Skip to main content
Frontiers in Cellular Neuroscience logoLink to Frontiers in Cellular Neuroscience
. 2019 Feb 12;13:44. doi: 10.3389/fncel.2019.00044

Astrocytes in Neuropathologies Affecting the Frontal Cortex

Ulla-Kaisa Peteri 1, Mikael Niukkanen 1, Maija L Castrén 1,*
PMCID: PMC6379461  PMID: 30809131

Abstract

To an increasing extent, astrocytes are connected with various neuropathologies. Astrocytes comprise of a heterogeneous population of cells with region- and species-specific properties. The frontal cortex exhibits high levels of plasticity that is required for high cognitive functions and memory making this region especially susceptible to damage. Aberrations in the frontal cortex are involved with several cognitive disorders, including Alzheimer’s disease, Huntington’s disease and frontotemporal dementia. Human induced pluripotent stem cells (iPSCs) provide an alternative for disease modeling and offer possibilities for studies to investigate pathological mechanisms in a cell type-specific manner. Patient-specific iPSC-derived astrocytes have been shown to recapitulate several disease phenotypes. Addressing astrocyte heterogeneity may provide an improved understanding of the mechanisms underlying neurodegenerative diseases.

Keywords: astrocyte, frontal cortex, Alzheimer’s disease, Huntington’s disease, frontotemporal dementia, neurodegeneration, induced pluripotent stem cells

Introduction

Astrocytes are implicated as active mediators of synaptic activity, synaptogenesis and neurogenesis and are crucial in maintaining extracellular homeostasis and controlling blood-brain barrier permeability (Zhao et al., 2015; Allen and Lyons, 2018; Marina et al., 2018). Considering their versatile role in regulating brain function, it is no surprise that astrocyte malfunctions have been connected to various neurodegenerative disorders (Phatnani and Maniatis, 2015). Astrocytes are known as a morphologically and functionally diverse population of cells that differ both between distinct brain regions as well as within specific areas (Vasile et al., 2017). This diversity is reflected in their pathological features observed in psychiatric disorders (Rajkowska et al., 2002; Wallingford et al., 2017). Drugs used to treat mood disorders affect astrocytes as well and antidepressant effect on neurons is considered to be partially due to induction of astrocytic release of trophic factors (Marathe et al., 2018). Combined astrocyte and neuron-mediated effects also influence responses of antipsychotics (Khan et al., 2001). However, typical and atypical antipsychotics may display differential effects on the gliotransmitter release and the inflammatory response of astrocytes (Tanahashi et al., 2012; Bobermin et al., 2018). Understanding the function of astrocytes is therefore crucial for disease modeling and for developing treatments. Heterogeneity of the cell population and species-specific differences pose a challenge in the study of astrocytes. A method developed by Takahashi et al. (2007) allows the reprogramming of somatic cells into iPSCs, which can be used to generate patient-specific cells of a desired type (Takahashi et al., 2007). In this review, we describe how iPSC-derived astrocytes have been used to model neurodegenerative disorders involving frontal lobe malfunctions.

Astrocyte Heterogeneity in the Brain

The generation of cortical glia is initiated once neurogenesis has been completed. The temporal patterning is based on a positive feedback signal from new-born neurons (Molofsky and Deneen, 2015; Takouda et al., 2017). Astrocyte progenitors migrate radially, obtaining their region specific properties upon maturation and this process continues postnatally (Colombo et al., 1997; Tsai et al., 2012). Several genes enriched in neuronal progenitors are also expressed in astrocytes, suggesting that astrocytes retain some proliferative potential even in the mature brain (Cahoy et al., 2008). However, only a distinct subset of astrocytes show neurogenic potential (Ghashghaei et al., 2007; Bardehle et al., 2013).

Mature astrocytes can be distinguished based on their morphology and functional properties. In the human cortex, astrocytes are morphologically categorized into four subtypes; interlaminar, protoplasmic, varicose projections and fibrous astroglia, located in the layers I and II, III and IV, V and VI and in the white matter, respectively (Vasile et al., 2017). The brain also contains other, both morphologically and functionally, distinct astrocytes such as elongated radial glia-like tanycytes and unipolar BG with several radially ascending processes. Tanycytes specialized in the regulation of neuroendocrine functions are located in the hypothalamus (Prevot et al., 2018) while BG modulate the efficacy of the synaptic transmission of Purkinje cells in the cerebellum (De Zeeuw and Hoogland, 2015).

Some astrocyte subtypes found in the human cortex are not represented in the rodent brain. Furthermore, human cortical astrocytes exceed their mouse counterparts both in complexity and size, and propagate calcium signals several times faster (Oberheim et al., 2009). Although human and mouse astrocytes share similar properties related to their effects on synapse formation, they differ in their function and transcriptional profiles (Zhang et al., 2016). Species-specific functional differences in glial cells are supported by improved learning and memory in chimeric animals following engrafting human glia into mouse brain (Han et al., 2013).

A higher relative number of astrocytes in the human frontal cortex, compared to that of many other species including other primates, is thought to be due to the high metabolomic cost of maintaining a bigger brain size (Bass et al., 1971; Sherwood et al., 2006). One of the key mechanism’s astrocytes apply to provide energy to neurons is via the astrocyte-neuron lactate shuttle. This metabolomic coupling is known to be crucial for memory formation (Alberini et al., 2018). Astrocytes respond to neuronal activity with spatially and temporally regulated Ca2+ fluctuations that shape neuronal activity via the regulation of the gliotransmitter release (Semyanov, 2019). Molecular and functional variations in astrocytes are considered to contribute to differences in distinct neural circuit signaling (Chai et al., 2017; Morel et al., 2017; Xin and Bonci, 2018). Another central role of astrocytes is the regulation of neurotransmitter uptake including glutamate via excitatory amino acid transporters 1 and 2 (EAAT1, EAAT2, respectively) in humans, or glutamate/aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1) in rodents (Roberts et al., 2014; Meunier et al., 2017). Regulation of the extracellular neurotransmitter levels is affected in a number of neuropsychiatric disorders (John et al., 2012).

Astrocytes as Mediators of Pathologies Affecting the Frontal Cortex

The frontal cortex is responsible for higher executive functions such as cognition and working memory (Fuster, 2002). The expression of genes involved in processes mediating synaptic plasticity, memory and learning is, respectively, enriched in a human and primate frontal cortex (Sjostedt et al., 2015; Garcia-Cabezas et al., 2017). A high level of flexibility is necessary for learning and memory functions but may also lead to increased structural vulnerability, which may explain why aberrations in the frontal cortex are connected to several neuropathologies (John et al., 2012; Feresten et al., 2013; Torres-Platas et al., 2016).

Astrocytes contribute to the regulation of neuronal activity that is altered in several frontal cortex pathologies (Braun et al., 2009; Cao et al., 2013; Lima et al., 2014; Bull et al., 2015; Ebrahimi et al., 2016; Beamer et al., 2017). A common feature for brain diseases is the activation of astrocytes into an inflammatory, reactive state (Chanaday and Roth, 2016). White matter astrocytes in the frontal cortex appear to be especially vulnerable to ischemic stroke, leading to disrupted gliovascular interactions caused by astrogliosis (Chen et al., 2016). Astrogliosis in the frontal cortex, upon aging, is also linked to mood disorders (Miguel-Hidalgo et al., 2000; Narita et al., 2006). Neurodegenerative disorders have overlapping characteristics suggesting common underlying pathological mechanisms. For instance, CTE caused by repeated head injuries, displays a similar accumulation of neurofibrillary tangles to that in AD but can be differentiated from AD by astrocytic tangles that are considered a hallmark of CTE (Turner et al., 2016; Hsu et al., 2018). Below, examples of neurodegenerative disorders with fronto-temporal pathologies and studies employing iPSC-derived astrocytes are described.

