Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: Ageing Res Rev. 2020 Feb 24;59:101039. doi: 10.1016/j.arr.2020.101039

Astrocyte Mitochondria: Central players and potential therapeutic targets for neurodegenerative diseases and injury

JL Gollihue 1, CM Norris 1
PMCID: PMC7422487  NIHMSID: NIHMS1616910  PMID: 32105849

Abstract

Mitochondrial function has long been the focus of many therapeutic strategies for ameliorating age-related neurodegeneration and cognitive decline. Historically, the role of mitochondria in non-neuronal cell types has been overshadowed by neuronal mitochondria, which are responsible for the bulk of oxidative metabolism in the brain. Despite this neuronal bias, mitochondrial function in glial cells, particularly astrocytes, is increasingly recognized to play crucial roles in overall brain metabolism, synaptic transmission, and neuronal protection. Changes in astrocytic mitochondrial function appear to be intimately linked to astrocyte activation/reactivity found in most all age-related neurodegenerative diseases. Here, we address the importance of mitochondrial function to astrocyte signaling and consider how mitochondria could contribute to both the detrimental and protective properties of activated astrocytes. Strategies for protecting astrocytic mitochondrial function, promoting bidirectional transfer of mitochondria between astrocytes and neurons, and transplanting healthy mitochondria to diseased nervous tissue are also discussed.

Keywords: Mitochondria, astrocytes, aging, neurodegeneration, mitocentric therapy

1.1. Introduction

As the body grows older and slowly moves further away from physiologic homeostasis, many changes in all of the bodily systems occur. Arguably, one of the most devastating is the changes that occur in the central nervous system- leading to loss of cognitive, motor, and emotional function that in essence make us who we are. Dementia is one such disease that progresses steadily with age, and to date there are not many effective treatments to stave off the changes that occur. Many studies have focused on salvaging neuronal function, but there are many cell types in the central nervous system that directly interact with neurons and can be a target for therapeutics, such as astrocytes.

1.2. Astrocytes

Astrocytes are the predominant glial cells found in all areas of the brain, providing uniform, non-overlapping coverage (Bushong et al., 2002) in a network of astrocytes connected by gap junctions. Pertinent to synaptic health, astrocytes guide the development and directionality of synapses (Powell and Geller, 1999), regulate transmission efficiency at pre and post-synaptic sites (Ullian et al., 2001), and modulate synapse formation and turnover (Allen et al., 2012; Bialas and Stevens, 2013; Blanco-Suarez et al., 2018; Christopherson et al., 2005; Diniz et al., 2012; Fossati et al., 2019; Gomez-Casati et al., 2010; Kucukdereli et al., 2011; Mauch et al., 2001; Vainchtein et al., 2018). Astrocytic endfeet enwrap nearly the entire cerebrovasculature creating a physical barrier known as the blood brain barrier (BBB) (Abbott et al., 2006) allowing astrocytes to shuttle metabolites from the blood to other neural cells, exchange water molecules between blood vessels and the brain parenchyma, take up potassium and glutamate to protect neurons from excitotoxicity (Nielsen et al., 1997; Price et al., 2002; Simard et al., 2003) and link elevations in neuronal activity to elevations in blood flow through a process called neurovascular coupling (NVC) (Gordon et al., 2007; Koehler et al., 2009).

It’s well established that astrocytes use sophisticated Ca2+ signaling networks and mechanisms to communicate directly with neurons and other astrocytes (for a more in-depth review see (Rossi, 2015; Verkhratsky, 2019) and (Rusakov, 2015)). Activation of a variety of receptors on the astrocyte cell membrane trigger Ca2+ release from internal stores, which can spread to neighboring astrocytes through gap junctions, creating Ca2+ waves across large interconnected astrocyte networks (Charles et al., 1991). Ca2+ waves are most prevalent in astrocytic processes (Grosche et al., 1999) creating heterogenous microdomains within the astrocyte (Panatier et al., 2011{Bindocci, 2017 #717)}. These waves can be generated spontaneously without neuronal input (Nett et al., 2002{Parri, 2001 #60)} or initiated by neurotransmitters, thus allowing neurons to influence Ca2+ levels in astrocytes and play an important role in synchronizing astrocytic Ca2+ transients (Aguado et al., 2002). Once initiated in astrocytes, Ca2+ signals may have a major impact on the physiology of nearby neurons and other cellular constituents of the neurovascular unit. For instance, astrocytic Ca2+ transients can be shaped by the mitochondrial sodium/Ca2+ exchanger, altering exocytotic release of a variety of gliotransmitters including glutamate, ATP, and D-serine, at the synapse (Parnis et al., 2013). These gliotransmitters can act upon neuronal receptors, providing a route for direct astrocyte-neuron communication (for review see (Araque et al., 2014)).

