreted matricellular proteins in central nervous system development and function. J Cell Commun Signal. 2009;3:167–176. doi: 10.1007/s12079-009-0078-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Stevens B. Neuron-astrocyte signaling in the development and plasticity of neural circuits. Neurosignals. 2008;16:278–288. doi: 10.1159/000123038. [DOI] [PubMed] [Google Scholar]
  • 250.Risher WC, Eroglu C. Thrombospondins as key regulators of synaptogenesis in the central nervous system. Matrix Biol. 2012;31:170–177. doi: 10.1016/j.matbio.2012.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Rama Rao KV, Curtis KM, Johnstone JT, Norenberg MD. Amyloid-beta inhibits thrombospondin 1 release from cultured astrocytes: effects on synaptic protein expression. J Neuropathol Exp Neurol. 2013;72:735–744. doi: 10.1097/NEN.0b013e31829bd082. [DOI] [PubMed] [Google Scholar]
  • 252.Buee L, Hof PR, Roberts DD, Delacourte A, Morrison JH, Fillit HM. Immunohistochemical identification of thrombospondin in normal human brain and in Alzheimer’s disease. Am J Pathol. 1992;141:783–788. [PMC free article] [PubMed] [Google Scholar]
  • 253.Satoh K, Suzuki N, Yokota H. ADAMTS-4 (a disintegrin and metalloproteinase with thrombospondin motifs) is transcriptionally induced in beta-amyloid treated rat astrocytes. Neurosci Lett. 2000;289:177–180. doi: 10.1016/s0304-3940(00)01285-4. [DOI] [PubMed] [Google Scholar]
  • 254.Tyzack GE, Sitnikov S, Barson D, Adams-Carr KL, Lau NK, Kwok JC, et al. Astrocyte response to motor neuron injury promotes structural synaptic plasticity via STAT3-regulated TSP-1 expression. Nat Commun. 2014;5:4294. doi: 10.1038/ncomms5294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Chen JK, Zhan YJ, Yang CS, Tzeng SF. Oxidative stress-induced attenuation of thrombospondin-1 expression in primary rat astrocytes. J Cell Biochem. 2011;112:59–70. doi: 10.1002/jcb.22732. [DOI] [PubMed] [Google Scholar]
  • 256.Cross AK, Haddock G, Stock CJ, Allan S, Surr J, Bunning RA, et al. ADAMTS-1 and -4 are up-regulated following transient middle cerebral artery occlusion in the rat and their expression is modulated by TNF in cultured astrocytes. Brain Res. 2006;1088:19–30. doi: 10.1016/j.brainres.2006.02.136. [DOI] [PubMed] [Google Scholar]
  • 257.Sennlaub F, Valamanesh F, Vazquez-Tello A, El-Asrar AM, Checchin D, Brault S, et al. Cyclooxygenase-2 in human and experimental ischemic proliferative retinopathy. Circulation. 2003;108:198–204. doi: 10.1161/01.CIR.0000080735.93327.00. [DOI] [PubMed] [Google Scholar]

RESOURCES