Alzheimer’s Disease

Alzheimer’s disease is a progressive neurodegenerative disease that manifests through cognitive impairment, motor abnormalities and behavioral changes. AD pathology is hallmarked by the accumulation of insoluble amyloid-β (Aβ) plaques, amyloid deposits in the blood vessel walls and aggregation of the microtubule protein tau, within neurons. The abnormalities seen in AD usually occur first in the frontotemporal region then spread progressively to other areas of the neocortex (Masters et al., 2015).

The contribution of astrocytes in AD pathology comprises of both the loss of neuroprotective features as well as the acquirement of pathological properties. Initially, astrocytes uptake and degrade Aβ and have a neuroprotective role. However, disease progression often leads to impaired astrocytic Aβ clearance and induces toxic gain-of-functions that contribute to disease progression (Garwood et al., 2017). Neural plaques adjacent to GFAP expressing astrocytes are known to induce hypertrophy and there is also evidence showing that astrocyte reactivity may precede plaque formation (Teneka et al., 2005; Olabarria et al., 2010; Rodrieguez-Veitez et al., 2015). Morphological aberrances in AD astrocytes that compromise vascular coverage are detrimental to neurovascular regulation, while disrupted potassium (K+) mediated neurovascular coupling, due to downregulation of K+ channels Kir4.1 and BKCa, result in abnormal regional cerebral blood flow (Acosta et al., 2017).

Aβ has been shown to alter the expression of metabotropic glutamate receptor 5 (mGluR5) and nicotinic acetylcholine receptors (nAchRs) in astrocytes, which leads to changes in Ca2+ homeostasis and signaling (Haughey and Mattson, 2003; Xiu et al., 2005; Lim et al., 2013). Excitotoxicity is a common characteristic of AD and astrocytes contribute to excessive glutamate signaling. Insufficient clearance of glutamate is connected to the reduced expression of glutamate transporters and their aberrant trafficking, which has been linked to altered cholesterol synthesis (Masliah et al., 1996; Tian et al., 2010; Merlini et al., 2011; Talantova et al., 2013). Furthermore, the release of glutamate has been shown to be enhanced in AD astrocytes (Talantova et al., 2013).

Reactivity is a common feature of AD astrocytes. Aβ induces the astrocytic release of pro-inflammatory mediators and, in turn, pro-inflammatory signals stimulate astrocytic Aβ production leading to a positive feedback loop between astrocyte Aβ response and production (Gonzáles-Reyes et al., 2017). S100β-positive astrocytes are connected to AD pathology and they are reduced following immunization against Aβ (Neus Bosch et al., 2015). S100β expressed by astrocytes is important for the regulation of neuronal oscillations associated with cognitive flexibility and depressive behavior (Stroth and Svenningsson, 2015; Brockett et al., 2018).

Glucose hypometabolism can precede clinical symptoms of AD (Mosconi et al., 2006). There is evidence that carriers of apolipoprotein E𝜀4 (APOE𝜀4) allele, with an increased risk for AD, have lower levels of glucose metabolism in various brain regions, including the prefrontal cortex, before the manifestation of clinical symptoms (Reiman et al., 2004). Dementia in AD is related to altered lactate processing. Under normal circumstances lactate-producing enzymes are down-regulated with age and an increase in the expression of these enzymes improve memory in wild type mice but leads to memory deficits in AD mice (Harris et al., 2016). Astrocyte defects in AD have been described extensively in a recent review (Acosta et al., 2017).

Huntington’s Disease

Huntington’s disease (HD) is characterized by motor dysfunction, cognitive impairment and neuropsychiatric features. HD is an inherited neurological disorder caused by CAG trinucleotide repeat expansion in the gene encoding Htt. The expansion gives rise to a mutated form of Htt (mHtt) with an abnormally long polyglutamine sequence which leads to the formation of mHtt aggregates (Bates et al., 2015). Clearance of aggregates is more efficient from astrocytes than from neurons, rendering astrocytes more resistant to mHtt accumulation (Zhao et al., 2016; Jansen et al., 2017; Zhao T. et al., 2017). Eventual accumulation of mHtt into astrocytes results in altered glutamate homeostasis and, sub-sequentially, neuronal excitotoxicity (Shin et al., 2005; Bradford et al., 2009). In addition to the enhanced release of glutamate, the presence of mHtt in astrocytes decreases the expression of glutamate transporters in an age-dependent manner (Lievens et al., 2001; Estrada-Sanchez et al., 2009; Faideau et al., 2010; Lee et al., 2013). However, excitotoxicity in HD neurons has also been reported without defects in the glutamate clearance (Parsons et al., 2016).

Huntington’s disease astrocytes possess an altered K+ signaling due to the decreased expression of Kir4.1 (Tong et al., 2014; Zhang et al., 2018). Restoration of Kir4.1 function can ameliorate impaired GLT1-mediated homeostasis and, sub-sequentially astrocyte Ca2+ signaling, implying a causative effect of Kir4.1 dysfunction on these mechanisms (Tong et al., 2014; Jiang et al., 2016). Kir4.1 defects precede the appearance of reactive astrocytes, indicating that inflammation is a secondary effect of HD pathology possibly induced by neurotoxicity (Tong et al., 2014).

Both reactive astrocytes and microglia have been implicated in the pathogenesis of HD (Khakh et al., 2017). Microglia promote the reactivity of astrocytes via the secretion of pro-inflammatory factors such as Il-1α, TNFα, and C1q (Liddelow et al., 2017). Reactive astrocytes have an impaired ability for synaptic maintenance and decreased phagocytic capacity (Bradford et al., 2009; Haim et al., 2015). Additionally, they promote degeneration of a subset of neurons and mature oligodendrocytes (Liddelow et al., 2017). Activation of the JAK/STAT3 pathway appears to be a common pathological feature of HD and AD. Astrocyte specific inhibition of this pathway, in animal models, reduces the reactive astrocyte phenotype (Haim et al., 2015). Interestingly, some studies have shown that reactive astrocytes can also have a neuroprotective role in HD (Haim et al., 2015; Liddelow et al., 2017).

Accumulation of mHtt disrupts exosome secretion from astrocytes (Hong et al., 2017). This can be connected to the reduced BDNF release from astrocytes (Hong et al., 2016). BDNF signaling is associated with HD pathogenesis and restoration of BDNF release from astrocytes has been shown to have neuroprotective effects (Giralt et al., 2010; Hong et al., 2016; Reick et al., 2016). However, there is also evidence that at early stages of HD, TrkB signaling is altered due to an indirect effect of p75 neurotrophic receptor (p75NTR) activity, indicating that signaling defects may precede aberrant secretion of BDNF, a ligand of both TrkB and p75NTR (Plotkin et al., 2014).

Frontotemporal Dementia

Frontotemporal dementia (FTD) is an umbrella term for neurodegenerative diseases affecting the frontal or temporal lobes. Behavioral changes and deficits in executive functioning and language characterize FTD (Bang et al., 2015). The role of astrocytes in FTD is not fully understood. However, FTD pathology is known to involve astrogliosis that occurs at an early stage of the disease progression and precedes neuronal loss (Su et al., 2000; Kersaitis et al., 2004). Astrocytic degeneration is marked by the expression of apoptotic markers, such as caspase-3, and morphological changes (Su et al., 2000; Broe et al., 2004). Apoptotic astrocytes in FTD have been correlated with the degree of frontotemporal atrophy and significant astrogliosis has been observed to overlap with areas showing disturbed cerebral perfusion (Martinac et al., 2001; Broe et al., 2004). In theory, astrocyte degeneration could cause disruptions similar to those seen in AD and HD, but the possible role of astrocyte degeneration in FTD pathogenesis remains unclear (Su et al., 2000).