1.3. Astrocyte Activation

Astrocytes undergo many morphologic/biological changes during aging, disease, and injury, collectively referred to as “astrocyte activation” or “astrocyte reactivity”. Reactive astrocytes are often morphologically distinct from “resting” astrocytes and are usually characterized as more ramified, larger, with elongated processes (Burda et al., 2016; Wilhelmsson et al., 2006). Astrocyte activation is complex, with ongoing debates over the merits and detriments of astrocytic activation. Detrimental astrocytes appear to exhibit increased ROS and Ca2+ dysregulation (Ishii et al., 2017), release numerous cytokines, chemokines, and ROS involved in chronic neuroinflammation (Choi et al., 2014; Clarke et al., 2018; Farina et al., 2007; Zhu et al., 2009), impair glutamate uptake leading to excitotoxicity and neuronal degeneration (Dossi et al., 2018; Schousboe and Waagepetersen, 2005; Sompol et al., 2017), create abnormal synapses resulting in epilepsy and neuropathic pain (Boroujerdi et al., 2008), or cause the loss of synapses leading to impaired synaptic plasticity and cognitive decline (Furman et al., 2012; Furman et al., 2016; Hong et al., 2016; Shi et al., 2017; Stevens et al., 2007). In contrast, beneficial astrocytes release an array of factors that aid in synaptic development, repair, and rewiring including thrombospondin, matricellular proteins (SPARC-1 and hevin), TGF-beta, IL-33, D-serine, glypicans, BDNF, pentraxin 3, Chrd11, C3 and cholesterol to name just a few (Allen et al., 2012; Bialas and Stevens, 2013; Blanco-Suarez et al., 2018; Christopherson et al., 2005; Diniz et al., 2012; Fossati et al., 2019; Gomez-Casati et al., 2010; Kucukdereli et al., 2011; Mauch et al., 2001; Schafer et al., 2012; Vainchtein et al., 2018). Beneficial astrocytes also assist microglia in clearing dead cells, cellular debris, and/or protein aggregates (Gomez-Arboledas et al., 2018; Wakida et al., 2018; Wyss-Coray et al., 2003), and may assume greater glycogen storage capacity resulting in better memory formation and neuronal survival (Matsui et al., 2017; Waitt et al., 2017). Finally, astrocyte-containing glial scars, formed around injured tissue and protein aggregates, have long been thought to provide a physical barrier for shielding healthy tissue from damaging factors (Faulkner et al., 2004; Okada et al., 2006; Wang et al., 2018), though deleterious properties of glial scars have also been described (Liddelow and Barres, 2015; Liddelow and Barres, 2017; Perez-Nievas and Serrano-Pozo, 2018; Sofroniew and Vinters, 2010).

1.4. Mitochondrial Function in Astrocytes

Despite their heavy reliance on glycolysis for energy production (Hamberger and Hyden, 1963; Pellerin and Magistretti, 1994; Zhang et al., 2014), astrocytes are responsible for 20% of the brain’s total oxygen consumption (Bluml et al., 2002), most of which occurs during oxidative phosphorylation within astrocytic mitochondria en route to the production of adenosine triphosphate (ATP). In addition to oxidative metabolism, mitochondria also serve as storage organelles for Ca2+ and play an important role in intracellular Ca2+ sequestration and signaling. Elevations in cytosolic Ca2+ appear to promote the immobilization of mitochondria near highly active synapses (Stephen et al., 2015) and fine processes where Ca2+ transients occur most frequently (Bindocci et al., 2017). Mitochondria are strategically located in these microdomains to meet higher local energy demands (Jackson and Robinson, 2018), shape Ca2+ fluctuations (Agarwal et al., 2017; Gunter et al., 2004), and/or modulate Ca2+ -dependent processes such as gliotransmission (Parnis et al., 2013; Stephen et al., 2015). Regardless of the exact function, mitochondria loss in these microdomains is associated with neurodegeneration, suggesting an essential role in astrocyte-neuron synergy. Additionally, it was shown that a decrease in healthy mitochondria in astrocytic processes correlated to neuronal loss after ischemia, even though the number of astrocytes remained stable, indicating that the loss of mitochondria at the astrocyte-synapse junctions may play a part in neuronal loss (Ito et al., 2009). Mitochondria can also be found in cerebral blood-vessel associated astrocyte endfeet, which are also highly metabolically active and exhibit dynamic Ca2+ signaling (Kacem et al., 1998; Stobart et al., 2018). However, very little is known about the role of the mitochondria in these specialized microdomains.

2.1. Discussion

2.2. Dysfunction of astrocytic mitochondria can cause deleterious actions on neurons

As in other cell types, astrocytic mitochondria are vulnerable to aging, injury and disease. In fact, many of the same stimuli that trigger robust astrocyte activation (for example, ischemia, traumatic tissue injury, inflammation, cytokines) also lead to astrocytic mitochondrial dysfunction (Figure 1), the telltale signs of which include calcium dysregulation, MPTP, the excessive generation of reactive oxygen species (ROS) and cell death cascades. Due to the intimate relationships between astrocytes, these factors can possibly have far-reaching effects across the astrocyte network, causing further astrocyte activation and exacerbating the astrocyte activation/ mitochondria dysfunction loop. Oxidative stress in astrocytes, triggered by pathogenic factors such as amyloid-beta, has been shown to inflict extensive damage to surrounding neurons (Abramov et al., 2004). Neurotoxicity initiated by astrocytic mitochondrial dysfunction may arise from the release of a number of proinflammatory cytokines, ROS, and other inflammatory factors (Kubik and Philbert, 2015; Min et al., 2015; Nahirnyj et al., 2013).

Figure 1. Factors that activate astrocytes also influence mitochondrial health and function.

Figure 1.