Modeling Psychopathologies Using Human Cells

In recent years, a number of astrocyte differentiation methods have been developed and advances on the use of iPSC-derived astrocytes have been reviewed in a recent paper (Zheng et al., 2018). Below, the application of iPSC-derived astrocytes and their use to model frontal cortex defects are discussed. The studies represented are summarized in Table 1.

Table 1.

Summary of studies on iPSC-derived astrocytes in modeling frontocortical pathologies.

Disease Mutation Astrocyte differentiation Key findings Reference
AD PSEN (FAD)
APOE4+/+ (SAD)
iPSC-derived NPC conversion to astroglia in the presence of CNTF, BMP2, FGF2, EGF. Both SAD and FAD astrocyted exhibited reduced morphological heterogeneity, aberrant expression of S100β, and altered cytokine secretion. Altered EAAT1 distribution only seen in SAD. Jones et al., 2017
AD PSEN1 ΔE9 Expansion of NPCs in suspension culture in the presence of FGF2 and EGF. Astrocyte differentiation in the presence of CNTF and BMP4. Astrocytes produce Aβ with aberrant uptake. Altered cytokine secretion, increased production of ROS. Induce aberrant Ca2+ signaling in healthy neurons. Oksanen et al., 2017
AD APOE𝜀4/𝜀4
APOE𝜀3/𝜀3
Neural induction in suspension culture followed by neural rosette formation and generation of NPCs. Astrocyte differentiation in the presence of CNTF, BMP4, and Heregulin-β. ApoE isoforms have distinct properties with APOE𝜀3/𝜀3 astrocytes having greater neuroprotective and synaptogenetic potential. Zhao J. et al., 2017
AD APOE4 APOE3 NPCs generation in an adherent culture. Astrocyte differentiation in the presence of FGF2 and BMP4. Aberrant production and uptake of Aβ. In 3D culture Aβ starts to accumulates in the organoids. Changes in the gene expressions related to lipid metabolism. Lin et al., 2018
AD APP-KO
APPswe/swe APP V717F
NPCs were generated in an adherent culture. Astrocyte differentiation in a suspension culture in the presence of FBS and EGF. Astrocytes have aberrant cholesterol metabolism. Lipoprotein and Aβ endocytosis are impaired. Fong et al., 2018
HD Hit NPCs were generated in suspension in the presence of growth factors. Astrocyte differentiation was induced by plating the NPCs in the absence of FGF2. Generated astrocytes exhibited enhanced cytoplasmic vacuolation under basal conditions. Juopperi et al., 2012
HD Hit Astrocyte differentiation of iPSCs in neural differentiation medium in the presence of CNTF. Blocking soluble TNFα suppresses pathological inflammatory response in astrocytes. Hsiao et al., 2014
HD Hit Astrocyte differentiation of iPSCs in neural differentiation medium in the presence of CNTF. Increased inflammatory response and expression of VEGF-A in HD-astrocytes lead to compromised vascular reactivity. Hsiao et al., 2015
FTD/ALS TDP-43
M337V
NPCs were cultured in the presence of LIF and EGF in followed by expansion with FGF2 and EGF. Terminal differentiation into astrocytes induced by growth factor withdrawal. Astrocytes showed accumulation of cytoplasmic TDP-43 resulting in lowered cell survival. Serio et al., 2013
FTD MAPT
N279K
NPC differentiation induced by lentiviral induction of SOX10 followed by treatment with SAG, PDGF, FGF2, NT3, IGF, and LDN. Astrocyte differentiation in the presence of IGF, CNTF, and dbcAMP Astrocytes showed changes in TAU expression, hypertrophy, increased vulnerability to oxidative stress and altered transcriptomic profile. In co-culture system FTD astrocytes altered responses to oxidative stress in healthy neurons. Hallmann et al., 2017

Defects in both the clearance and production of Aβ, associated with AD, can also be seen in iPSC-derived AD astrocytes and appear to involve aberrant lipid metabolism (Oksanen et al., 2017; Fong et al., 2018; Lin et al., 2018). When studying the effects of APOE genotype Lin et al. (2018) demonstrated that APOE4 astrocytes show differences in the transcriptomic profile compared to isogenic APOE3 cells, as well as a diminished ability in clearing Aβ (Lin et al., 2018). The role of ApoE in the Aβ clearance is still unresolved and some studies claim that ApoE is crucial for the degradation and removal of Aβ, while others have shown that ApoE promotes neurodegeneration (Holtzman et al., 1999; Koistinaho et al., 2004; Liao et al., 2014; Shi et al., 2017). In co-culture studies APOE3 exhibited a greater ability to promote neuronal support and synaptogenesis (Zhao J. et al., 2017). Different properties of APOE isoforms in human astrocytes are in agreement with previous studies in mice (Wang et al., 2005). Jones et al. (2017) studied the function of AD astrocytes generated from iPSCs modeling early-onset FAD with mutation in PSEN and late-onset SAD with the APOE4 genotype. Both FAD and SAD astrocytes showed reduced morphological heterogeneity and aberrant expression of S100β. However, altered distribution of EAAT1 was only seen in SAD astrocytes (Jones et al., 2017). Altered secretion of inflammatory cytokines was found in both FAD and SAD, as well as in astrocytes with the PSEN1 ΔE9 genotype generated by Oksanen et al. (Jones et al., 2017; Oksanen et al., 2017). PSEN1 ΔE9 astrocytes also displayed changes in Ca2+ homeostasis, mitochondrial metabolism, ROS production and lactate secretion, thus covering all classical features of AD pathology (Oksanen et al., 2017).

Inflammatory responses were studied by Hsiao et al. (2015) in iPSC-derived HD astrocytes and an increase in the expression of VEGF-A, with further up-regulation after inflammatory cytokine treatment, was found. This leads to the enhanced proliferation of endothelial cells and the compromised survival of pericytes. As a result, poor pericyte coverage of blood vessels cause vascular reactivity and disrupts the blood-brain-barrier (Hsiao et al., 2015). Additionally, they demonstrated that the TNFα inhibitor XPro1595 successfully suppressed the inflammatory responses both in human astrocytes as well as primary astrocytes propagated from the brain of a transgenic HD mouse model (R6/2) (Hsiao et al., 2014). Juopperi et al. (2012) showed that HD astrocytes display increased cytoplasmic vacuolization (Juopperi et al., 2012). This phenotype is also present in HD lymphoblasts (Nagata et al., 2004; Martinez-Vicente et al., 2010). The findings in iPSC-derived HD astrocytes are consistent with astrogliosis as a key characteristic of HD pathology.

Frontotemporal dementia astrocytes, derived from iPSCs with mutations in genes encoding microtubule-associated protein TAU (MAPT) and TDP-34, demonstrated an increased susceptibility to oxidative stress and compromised survival (Serio et al., 2013; Hallmann et al., 2017). In M337V TDP-34 astrocytes, lowered survival paralleled the accumulation of TDP-43 (Serio et al., 2013). This phenomenon has been implicated in astrocyte dysfunction in CTE (Jayakumar et al., 2017). In N279K MAPT astrocytes, the expression of 4R-TAU isoform was increased as reported in FTD patients (Ghetti et al., 2015; Hallmann et al., 2017). N279K MAPT astrocytes displayed morphological changes and increased GFAP expression, usually linked to reactivity, as well as altered gene expression profiles. In co-culture assays with healthy neurons, N279K MAPT astrocytes increased the vulnerability of neurons to oxidative stress (Hallmann et al., 2017). However, M337V TDP-34 astrocytes did not exert toxic effects on neurons, although astrocytic expression of mutated TDP-43 has been reported to induce neuronal cell death (Tong et al., 2013; Serio et al., 2013) suggesting that other cell types, such as microglia, are required for the neurotoxic effect. Altogether, the results indicate that astrocyte degeneration is a common feature of FTD.

Conclusion

An increasing number of studies have connected astrocyte defects to frontal cortex pathologies. Species-specificity of astrocytes poses a challenge in translating results obtained from animal studies to humans, and patient-derived iPSCs offer an alternative to disease modeling. Studies presented above demonstrate that iPSC-derived astrocytes successfully recapitulate various disease phenotypes. Further challenges still include addressing the heterogeneity within the astrocyte population and developing protocols to generate regionally defined human astrocyte subtypes.

Author Contributions

U-KP and MN wrote the manuscript in consultation with MC. MC performed the critical revision of the paper.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The authors wish to express their thanks for the financial support of the Academy of Finland, the Arvo and Lea Ylppö Foundation, the Finnish Konkordia Fund, the Finnish Cultural Foundation, and Finnish Brain Foundation.

Abbreviations

5-HT

5-hydroxytryptamine receptor

amyloid β

AD

Alzheimer’s disease

APOE

apolipoprotein E

APP

amyloid precursor protein

BDNF

brain-derived neurotrophic factor

BG

Bergmann glia

cAMP

cyclic adenosine monophosphate

CTE

chronic traumatic encephalopathy

C1q

complement component 1q

EAAT

excitatory amino acid transporter

FAD

familial Alzheimer’s disease

FTD

frontotemporal dementia

GFAP

glial fibrillary acidic protein

GLAST

glutamate aspartate transporter

GLT-1

glutamate transporter 1

HD

Huntington’s disease

Htt

huntingtin

Il-1α

interleukin 1α

iPSC

induced pluripotent stem cell

JAK/STAT3

Janus kinase/signal transducers and activators of transcription 3

ROS

reactive oxygen species

SAD

sporadic Alzheimer’s disease

TNFα

tumor necrosis factor α

TDP-34

transactive response DNA-binding protein 34

TrkB

tyrosine-related kinase B

VEGF-A

vascular endothelial growth factor A

References

  1. Acosta C., Anderson H. D., Anderson C. M. (2017). Astrocyte dysfunction in Alzheimer disease. J. Neurosci. Res. 95 2430–2447. 10.1002/jnr.24075 [DOI] [PubMed] [Google Scholar]
  2. Alberini C. M., Cruz E., Descalzi G., Bessieres B., Gao V. (2018). Astrocyte glycogen and lactate: new insights into learning and memory mechanisms. Glia 66 1244–1262. 10.1002/glia.23250 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Allen N. J., Lyons D. A. (2018). Glia as architects of central nervous system formation and function. Science 362 181–185. 10.1126/science.aat0473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bang J., Spina S., Miller B. L. (2015). Non-Alzheimer’s dementia 1: frontotemporal dementia. Lancet 386 1672–1682. 10.1016/S0140-6736(15)00461-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bardehle S., Krüger M., Buggenthin F., Schwausch J., Ninkovic J., Clevers H., et al. (2013). Live imaging of astrocyte responses to acute injury reveals selective juxtavascular proliferation. Nat. Neurosci. 16:580. 10.1038/nn.3371 [DOI] [PubMed] [Google Scholar]
  6. Bass N. H., Hess H. H., Pope A., Thalheimer C. (1971). Quantitative cytoarchitectonic distribution of neurons, glia, and DNA in rat cerebral cortex. J. Comp. Neurol. 143 481–490. 10.1002/cne.901430405 [DOI] [PubMed] [Google Scholar]
  7. Bates G. P., Dorsey R., Gusella J. F., Hayden M. R., Kay C., Leavitt B. R., et al. (2015). Huntington disease. Nat. Rev. Dis. Primers 1:15005. 10.1038/nrdp.2015.5 [DOI] [PubMed] [Google Scholar]
  8. Beamer E., Kovacs G., Sperlagh B. (2017). ATP released from astrocytes modulates action potential threshold and spontaneous excitatory postsynaptic currents in the neonatal rat prefrontal cortex. Brain Res. Bull. 135 129–142. 10.1016/j.brainresbull.2017.10.006 [DOI] [PubMed] [Google Scholar]
  9. Bobermin L. D., da Silva A., Souza D. O., Quincozes-Santos A. (2018). Differential effects of typical and atypical antipsychotics on astroglial cells in vitro. Int. J. Dev. Neurosci. 69 1–9. 10.1016/j.ijdevneu.2018.06.001 [DOI] [PubMed] [Google Scholar]
  10. Bradford J., Shin J.-Y., Roberts M., Wang C.-E., Li X.-J., Li S. (2009). Expression of mutant huntingtin in mouse brain astrocytes causes age-dependent neurological symptoms. Proc. Natl. Acad. Sci. U.S.A. 10622480–22485. 10.1073/pnas.0911503106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Braun K., Antemano R., Helmeke C., Buchner M., Poeggel G. (2009). Juvenile separation stress induces rapid region- and layer-specific changes in S100ss- and glial fibrillary acidic protein-immunoreactivity in astrocytes of the rodent medial prefrontal cortex. Neuroscience 160 629–638. 10.1016/j.neuroscience.2009.02.074 [DOI] [PubMed] [Google Scholar]
  12. Brockett A. T., Kane G. A., Monari P. K., Briones B. A., Vigneron P. A., Barber G. A., et al. (2018). Evidence supporting a role for astrocytes in the regulation of cognitive flexibility and neuronal oscillations through the Ca2+ binding protein S100beta. PLoS One 13:e0195726. 10.1371/journal.pone.0195726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Broe M., Kril J., Halliday G. M. (2004). Astrocytic degeneration relates to the severity of disease in frontotemporal dementia. Brain 127(Pt 10), 2214–2220. 10.1093/brain/awh250 [DOI] [PubMed] [Google Scholar]
  14. Bull C., Syed W. A., Minter S. C., Bowers M. S. (2015). Differential response of glial fibrillary acidic protein-positive astrocytes in the rat prefrontal cortex following ethanol self-administration. Alcohol. Clin. Exp. Res. 39 650–658. 10.1111/acer.12683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cahoy J. D., Emery B., Kaushal A., Foo L. C., Zamanian J. L., Christopherson K. S., et al. (2008). A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28 264–278. 10.1523/JNEUROSCI.4178-07.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cao X., Li L. P., Wang Q., Wu Q., Hu H. H., Zhang M., et al. (2013). Astrocyte-derived ATP modulates depressive-like behaviors. Nat. Med. 19 773–777. 10.1038/nm.3162 [DOI] [PubMed] [Google Scholar]
  17. Chai H., Diaz-Castro B., Shigetomi E., Monte E., Octeau J. C., Yu X., et al. (2017). Neural circuit-specialized astrocytes: transcriptomic, proteomic, morphological, and functional evidence. Neuron 95 531–549.e9. 10.1016/j.neuron.2017.06.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chanaday N. L., Roth G. A. (2016). Microglia and astrocyte activation in the frontal cortex of rats with experimental autoimmune encephalomyelitis. Neuroscience 314 160–169. 10.1016/j.neuroscience.2015.11.060 [DOI] [PubMed] [Google Scholar]
  19. Chen A., Akinyemi R. O., Hase Y., Firbank M. J., Ndung’u M. N., Foster V., et al. (2016). Frontal white matter hyperintensities, clasmatodendrosis and gliovascular abnormalities in ageing and post-stroke dementia. Brain 139 242–258. 10.1093/brain/awv328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Colombo J. A., Lipina S., Yanez A., Puissant V. (1997). Postnatal development of interlaminar astroglial processes in the cerebral cortex of primates. Int. J. Dev. Neurosci. 15 823–833. 10.1016/S0736-5748(97)00043-9 [DOI] [PubMed] [Google Scholar]