Many of the pathogenic factors (either by loss of tissue homeostasis or release of molecular signals) that are responsible for activating astrocytes can also trigger mitochondrial dysfunction. Activation of astrocytes increases the activity burden placed upon their mitochondria, which may in turn cause dysfunction and the release of an array of factors within the cell. These factors can themselves cause activation or damage to the astrocyte. Due to the nature of this bidirectional loop, it is difficult to assess whether the pathogenic factors cause astrocyte activation, which subsequently causes mitochondrial dysfunction, or vice versa.

Breakdown of the astrocytic mitochondrial membrane potential by adding fluorocitrate to primary astrocytes caused a delayed dissipation of the mitochondria membrane potential leading to reduced glutamate uptake (Voloboueva et al., 2007). Consequently, neurons co-cultured with fluorocitrate-treated astrocytes exhibited increased vulnerability to glutamate-mediated excitotoxic death. It is interesting to note that the deleterious effects of fluorocitrate on astrocytes was hastened in the presence of co-cultured neurons, suggesting that neurons actively tax the energy output of astrocytic mitochondria. Similarly, it was shown that there was increased neuronal death following ischemic injury when specifically targeting astrocytic electron transport chain for damage in vivo (Fiebig et al., 2019). These results infer that functional astrocytic mitochondria are necessary for the support functions that astrocytes provide to neurons following injury. If mitochondrial dysfunction in astrocytes is harmful to neurons, it is logical to assume that treating astrocytic mitochondria can then be beneficial to neurons.

Similar to neurons, activated astrocytes exhibit extensive Ca2+ dysregulation during aging, injury, and disease characterized by elevated cytosolic Ca2+ levels, augmented Ca2+ transients, and/or more frequent Ca2+ oscillations (Agulhon et al., 2008; Kuchibhotla et al., 2009). Excess accumulation of Ca2+ into the mitochondria, which often occurs when other Ca2+ regulatory mechanisms are challenged, depletes the mitochondrial membrane potential, leading to impaired ATP production, and, in extreme cases, generation of the mitochondrial permeability transition pore (MPTP) (Hunter et al., 1976) resulting in release of Ca2+, cytochrome c, ROS and other cytotoxic factors into the cytoplasm (Krajewski et al., 1999). Severe Ca2+ dysregulation in astrocytes likely leads to aberrant activation of Ca2+ -dependent enzymes, such as calpain proteases and calcineurin phosphatases (Feng et al., 2011; Norris et al., 2005; Pleiss et al., 2016; Price et al., 2018; Shields et al., 2000; Shields et al., 1998), which in turn can exacerbate mitochondrial dysfunction. Consequently, inhibition of calcineurin signaling in astrocytes under a variety of injurious conditions helps maintain protective properties of astrocytes (e.g. glutamate uptake) and reduces excitotoxic damage to neurons (Furman et al., 2012; Sompol et al., 2017), perhaps through the dampening of vicious feedback cycles between oxidative stress and Ca2+ dysregulation.

2.3. Bidirectional exchange of mitochondria between astrocytes and neurons

Mitochondria may also be central to many of the neuroprotective functions associated with astrocyte activation. Presumably, the release of growth factors, establishment of protective glial scars, and increased synaptic support all depend critically on the energy output of astrocytic mitochondria. Recent work has established that astrocytes can’t mount a protective proliferative response following acute brain lesions if mitochondrial function is impaired (Fiebig et al., 2019). Astrocytes, like many other cell types, dispose of their own damaged mitochondria via autophagy (mitophagy) which is elevated following acute injury (Quintana et al., 2019), perhaps to minimize the deleterious consequences of oxidative stress and Ca2+ dysregulation discussed above. Not only do astrocytes actively degrade their own damaged mitochondria, they can also take up and dispose of mitochondria released from other cells. This form of “transmitophagy” was first reported by Davis et al (2014), who showed that fluorescently tagged mitochondria from otherwise healthy retinal ganglion cells were present (in high numbers) in astrocytic lysosomes. It’s unclear whether astrocytes take a leading role in disposing of neuron-derived mitochondria in the context of neurodegenerative disease or following acute brain injury.

Interestingly, the intercellular transport of mitochondria in astrocyte-neuron networks appears to work in both directions. But, while neurons tend to hand off damaged mitochondria to astrocytes for degradation, it seems that activated astrocytes can donate some of their healthy mitochondria to nearby damaged neurons, which in turn, leads to greater neuronal viability. Release of mitochondria-containing vesicles from primary astrocytes was found to depend on a Ca2+ -CD38-cADPR signaling pathway (Hayakawa et al., 2016). Upregulation of astrocytic CD38 using a CRISPR/Cas9 system significantly augmented the release of mitochondria-containing vesicles, that, when added to oxygen-glucose starved neurons, restored neuronal energy production. Hayakawa et al subsequently validated these observations in intact mice by showing that fluorescently tagged astrocytic mitochondria are passed to neurons after a transient ischemic insult. Astrocyte-derived mitochondria appeared to fuse with neuronal mitochondria in peri-infarct neocortex and were associated with the upregulation of cell survival pathways. Moreover, blockade of CD38 in infarcted mice had a negative impact on a number of functional outcome measures, suggesting that astrocyte-mediated mitochondria release occurs naturally in the brain as long as CD38 signaling is maintained. It is presently unknown whether a similar neuroprotective mechanism is used in other, more chronic neurodegenerative conditions.

2.4. Mito-Centric Therapies

Mitochondrial-targeting strategies have long been investigated for their potential to treat brain injury and neurodegenerative disease. Administration of antioxidants, mild uncouplers, MPTP inhibitors, and alternative biofuels are among the most commonly used strategies. In the fields of neurotrauma and neurodegeneration, these treatments are typically aimed at providing neuroprotection through the improvement of neuronal mitochondrial function. However, as outlined in this review, astrocytic mitochondrial health may be just as important to overall neuronal survival and adaptation to injury/disease. By targeting the main support cells of the brain (i.e. astrocytes) many other cell types besides neurons may reap substantial benefits. For research (and possibly clinical) purposes, astrocytes can be selectively targeted with recombinant viral vectors, nanoparticles, or specialized peptides (Figure 2) (Bosson et al., 2017; Cannon et al., 2011; Furman et al., 2012; Terashima et al., 2018; Valiante et al., 2015). Valori et al. (2019) published an excellent review regarding the possible hurdles to translation of astrocyte-centered therapy as well as current approaches (Valori et al., 2019). Evidence for imparting neuroprotection through the selective targeting of astrocyte signaling pathways has already been established in several different disease models including acute cortical lesions (Wilhelmsson et al., 2004), β-amyloid expression (Ceyzeriat et al., 2018; Fernandez et al., 2016; Furman et al., 2012; Sompol et al., 2017), spinal cord injury (Gaudet and Fonken, 2018), chronic pain (Li et al., 2019), and CNS injury (Furman et al., 2016; Huang et al., 2019). Astrocyte-directed therapeutics to protect and/or maintain healthy mitochondrial function could include a variety of antioxidants, MPTP modulators, and alternative biofuels utilized in oxidative phosphorylation. Alternatively, strategies could focus on the release of healthy mitochondria from astrocytes through the stimulation of CD38-cADPR signaling.

Figure 2. Possible mitochondria-centric astrocyte therapies.

Figure 2.

Healthy mitochondrial function is key to proper astrocyte function. Targeting mitochondria to increase astrocytic vitality and thereby support neuronal and vascular health is becoming a popular potential therapeutic. Multiple avenues to target mitochondrial health are available, including delivery of drugs specifically to the mitochondria via AAVs, peptides, and nanoparticles. Supplementing the astrocyte with healthy mitochondria, or conversely promoting the degradation of unhealthy mitochondria, are also viable potential therapies.

Endogenous transcellular mitochondrial donation has also inspired a variety of exogenous mitochondrial delivery strategies, whereby healthy mitochondria are harvested from an allogeneic or, preferably an autologous source, and then transplanted into damaged or diseased tissue. Mitochondrial transplantation has already shown considerable promise for limiting ischemic damage to the heart, especially in pediatric patients with coronary artery disease (Emani and McCully, 2018) where it is being intensively investigated in clinical trials. While there are presently no similar clinical trials underway for CNS disorders, mitochondrial transplantation has shown significant neuroprotective and/or functional benefits in diverse experimental models (Chang et al., 2016; Cowan et al., 2016; Gollihue et al., 2018; Gollihue et al., 2017; Hayakawa et al., 2016; McCully et al., 2009). When injected directly into gliomas of living mice, isolated mitochondria led to a metabolic switch from glycolysis to aerobic respiration coincident with decreased glioma growth (Sun C, 2019). In a mouse model of stroke, transplantation of mitochondria to the infarct region resulted in neuronal uptake of mitochondria. Similarly, after stroke in these mice there is evidence of mitochondrial transfer from astrocytes to neurons resulting in increased pro-survival signaling, reduced neuronal death, and ameliorated motor and neurologic deficits (Hayakawa et al., 2016). Using a modified transplant procedure, supplemented with the peptide carrier protein, PEP1, mitochondrial delivery to the medial forebrain bundle of a Parkinson’s model rats led to greater neuronal sparing in the substantia nigra and improved locomotive functional outcome (Chang et al., 2013). More recently, we found that transplant of viable mitochondria to injured rat spinal cord helped to maintain acute bioenergetics, though no long term benefits were observed for tissue sparing or the recovery of essential sensory-motor functions (Gollihue, Patel et al. 2018). Together, these studies show that exogenous mitochondrial incorporation into neurons is possible in vivo, and provide proof-of-principle that transplants can result in measurable improvements for many different physiological outcomes.

3.1. Summary and Conclusions

Astrocytes are highly metabolically active cells in the central nervous system that have a variety of functions- one of the most important being neuronal support. Mitochondria produce energy to meet the energy demands of astrocytes. Astrocytic mitochondrial dysfunction is incidentally highly detrimental, leading to insufficient energy production, Ca2+ dysregulation, inflammatory response induction, and glutamate dysregulation. Treating mitochondria in astrocytes can directly benefit neuronal survival because of the interdependence of neurons and astrocytes. Additionally, astrocytes have an inherent ability to donate their mitochondria to neurons in need- a process that could be exploited for neuroprotection in aging, injury, and disease. Supplying exogenous healthy mitochondria to injured or susceptible tissue could have far-reaching effects in neuroprotection, though there does not seem to be a one size-fits-all technique that is applicable in all instances of CNS injury. Further research to optimize disease-specific techniques for targeting astrocytic mitochondrial health are needed.