  21. De Zeeuw C. I., Hoogland T. M. (2015). Reappraisal of Bergmann glial cells as modulators of cerebellar circuit function. Front. Cell. Neurosci. 9:246. 10.3389/fncel.2015.00246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Ebrahimi M., Yamamoto Y., Sharifi K., Kida H., Kagawa Y., Yasumoto Y., et al. (2016). Astrocyte-expressed FABP7 regulates dendritic morphology and excitatory synaptic function of cortical neurons. Glia 64 48–62. 10.1002/glia.22902 [DOI] [PubMed] [Google Scholar]
  23. Estrada-Sanchez A. M., Montiel T., Segovia J., Massieu L. (2009). Glutamate toxicity in the striatum of the R6/2 Huntington’s disease transgenic mice is age-dependent and correlates with decreased levels of glutamate transporters. Neurobiol. Dis. 34 78–86. 10.1016/j.nbd.2008.12.017 [DOI] [PubMed] [Google Scholar]
  24. Faideau M., Kim J., Cormier K., Gilmore R., Welch M., Auregan G., et al. (2010). In vivo expression of polyglutamine-expanded huntingtin by mouse striatal astrocytes impairs glutamate transport: a correlation with Huntington’s disease subjects. Hum. Mol. Genet. 19 3053–3067. 10.1093/hmg/ddq212 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Feresten A. H., Barakauskas V., Ypsilanti A., Barr A. M., Beasley C. L. (2013). Increased expression of glial fibrillary acidic protein in prefrontal cortex in psychotic illness. Schizophr. Res. 150 252–257. 10.1016/j.schres.2013.07.024 [DOI] [PubMed] [Google Scholar]
  26. Fong L. K., Yang M. M., Dos Santos Chaves R., Reyna S. M., Langness V. F., Woodruff G., et al. (2018). Full-length amyloid precursor protein regulates lipoprotein metabolism and amyloid-beta clearance in human astrocytes. J. Biol. Chem. 293 11341–11357. 10.1074/jbc.RA117.000441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Fuster J. M. (2002). Frontal lobe and cognitive development. J. Neurocytol. 31 373–385. 10.1023/A:1024190429920 [DOI] [PubMed] [Google Scholar]
  28. Garcia-Cabezas M. A., Joyce M. K. P., John Y. J., Zikopoulos B., Barbas H. (2017). Mirror trends of plasticity and stability indicators in primate prefrontal cortex. Eur. J. Neurosci. 46 2392–2405. 10.1111/ejn.13706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Garwood C. J., Ratcliffe L. E., Simpson J. E., Heath P. R., Ince P. G., Wharton S. B. (2017). Review: astrocytes in Alzheimer’s disease and other age-associated dementias: a supporting player with a central role. Neuropathol. Appl. Neurobiol. 43 281–298. 10.1111/nan.12338 [DOI] [PubMed] [Google Scholar]
  30. Ghashghaei H. T., Weimer J. M., Schmid R. S., Yokota Y., McCarthy K. D., Popko B., et al. (2007). Reinduction of ErbB2 in astrocytes promotes radial glial progenitor identity in adult cerebral cortex. Genes Dev. 21 3258–3271. 10.1101/gad.1580407 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Ghetti B., Oblak A. L., Boeve B. F., Johnson K. A., Dickerson B. C., Goedert M. (2015). Invited review: frontotemporal dementia caused by microtubule-associated protein tau gene (MAPT) mutations: a chameleon for neuropathology and neuroimaging. Neuropathol. Appl. Neurobiol. 41 24–46. 10.1111/nan.12213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Giralt A., Friedman H. C., Caneda-Ferron B., Urban N., Moreno E., Rubio N., et al. (2010). BDNF regulation under GFAP promoter provides engineered astrocytes as a new approach for long-term protection in Huntington’s disease. Gene Ther. 17 1294–1308. 10.1038/gt.2010.71 [DOI] [PubMed] [Google Scholar]
  33. Gonzáles-Reyes R. E., Nava-Mesa M. O., Vargas-Sánchez K., Ariza-Salamanca D., Mora-Muñoz L. (2017). Involvement of astrocytes in Alzheimer’s disease from a neuroinflammatory and oxidative stress perspective. Front. Mol. Neurosci. 10:427 10.3389/fnmol.2017.00427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Haim L. B., Ceyzériat K., Carrillo-de Sauvage M. A., Aubry F., Auregan G., Guillermier M., et al. (2015). The JAK/STAT3 pathway is a common inducer of astrocyte reactivity in Alzheimer’s and Huntington’s diseases. J. Neurosci. 35 2817–2829. 10.1523/JNEUROSCI.3516-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Hallmann A. L., Arauzo-Bravo M. J., Mavrommatis L., Ehrlich M., Ropke A., Brockhaus J., et al. (2017). Astrocyte pathology in a human neural stem cell model of frontotemporal dementia caused by mutant TAU protein. Sci. Rep. 7:42991. 10.1038/srep42991 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Han X., Chen M., Wang F., Windrem M., Wang S., Shanz S., et al. (2013). Forebrain engraftment by human glial progenitor cells enhances synaptic plasticity and learning in adult mice. Cell Stem Cell 12 342–353. 10.1016/j.stem.2012.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Harris R. A., Tindale L., Lone A., Singh O., Macauley S. L., Stanley M., et al. (2016). Aerobic glycolysis in the frontal cortex correlates with memory performance in wild-type mice but not the APP/PS1 mouse model of cerebral amyloidosis. J. Neurosci. 36 1871–1878. 10.1523/JNEUROSCI.3131-15.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Haughey N. J., Mattson M. P. (2003). Alzheimer’s amyloid beta-peptide enhances ATP/gap junction-mediated calcium-wave propagation in astrocytes. Neuromolecular Med. 3 173–180. 10.1385/NMM:3:3:173 [DOI] [PubMed] [Google Scholar]
  39. Holtzman D. M., Bales K. R., Wu S., Bhat P., Parsadanian M., Fagan A. M., et al. (1999). Expression of human apolipoprotein E reduces amyloid-beta deposition in a mouse model of Alzheimer’s disease. J. Clin. Invest. 103 R15–R21. 10.1172/JCI6179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Hong Y., Zhao T., Li X.-J., Li S. (2016). Mutant huntingtin impairs BDNF release from astrocytes by disrupting conversion of Rab3a-GTP into Rab3a-GDP. J. Neurosci. 36 8790–8801. 10.1523/JNEUROSCI.0168-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Hong Y., Zhao T., Li X. J., Li S. (2017). Mutant huntingtin inhibits alphaB-crystallin expression and impairs exosome secretion from astrocytes. J. Neurosci. 37 9550–9563. 10.1523/JNEUROSCI.1418-17.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Hsiao H.-Y., Chen Y.-C., Huang C.-H., Chen C.-C., Hsu Y.-H., Chen H.-M., et al. (2015). Aberrant astrocytes impair vascular reactivity in Huntington disease. Ann. Neurol. 78 178–192. 10.1002/ana.24428 [DOI] [PubMed] [Google Scholar]
  43. Hsiao H. Y., Chiu F. L., Chen C. M., Wu Y. R., Chen H. M., Chen Y. C., et al. (2014). Inhibition of soluble tumor necrosis factor is therapeutic in Huntington’s disease. Hum. Mol. Genet. 23 4328–4344. 10.1093/hmg/ddu151 [DOI] [PubMed] [Google Scholar]