Acknowledgments

Funding: This work was supported by grants from the National Institutes of Health [T32AG057461 to JLG and RF1AG027297 to CMN]; the Hazel Embry Research Trust; and the Sylvia Mansbach Endowment.

References:

  1. Abbott NJ, Ronnback L, Hansson E, 2006. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci 7, 41–53. [DOI] [PubMed] [Google Scholar]
  2. Abramov AY, Canevari L, Duchen MR, 2004. Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase. J Neurosci 24, 565–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Agarwal A, Wu PH, Hughes EG, Fukaya M, Tischfield MA, Langseth AJ, Wirtz D, Bergles DE, 2017. Transient Opening of the Mitochondrial Permeability Transition Pore Induces Microdomain Calcium Transients in Astrocyte Processes. Neuron 93, 587–605 e587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Aguado F, Espinosa-Parrilla JF, Carmona MA, Soriano E, 2002. Neuronal activity regulates correlated network properties of spontaneous calcium transients in astrocytes in situ. J Neurosci 22, 9430–9444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Agulhon C, Petravicz J, McMullen AB, Sweger EJ, Minton SK, Taves SR, Casper KB, Fiacco TA, McCarthy KD, 2008. What is the role of astrocyte calcium in neurophysiology? Neuron 59, 932–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Allen NJ, Bennett ML, Foo LC, Wang GX, Chakraborty C, Smith SJ, Barres BA, 2012. Astrocyte glypicans 4 and 6 promote formation of excitatory synapses via GluA1 AMPA receptors. Nature 486, 410–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Araque A, Carmignoto G, Haydon PG, Oliet SH, Robitaille R, Volterra A, 2014. Gliotransmitters travel in time and space. Neuron 81, 728–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bialas AR, Stevens B, 2013. TGF-beta signaling regulates neuronal C1q expression and developmental synaptic refinement. Nat Neurosci 16, 1773–1782. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  9. Bindocci E, Savtchouk I, Liaudet N, Becker D, Carriero G, Volterra A, 2017. Three-dimensional Ca(2+) imaging advances understanding of astrocyte biology. Science 356. [DOI] [PubMed] [Google Scholar]
  10. Blanco-Suarez E, Liu TF, Kopelevich A, Allen NJ, 2018. Astrocyte-Secreted Chordin-like 1 Drives Synapse Maturation and Limits Plasticity by Increasing Synaptic GluA2 AMPA Receptors. Neuron 100, 1116–1132 e1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bluml S, Moreno-Torres A, Shic F, Nguy CH, Ross BD, 2002. Tricarboxylic acid cycle of glia in the in vivo human brain. NMR Biomed 15, 1–5. [DOI] [PubMed] [Google Scholar]
  12. Boroujerdi A, Kim HK, Lyu YS, Kim DS, Figueroa KW, Chung JM, Luo ZD, 2008. Injury discharges regulate calcium channel alpha-2-delta-1 subunit upregulation in the dorsal horn that contributes to initiation of neuropathic pain. Pain 139, 358–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bosson A, Paumier A, Boisseau S, Jacquier-Sarlin M, Buisson A, Albrieux M, 2017. TRPA1 channels promote astrocytic Ca(2+) hyperactivity and synaptic dysfunction mediated by oligomeric forms of amyloid-beta peptide. Mol Neurodegener 12, 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Burda JE, Bernstein AM, Sofroniew MV, 2016. Astrocyte roles in traumatic brain injury. Exp Neurol 275 Pt 3, 305–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bushong EA, Martone ME, Jones YZ, Ellisman MH, 2002. Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains. J Neurosci 22, 183–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cannon JR, Sew T, Montero L, Burton EA, Greenamyre JT, 2011. Pseudotype-dependent lentiviral transduction of astrocytes or neurons in the rat substantia nigra. Exp Neurol 228, 41–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Ceyzeriat K, Ben Haim L, Denizot A, Pommier D, Matos M, Guillemaud O, Palomares MA, Abjean L, Petit F, Gipchtein P, Gaillard MC, Guillermier M, Bernier S, Gaudin M, Auregan G, Josephine C, Dechamps N, Veran J, Langlais V, Cambon K, Bemelmans AP, Baijer J, Bonvento G, Dhenain M, Deleuze JF, Oliet SHR, Brouillet E, Hantraye P, Carrillo-de Sauvage MA, Olaso R, Panatier A, Escartin C, 2018. Modulation of astrocyte reactivity improves functional deficits in mouse models of Alzheimer's disease. Acta Neuropathol Commun 6, 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chang JC, Liu KH, Li YC, Kou SJ, Wei YH, Chuang CS, Hsieh M, Liu CS, 2013. Functional recovery of human cells harbouring the mitochondrial DNA mutation MERRF A8344G via peptide-mediated mitochondrial delivery. Neurosignals 21, 160–173. [DOI] [PubMed] [Google Scholar]
  19. Chang JC, Wu SL, Liu KH, Chen YH, Chuang CS, Cheng FC, Su HL, Wei YH, Kuo SJ, Liu CS, 2016. Allogeneic/xenogeneic transplantation of peptide-labeled mitochondria in Parkinson's disease: restoration of mitochondria functions and attenuation of 6-hydroxydopamine-induced neurotoxicity. Transl Res 170, 40–56 e43. [DOI] [PubMed] [Google Scholar]