  44. Hsu E. T., Gangolli M., Su S., Holleran L., Stein T. D., Alvarez V. E., et al. (2018). Astrocytic degeneration in chronic traumatic encephalopathy. Acta Neuropathol. 136 955–972. 10.1007/s00401-018-1902-3 [DOI] [PubMed] [Google Scholar]
  45. Jansen A. H., van Hal M., Op den Kelder I. C., Meier R. T., de Ruiter A. A., Schut M. H., et al. (2017). Frequency of nuclear mutant huntingtin inclusion formation in neurons and glia is cell-type-specific. Glia 65 50–61. 10.1002/glia.23050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Jayakumar A. R., Tong X. Y., Shamaladevi N., Barcelona S., Gaidosh G., Agarwal A., et al. (2017). Defective synthesis and release of astrocytic thrombospondin-1 mediates the neuronal TDP-43 proteinopathy, resulting in defects in neuronal integrity associated with chronic traumatic encephalopathy: in vitro studies. J. Neurochem. 140 645–661. 10.1111/jnc.13867 [DOI] [PubMed] [Google Scholar]
  47. Jiang R., Diaz-Castro B., Looger L. L., Khakh B. S. (2016). Dysfunctional calcium and glutamate signaling in striatal astrocytes from Huntington’s disease model mice. J. Neurosci. 36 3453–3470. 10.1523/JNEUROSCI.3693-15.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. John C. S., Smith K. L., Van’t Veer A., Gompf H. S., Carlezon W. A., Jr., et al. (2012). Blockade of astrocytic glutamate uptake in the prefrontal cortex induces anhedonia. Neuropsychopharmacology 37 2467–2475. 10.1038/npp.2012.105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Jones V. C., Atkinson-Dell R., Verkhratsky A., Mohamet L. (2017). Aberrant iPSC-derived human astrocytes in Alzheimer’s disease. Cell Death Dis. 8:e2696. 10.1038/cddis.2017.89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Juopperi T. A., Kim W. R., Chiang C. -H., Yu H., Margolis R. L., Ross C. A., et al. (2012). Astrocytes generated from patient induced pluripotent stem cells recapitulate features of Huntington’s disease patient cells. Mol. Brain 5:17. 10.1186/1756-6606-5-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Kersaitis C., Halliday G. M., Kril J. J. (2004). Regional and cellular pathology in frontotemporal dementia: relationship to stage of disease in cases with and without Pick bodies. Acta Neuropathol. 108 515–523. 10.1007/s00401-004-0917-0 [DOI] [PubMed] [Google Scholar]
  52. Khakh B. S., Beaumont V., Cachope R., Munoz-Sanjuan I., Goldman S. A., Grantyn R. (2017). Unravelling and exploiting astrocyte dysfunction in Huntington’s disease. Trends Neurosci. 40 422–437. 10.1016/j.tins.2017.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Khan Z. U., Koulen P., Rubinstein M., Grandy D. K., Goldman-Rakic P. S. (2001). An astroglia-linked dopamine D2-receptor action in prefrontal cortex. Proc. Natl. Acad. Sci. U.S.A. 98 1964–1969. 10.1073/pnas.98.4.1964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Koistinaho M., Lin S., Wu X., Esterman M., Koger D., Hanson J., et al. (2004). Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-β peptides. Nat. Med. 10 719–726. 10.1038/nm1058 [DOI] [PubMed] [Google Scholar]
  55. Lee W., Reyes R. C., Gottipati M. K., Lewis K., Lesort M., Parpura V., et al. (2013). Enhanced Ca2+-dependent glutamate release from astrocytes of the BACHD Huntington’s disease mouse model. Neurobiol. Dis. 58 192–199. 10.1016/j.nbd.2013.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Liao F., Hori Y., Hudry E., Bauer A. Q., Jiang H., Mahan T. E., et al. (2014). Anti-ApoE antibody given after plaque onset decreases Aβ accumulation and improves brain function in a mouse model of Aβ amyloidosis. J. Neurosci. 34 7281–7292. 10.1523/JNEUROSCI.0646-14.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Liddelow S. A., Guttenplan K. A., Clarke L. E., Bennett F. C., Bohlen C. J., Schirmer L., et al. (2017). Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541 481–487. 10.1038/nature21029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Lievens J. C., Woodman B., Mahal A., Spasic-Boscovic O., Samuel D., Kerkerian-Le Goff L., et al. (2001). Impaired glutamate uptake in the R6 Huntington’s disease transgenic mice. Neurobiol. Dis. 8 807–821. 10.1006/nbdi.2001.0430 [DOI] [PubMed] [Google Scholar]
  59. Lim D., Iyer A., Ronco V., Grolla A. A., Canonico P. L., Aronica E., et al. (2013). Amyloid beta deregulates astroglial mGluR5-mediated calcium signaling via calcineurin and Nf-kB. Glia 61 1134–1145. 10.1002/glia.22502 [DOI] [PubMed] [Google Scholar]
  60. Lima A., Sardinha V. M., Oliveira A. F., Reis M., Mota C., Silva M. A., et al. (2014). Astrocyte pathology in the prefrontal cortex impairs the cognitive function of rats. Mol. Psychiatry 19 834–841. 10.1038/mp.2013.182 [DOI] [PubMed] [Google Scholar]
  61. Lin Y. T., Seo J., Gao F., Feldman H. M., Wen H. L., Penney J., et al. (2018). APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 98 1141–1154.e7. 10.1016/j.neuron.2018.05.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Marathe S. V., D’Almeida P. L., Virmani G., Bathini P., Alberi L. (2018). Effects of monoamines and antidepressants on astrocyte physiology: implications for monoamine hypothesis of depression. J. Exp. Neurosci. 12:1179069518789149. 10.1177/1179069518789149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Marina N., Turovsky E., Christie I. N., Hosford P. S., Hadjihambi A., Korsak A., et al. (2018). Brain metabolic sensing and metabolic signaling at the level of an astrocyte. Glia 66 1185–1199. 10.1002/glia.23283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Martinac J. A., Craft D. K., Su J. H., Kim R. C., Cotman C. W. (2001). Astrocytes degenerate in frontotemporal dementia: possible relation to hypoperfusion. Neurobiol. Aging 22 195–207. 10.1016/S0197-4580(00)00231-1 [DOI] [PubMed] [Google Scholar]
  65. Martinez-Vicente M., Talloczy Z., Wong E., Tang G., Koga H., Kaushik S., et al. (2010). Cargo recognition failure is responsible for inefficient autophagy in Huntington’s disease. Nat. Neurosci. 13 567–576. 10.1038/nn.2528 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Masliah E., Alford M., Mallory M., Hansen L. (1996). Deficient glutamate transport is associated with neurodegeneration in Alzheimer’s disease. Ann. Neurol. 40 759–766. 10.1002/ana.410400512 [DOI] [PubMed] [Google Scholar]
  67. Masters C. L., Bateman R., Blennow K., Rowe C. C., Sperling R. A., Cummings J. L. (2015). Alzheimer’s disease. Nat. Rev. Dis. Primers 1:15056. 10.1038/nrdp.2015.56 [DOI] [PubMed] [Google Scholar]
  68. Merlini M., Meyer E. P., Ulmann-Schuler A., Nitsch R. M. (2011). Vascular β-amyloid and early astrocyte alterations impair cerebrovascular function and cerebral metabolism in transgenic arcAβ mice. Acta Neuropathol. 12 293–311. 10.1007/s00401-011-0834-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Meunier C., Wang N., Yi C., Dallerac G., Ezan P., Koulakoff A., et al. (2017). Contribution of astroglial Cx43 hemichannels to the modulation of glutamatergic currents by D-serine in the mouse prefrontal cortex. J. Neurosci. 37 9064–9075. 10.1523/JNEUROSCI.2204-16.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Miguel-Hidalgo J. J., Baucom C., Dilley G., Overholser J. C., Meltzer H. Y., Stockmeier C. A., et al. (2000). Glial fibrillary acidic protein immunoreactivity in the prefrontal cortex distinguishes younger from older adults in major depressive disorder. Biol. Psychiatry 48 861–873. 10.1016/S0006-3223(00)00999-9 [DOI] [PubMed] [Google Scholar]