  20. Charles AC, Merrill JE, Dirksen ER, Sanderson MJ, 1991. Intercellular signaling in glial cells: calcium waves and oscillations in response to mechanical stimulation and glutamate. Neuron 6, 983–992. [DOI] [PubMed] [Google Scholar]
  21. Choi SS, Lee HJ, Lim I, Satoh J, Kim SU, 2014. Human astrocytes: secretome profiles of cytokines and chemokines. PLoS One 9, e92325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Christopherson KS, Ullian EM, Stokes CC, Mullowney CE, Hell JW, Agah A, Lawler J, Mosher DF, Bornstein P, Barres BA, 2005. Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell 120, 421–433. [DOI] [PubMed] [Google Scholar]
  23. Clarke LE, Liddelow SA, Chakraborty C, Munch AE, Heiman M, Barres BA, 2018. Normal aging induces A1-like astrocyte reactivity. Proc Natl Acad Sci U S A 115, E1896–E1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Cowan DB, Yao R, Akurathi V, Snay ER, Thedsanamoorthy JK, Zurakowski D, Ericsson M, Friehs I, Wu Y, Levitsky S, Del Nido PJ, Packard AB, McCully JD, 2016. Intracoronary Delivery of Mitochondria to the Ischemic Heart for Cardioprotection. PloS one 11, e0160889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Diniz LP, Almeida JC, Tortelli V, Vargas Lopes C, Setti-Perdigao P, Stipursky J, Kahn SA, Romao LF, de Miranda J, Alves-Leon SV, de Souza JM, Castro NG, Panizzutti R, Gomes FC, 2012. Astrocyte-induced synaptogenesis is mediated by transforming growth factor beta signaling through modulation of D-serine levels in cerebral cortex neurons. J Biol Chem 287, 41432–41445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Dossi E, Vasile F, Rouach N, 2018. Human astrocytes in the diseased brain. Brain Res Bull 136, 139–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Emani SM, McCully JD, 2018. Mitochondrial transplantation: applications for pediatric patients with congenital heart disease. Transl Pediatr 7, 169–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Farina C, Aloisi F, Meinl E, 2007. Astrocytes are active players in cerebral innate immunity. Trends Immunol 28, 138–145. [DOI] [PubMed] [Google Scholar]
  29. Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV, 2004. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci 24, 2143–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Feng ZH, Hao J, Ye L, Dayao C, Yan N, Yan Y, Chu L, Shi FD, 2011. Overexpression of mu-calpain in the anterior temporal neocortex of patients with intractable epilepsy correlates with clinicopathological characteristics. Seizure 20, 395–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Fernandez AM, Hervas R, Dominguez-Fraile M, Garrido VN, Gomez-Gutierrez P, Vega M, Vitorica J, Perez JJ, Torres Aleman I, 2016. Blockade of the Interaction of Calcineurin with FOXO in Astrocytes Protects Against Amyloid-beta-Induced Neuronal Death. J Alzheimers Dis 52, 1471–1478. [DOI] [PubMed] [Google Scholar]
  32. Fiebig C, Keiner S, Ebert B, Schaffner I, Jagasia R, Lie DC, Beckervordersandforth R, 2019. Mitochondrial Dysfunction in Astrocytes Impairs the Generation of Reactive Astrocytes and Enhances Neuronal Cell Death in the Cortex Upon Photothrombotic Lesion. Front Mol Neurosci 12, 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Fossati G, Pozzi D, Canzi A, Mirabella F, Valentino S, Morini R, Ghirardini E, Filipello F, Moretti M, Gotti C, Annis DS, Mosher DF, Garlanda C, Bottazzi B, Taraboletti G, Mantovani A, Matteoli M, Menna E, 2019. Pentraxin 3 regulates synaptic function by inducing AMPA receptor clustering via ECM remodeling and beta1-integrin. EMBO J 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Furman JL, Sama DM, Gant JC, Beckett TL, Murphy MP, Bachstetter AD, Van Eldik LJ, Norris CM, 2012. Targeting astrocytes ameliorates neurologic changes in a mouse model of Alzheimer's disease. J Neurosci 32, 16129–16140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Furman JL, Sompol P, Kraner SD, Pleiss MM, Putman EJ, Dunkerson J, Mohmmad Abdul H, Roberts KN, Scheff SW, Norris CM, 2016. Blockade of Astrocytic Calcineurin/NFAT Signaling Helps to Normalize Hippocampal Synaptic Function and Plasticity in a Rat Model of Traumatic Brain Injury. J Neurosci 36, 1502–1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Gaudet AD, Fonken LK, 2018. Glial Cells Shape Pathology and Repair After Spinal Cord Injury. Neurotherapeutics 15, 554–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Gollihue JL, Patel SP, Eldahan KC, Cox DH, Donahue RR, Taylor BK, Sullivan PG, Rabchevsky AG, 2018. Effects of Mitochondrial Transplantation on Bioenergetics, Cellular Incorporation, and Functional Recovery after Spinal Cord Injury. J Neurotrauma 35, 1800–1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Gollihue JL, Patel SP, Mashburn C, Eldahan KC, Sullivan PG, Rabchevsky AG, 2017. Optimization of mitochondrial isolation techniques for intraspinal transplantation procedures. J Neurosci Methods 287, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Gomez-Arboledas A, Davila JC, Sanchez-Mejias E, Navarro V, Nunez-Diaz C, Sanchez-Varo R, Sanchez-Mico MV, Trujillo-Estrada L, Fernandez-Valenzuela JJ, Vizuete M, Comella JX, Galea E, Vitorica J, Gutierrez A, 2018. Phagocytic clearance of presynaptic dystrophies by reactive astrocytes in Alzheimer's disease. Glia 66, 637–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Gomez-Casati ME, Murtie JC, Rio C, Stankovic K, Liberman MC, Corfas G, 2010. Nonneuronal cells regulate synapse formation in the vestibular sensory epithelium via erbB-dependent BDNF expression. Proc Natl Acad Sci U S A 107, 17005–17010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Gordon GR, Mulligan SJ, MacVicar BA, 2007. Astrocyte control of the cerebrovasculature. Glia 55, 1214–1221. [DOI] [PubMed] [Google Scholar]
  42. Grosche J, Matyash V, Moller T, Verkhratsky A, Reichenbach A, Kettenmann H, 1999. Microdomains for neuron-glia interaction: parallel fiber signaling to Bergmann glial cells. Nat Neurosci 2, 139–143. [DOI] [PubMed] [Google Scholar]
  43. Gunter TE, Yule DI, Gunter KK, Eliseev RA, Salter JD, 2004. Calcium and mitochondria. FEBS Lett 567, 96–102. [DOI] [PubMed] [Google Scholar]
  44. Hamberger A, Hyden H, 1963. Inverse enzymatic changes in neurons and glia during increased function and hypoxia. J Cell Biol 16, 521–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Hayakawa K, Esposito E, Wang X, Terasaki Y, Liu Y, Xing C, Ji X, Lo EH, 2016. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 535, 551–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, Lemere CA, Selkoe DJ, Stevens B, 2016. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Huang L, Nakamura Y, Lo EH, Hayakawa K, 2019. Astrocyte Signaling in the Neurovascular Unit After Central Nervous System Injury. Int J Mol Sci 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Hunter DR, Haworth RA, Southard JH, 1976. Relationship between configuration, function, and permeability in calcium-treated mitochondria. J Biol Chem 251, 5069–5077. [PubMed] [Google Scholar]
  49. Ishii T, Takanashi Y, Sugita K, Miyazawa M, Yanagihara R, Yasuda K, Onouchi H, Kawabe N, Nakata M, Yamamoto Y, Hartman PS, Ishii N, 2017. Endogenous reactive oxygen species cause astrocyte defects and neuronal dysfunctions in the hippocampus: a new model for aging brain. Aging Cell 16, 39–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Ito U, Hakamata Y, Kawakami E, Oyanagi K, 2009. Degeneration of astrocytic processes and their mitochondria in cerebral cortical regions peripheral to the cortical infarction: heterogeneity of their disintegration is closely associated with disseminated selective neuronal necrosis and maturation of injury. Stroke 40, 2173–2181. [DOI] [PubMed] [Google Scholar]
  51. Jackson JG, Robinson MB, 2018. Regulation of mitochondrial dynamics in astrocytes: Mechanisms, consequences, and unknowns. Glia 66, 1213–1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Kacem K, Lacombe P, Seylaz J, Bonvento G, 1998. Structural organization of the perivascular astrocyte endfeet and their relationship with the endothelial glucose transporter: a confocal microscopy study. Glia 23, 1–10. [PubMed] [Google Scholar]
  53. Koehler RC, Roman RJ, Harder DR, 2009. Astrocytes and the regulation of cerebral blood flow. Trends Neurosci 32, 160–169. [DOI] [PubMed] [Google Scholar]
  54. Krajewski S, Krajewska M, Ellerby LM, Welsh K, Xie Z, Deveraux QL, Salvesen GS, Bredesen DE, Rosenthal RE, Fiskum G, Reed JC, 1999. Release of caspase-9 from mitochondria during neuronal apoptosis and cerebral ischemia. Proc Natl Acad Sci U S A 96, 5752–5757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Kubik LL, Philbert MA, 2015. The role of astrocyte mitochondria in differential regional susceptibility to environmental neurotoxicants: tools for understanding neurodegeneration. Toxicol Sci 144, 7–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ, 2009. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science 323, 1211–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Kucukdereli H, Allen NJ, Lee AT, Feng A, Ozlu MI, Conatser LM, Chakraborty C, Workman G, Weaver M, Sage EH, Barres BA, Eroglu C, 2011. Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc Natl Acad Sci U S A 108, E440–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Li T, Chen X, Zhang C, Zhang Y, Yao W, 2019. An update on reactive astrocytes in chronic pain. J Neuroinflammation 16, 140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Liddelow S, Barres B, 2015. SnapShot: Astrocytes in Health and Disease. Cell 162, 1170–1170 e1171. [DOI] [PubMed] [Google Scholar]
  60. Liddelow SA, Barres BA, 2017. Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity 46, 957–967. [DOI] [PubMed] [Google Scholar]
  61. Matsui T, Omuro H, Liu YF, Soya M, Shima T, McEwen BS, Soya H, 2017. Astrocytic glycogen-derived lactate fuels the brain during exhaustive exercise to maintain endurance capacity. Proc Natl Acad Sci U S A 114, 6358–6363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Mauch DH, Nagler K, Schumacher S, Goritz C, Muller EC, Otto A, Pfrieger FW, 2001. CNS synaptogenesis promoted by glia-derived cholesterol. Science 294, 1354–1357. [DOI] [PubMed] [Google Scholar]