  71. Molofsky A. V., Deneen B. (2015). Astrocyte development: a guide for the perplexed. Glia 63 1320–1329. 10.1002/glia.22836 [DOI] [PubMed] [Google Scholar]
  72. Morel L., Chiang M. S. R., Higashimori H., Shoneye T., Iyer L. K., Yelick J., et al. (2017). Molecular and functional properties of regional astrocytes in the adult brain. J. Neurosci. 37 8706–8717. 10.1523/JNEUROSCI.3956-16.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Mosconi L., Sorbi S., de Leon M. J., Li Y., Nacmias B., Myoung P. S., et al. (2006). Hypometabolism exceeds atrophy in presymptomatic early-onset familial Alzheimer’s disease. J. Nucl. Med. 47 1778–1786. [PubMed] [Google Scholar]
  74. Nagata E., Sawa A., Ross C. A., Snyder S. H. (2004). Autophagosome-like vacuole formation in Huntington’s disease lymphoblasts. Neuroreport 15 1325–1328. 10.1097/01.wnr.0000127073.66692.8f [DOI] [PubMed] [Google Scholar]
  75. Narita M., Kuzumaki N., Narita M., Kaneko C., Tamai E., Khotib J., et al. (2006). Age-related emotionality is associated with cortical delta-opioid receptor dysfunction-dependent astrogliosis. Neuroscience 137 1359–1367. 10.1016/j.neuroscience.2005.10.067 [DOI] [PubMed] [Google Scholar]
  76. Neus Bosch M., Pugliese M., Andrade C., Gimeno-Bayon J., Mahy N., Rodriguez M. J. (2015). Amyloid-beta immunotherapy reduces amyloid plaques and astroglial reaction in aged domestic dogs. Neurodegener. Dis. 15 24–37. 10.1159/000368672 [DOI] [PubMed] [Google Scholar]
  77. Oberheim N. A., Takano T., Han X., He W., Lin J. H. C., Wang F., et al. (2009). Uniquely hominid features of adult human astrocytes. J. Neurosci. 29 3276–3287. 10.1523/JNEUROSCI.4707-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Oksanen M., Petersen A. J., Naumenko N., Puttonen K., Lehtonen Š., Gubert Olivé M., et al. (2017). PSEN1 mutant iPSC-derived model reveals severe astrocyte pathology in Alzheimer’s disease. Stem Cell Rep. 9 1885–1897. 10.1016/j.stemcr.2017.10.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Olabarria M., Noristani H. N., Verkhratsky A., Rodríguez J. J. (2010). Concomitant astroglial atrophy and astrogliosis in a triple transgenic animal model of Alzheimer’s disease. Glia 58 831–838. 10.1002/glia.20967 [DOI] [PubMed] [Google Scholar]
  80. Parsons M. P., Vanni M. P., Woodard C. L., Kang R., Murphy T. H., Raymond L. A. (2016). Real-time imaging of glutamate clearance reveals normal striatal uptake in Huntington disease mouse models. Nat. Commun. 7:11251. 10.1038/ncomms11251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Phatnani H., Maniatis T. (2015). Astrocytes in neurodegenerative disease. Cold Spring Harb. Perspect. Biol. 7:a020628. 10.1101/cshperspect.a020628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Plotkin J. L., Joshua L., Day M., Peterson Jayms D., Xie Z., Kress Geraldine J.et al. (2014). Impaired TrkB receptor signaling underlies corticostriatal dysfunction in Huntington’s disease. Neuron 83 178–188. 10.1016/j.neuron.2014.05.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Prevot V., Dehouck B., Sharif A., Ciofi P., Giacobini P., Clasadonte J. (2018). The versatile tanycyte: a hypothalamic integrator of reproduction and energy metabolism. Endocr. Rev. 39 333–368. 10.1210/er.2017-00235 [DOI] [PubMed] [Google Scholar]
  84. Rajkowska G., Miguel-Hidalgo J. J., Makkos Z., Meltzer H., Overholser J., Stockmeier C. (2002). Layer-specific reductions in GFAP-reactive astroglia in the dorsolateral prefrontal cortex in schizophrenia. Schizophr. Res. 57 127–138. 10.1016/S0920-9964(02)00339-0 [DOI] [PubMed] [Google Scholar]
  85. Reick C., Ellrichmann G., Tsai T., Lee D. H., Wiese S., Gold R., et al. (2016). Expression of brain-derived neurotrophic factor in astrocytes - Beneficial effects of glatiramer acetate in the R6/2 and YAC128 mouse models of Huntington’s disease. Exp. Neurol. 285 12–23. 10.1016/j.expneurol.2016.08.012 [DOI] [PubMed] [Google Scholar]
  86. Reiman E. M., Chen K., Alexander G. E., Caselli R. J., Bandy D., Osborne D., et al. (2004). Functional brain abnormalities in young adults at genetic risk for late-onset Alzheimer’s dementia. Proc. Natl. Acad. Sci. U.S.A. 101 284–289. 10.1073/pnas.2635903100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Roberts R. C., Roche J. K., McCullumsmith R. E. (2014). Localization of excitatory amino acid transporters EAAT1 and EAAT2 in human postmortem cortex: a light and electron microscopic study. Neuroscience 277 522–540. 10.1016/j.neuroscience.2014.07.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Rodrieguez-Veitez E., Ni R., Gulyás B., Tóth M., Häggkvist J., Halldin C., et al. (2015). Astrocytosis precedes amyloid plaque deposition in Alzheimer APPswe transgenic mouse brain: a correlative positron emission tomography and in vitro imaging study. Eur. J. Nucl. Med. Mol. Imaging 42 1119–1132. 10.1007/s00259-015-3047-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Semyanov A. (2019). Spatiotemporal pattern of calcium activity in astrocytic network. Cell Calcium 78 15–25. 10.1016/j.ceca.2018.12.007 [DOI] [PubMed] [Google Scholar]
  90. Serio A., Bilican B., Barmada S. J., Ando D. M., Zhao C., Siller R., et al. (2013). Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proc. Natl. Acad. Sci. U.S.A. 110 4697–4702. 10.1073/pnas.1300398110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Sherwood C. C., Stimpson C. D., Raghanti M. A., Wildman D. E., Uddin M., Grossman L. I., et al. (2006). Evolution of increased glia–neuron ratios in the human frontal cortex. Proc. Natl. Acad. Sci. U.S.A. 103 13606–13611. 10.1073/pnas.0605843103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Shi Y., Yamada K., Liddelow S. A., Smith S. T., Zhao L., Luo W., et al. (2017). ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549 523–527. 10.1038/nature24016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Shin J. Y., Fang Z. H., Yu Z. X., Wang C. E., Li S. H., Li X. J. (2005). Expression of mutant huntingtin in glial cells contributes to neuronal excitotoxicity. J. Cell Biol. 171 1001–1012. 10.1083/jcb.200508072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Sjostedt E., Fagerberg L., Hallstrom B. M., Haggmark A., Mitsios N., Nilsson P., et al. (2015). Defining the human brain proteome using transcriptomics and antibody-based profiling with a focus on the cerebral cortex. PLoS One 10:e0130028. 10.1371/journal.pone.0130028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Stroth N., Svenningsson P. (2015). S100B interacts with the serotonin 5-HT7 receptor to regulate a depressive-like behavior. Eur. Neuropsychopharmacol. 25 2372–2380. 10.1016/j.euroneuro.2015.10.003 [DOI] [PubMed] [Google Scholar]
  96. Su J. H., Nichol K. E., Sitch T., Sheu P., Chubb C., Miller B. L., et al. (2000). DNA damage and activated caspase-3 expression in neurons and astrocytes: evidence for apoptosis in frontotemporal dementia. Exp. Neurol. 163 9–19. 10.1006/exnr.2000.7340 [DOI] [PubMed] [Google Scholar]
  97. Takahashi K., Tanabe K., Ohnuki M., Narita M., Ichisaka T., Tomoda K., et al. (2007). Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131 861–872. 10.1016/j.cell.2007.11.019 [DOI] [PubMed] [Google Scholar]
  98. Takouda J., Katada S., Nakashima K. (2017). Emerging mechanisms underlying astrogenesis in the developing mammalian brain. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 93 386–398. 10.2183/pjab.93.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Talantova M., Sanz-Blasco S., Zhang X., Xia P., Akhtar M. W., Okamoto S.-I., et al. (2013). Aβ induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proc. Natl. Acad. Sci. U.S.A. 110E2518–E2527. 10.1073/pnas.1306832110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Tanahashi S., Yamamura S., Nakagawa M., Motomura E., Okada M. (2012). Clozapine, but not haloperidol, enhances glial D-serine and L-glutamate release in rat frontal cortex and primary cultured astrocytes. Br. J. Pharmacol. 165 1543–1555. 10.1111/j.1476-5381.2011.01638.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Teneka M. T., Sastre M., Dumitrescu-Ozimek L., Dewachter I., Walter J., Klockgether T., et al. (2005). Focal glial activation coincides with increased BACE1 activation and precedes amyloid plaque deposition in APP[V717I] transgenic mice. J. Neuroinflammation 2:22. 10.1186/1742-2094-2-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Tian G., Kong Q., Lai L., Ray-Chaudhury A., Lin C.-L. (2010). 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. 113 978–989. 10.1111/j.1471-4159.2010.06661.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Tong J., Huang C., Bi F., Wu Q., Huang B., Liu X., et al. (2013). Expression of ALS-linked TDP-43 mutant in astrocytes causes non-cell-autonomous motor neuron death in rats. EMBO J. 32 1917–1926. 10.1038/emboj.2013.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Tong X., Ao Y., Faas G. C., Nwaobi S. E., Xu J., Haustein M. D., et al. (2014). Astrocyte Kir4.1 ion channel deficits contribute to neuronal dysfunction in Huntington’s disease model mice. Nat. Neurosci. 17 694–703. 10.1038/nn.3691 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Torres-Platas S. G., Nagy C., Wakid M., Turecki G., Mechawar N. (2016). Glial fibrillary acidic protein is differentially expressed across cortical and subcortical regions in healthy brains and downregulated in the thalamus and caudate nucleus of depressed suicides. Mol. Psychiatry 21 509–515. 10.1038/mp.2015.65 [DOI] [PubMed] [Google Scholar]
  106. Tsai H.-H., Li H., Fuentealba L. C., Molofsky A. V., Taveira-Marques R., Zhuang H., et al. (2012). Regional astrocyte allocation regulates CNS synaptogenesis and repair. Science 337 358–362. 10.1126/science.1222381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Turner R. C., Lucke-Wold B. P., Robson M. J., Lee J. M., Bailes J. E. (2016). Alzheimer’s disease and chronic traumatic encephalopathy: distinct but possibly overlapping disease entities. Brain Inj. 30 1279–1292. 10.1080/02699052.2016.1193631 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Vasile F., Dossi E., Rouach N. (2017). Human astrocytes: structure and functions in the healthy brain. Brain Struct. Funct. 222 2017–2029. 10.1007/s00429-017-1383-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Wallingford J., Scott A. L., Rodrigues K., Doering L. C. (2017). Altered developmental expression of the astrocyte-secreted factors hevin and SPARC in the fragile X mouse model. Front. Mol. Neurosci. 10:268. 10.3389/fnmol.2017.00268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Wang C., Wilson W. A., Moore S. D., Mace B. E., Maeda N., Schmechel D. E., et al. (2005). Human apoE4-targeted replacement mice display synaptic deficits in the absence of neuropathology. Neurobiol. Dis. 18 390–398. 10.1016/j.nbd.2004.10.013 [DOI] [PubMed] [Google Scholar]
  111. Xin W., Bonci A. (2018). Functional astrocyte heterogeneity and implications for their role in shaping neurotransmission. Front. Cell. Neurosci. 12:141. 10.3389/fncel.2018.00141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Xiu J., Nordberg A., Zhang J. T., Guan Z. Z. (2005). Expression of nicotinic receptors on primary cultures of rat astrocytes and up-regulation of the alpha7, alpha4 and beta2 subunits in response to nanomolar concentrations of the beta-amyloid peptide(1-42). Neurochem. Int. 47 281–290. 10.1016/j.neuint.2005.04.023 [DOI] [PubMed] [Google Scholar]
  113. Zhang X., Wan J. Q., Tong X. P. (2018). Potassium channel dysfunction in neurons and astrocytes in Huntington’s disease. CNS Neurosci. Ther. 24 311–318. 10.1111/cns.12804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Zhang Y., Sloan S. A., Clarke L. E., Caneda C., Plaza C. A., Blumenthal P. D., et al. (2016). Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89 37–53. 10.1016/j.neuron.2015.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Zhao J., Davis M. D., Martens Y. A., Shinohara M., Graff-Radford N. R., Younkin S. G., et al. (2017). APOE 𝜀4/𝜀4 diminishes neurotrophic function of human iPSC-derived astrocytes. Hum. Mol. Genet. 26 2690–2700. 10.1093/hmg/ddx155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Zhao T., Hong Y., Yin P., Li S., Li X.-J. (2017). Differential HspBP1 expression accounts for the greater vulnerability of neurons than astrocytes to misfolded proteins. Proc. Natl. Acad. Sci. 114 E7803–E7811. 10.1073/pnas.1710549114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Zhao T., Hong Y., Li S., Li X.-J. (2016). Compartment-dependent degradation of mutant huntingtin accounts for its preferential accumulation in neuronal processes. J. Neurosci. 36 8317–8328. 10.1523/JNEUROSCI.0806-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Zhao Z., Nelson A. R., Betsholtz C., Zlokovic B. V. (2015). Establishment and dysfunction of the blood-brain barrier. Cell 163 1064–1078. 10.1016/j.cell.2015.10.067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Zheng W., Li Q., Zhao C., Da Y., Zhang H.-L., Chen Z. (2018). Differentiation of glial cells from hiPSCs: potential applications in neurological diseases and cell replacement therapy. Front. Cell. Neurosci. 12:239. 10.3389/fncel.2018.00239 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Frontiers in Cellular Neuroscience are provided here courtesy of Frontiers Media SA

RESOURCES