  63. McCully JD, Cowan DB, Pacak CA, Toumpoulis IK, Dayalan H, Levitsky S, 2009. Injection of isolated mitochondria during early reperfusion for cardioprotection. American journal of physiology. Heart and circulatory physiology 296, H94–H105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Min SK, Park JS, Luo L, Kwon YS, Lee HC, Shim HJ, Kim ID, Lee JK, Shin HS, 2015. Assessment of C-phycocyanin effect on astrocytes-mediated neuroprotection against oxidative brain injury using 2D and 3D astrocyte tissue model. Sci Rep 5, 14418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Nahirnyj A, Livne-Bar I, Guo X, Sivak JM, 2013. ROS detoxification and proinflammatory cytokines are linked by p38 MAPK signaling in a model of mature astrocyte activation. PloS one 8, e83049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Nett WJ, Oloff SH, McCarthy KD, 2002. Hippocampal astrocytes in situ exhibit calcium oscillations that occur independent of neuronal activity. J Neurophysiol 87, 528–537. [DOI] [PubMed] [Google Scholar]
  67. Nielsen S, Nagelhus EA, Amiry-Moghaddam M, Bourque C, Agre P, Ottersen OP, 1997. Specialized membrane domains for water transport in glial cells: high-resolution immunogold cytochemistry of aquaporin-4 in rat brain. J Neurosci 17, 171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Norris CM, Kadish I, Blalock EM, Chen KC, Thibault V, Porter NM, Landfield PW, Kraner SD, 2005. Calcineurin triggers reactive/inflammatory processes in astrocytes and is upregulated in aging and Alzheimer's models. J Neurosci 25, 4649–4658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Okada S, Nakamura M, Katoh H, Miyao T, Shimazaki T, Ishii K, Yamane J, Yoshimura A, Iwamoto Y, Toyama Y, Okano H, 2006. Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury. Nat Med 12, 829–834. [DOI] [PubMed] [Google Scholar]
  70. Panatier A, Vallee J, Haber M, Murai KK, Lacaille JC, Robitaille R, 2011. Astrocytes are endogenous regulators of basal transmission at central synapses. Cell 146, 785–798. [DOI] [PubMed] [Google Scholar]
  71. Parnis J, Montana V, Delgado-Martinez I, Matyash V, Parpura V, Kettenmann H, Sekler I, Nolte C, 2013. Mitochondrial exchanger NCLX plays a major role in the intracellular Ca2+ signaling, gliotransmission, and proliferation of astrocytes. J Neurosci 33, 7206–7219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Pellerin L, Magistretti PJ, 1994. Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci U S A 91, 10625–10629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Perez-Nievas BG, Serrano-Pozo A, 2018. Deciphering the Astrocyte Reaction in Alzheimer's Disease. Front Aging Neurosci 10, 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Pleiss MM, Sompol P, Kraner SD, Abdul HM, Furman JL, Guttmann RP, Wilcock DM, Nelson PT, Norris CM, 2016. Calcineurin proteolysis in astrocytes: Implications for impaired synaptic function. Biochim Biophys Acta 1862, 1521–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Powell EM, Geller HM, 1999. Dissection of astrocyte-mediated cues in neuronal guidance and process extension. Glia 26, 73–83. [DOI] [PubMed] [Google Scholar]
  76. Price BR, Norris CM, Sompol P, Wilcock DM, 2018. An emerging role of astrocytes in vascular contributions to cognitive impairment and dementia. J Neurochem 144, 644–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Price DL, Ludwig JW, Mi H, Schwarz TL, Ellisman MH, 2002. Distribution of rSlo Ca2+-activated K+ channels in rat astrocyte perivascular endfeet. Brain Res 956, 183–193. [DOI] [PubMed] [Google Scholar]
  78. Quintana DD, Garcia JA, Sarkar SN, Jun S, Engler-Chiurazzi EB, Russell AE, Cavendish JZ, Simpkins JW, 2019. Hypoxia-reoxygenation of primary astrocytes results in a redistribution of mitochondrial size and mitophagy. Mitochondrion 47, 244–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Rossi D, 2015. Astrocyte physiopathology: At the crossroads of intercellular networking, inflammation and cell death. Prog Neurobiol 130, 86–120. [DOI] [PubMed] [Google Scholar]
  80. Rusakov DA, 2015. Disentangling calcium-driven astrocyte physiology. Nat Rev Neurosci 16, 226–233. [DOI] [PubMed] [Google Scholar]
  81. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, Stevens B, 2012. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Schousboe A, Waagepetersen HS, 2005. Role of astrocytes in glutamate homeostasis: implications for excitotoxicity. Neurotox Res 8, 221–225. [DOI] [PubMed] [Google Scholar]
  83. Shi Q, Chowdhury S, Ma R, Le KX, Hong S, Caldarone BJ, Stevens B, Lemere CA, 2017. Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice. Sci Transl Med 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Shields DC, Schaecher KE, Hogan EL, Banik NL, 2000. Calpain activity and expression increased in activated glial and inflammatory cells in penumbra of spinal cord injury lesion. J Neurosci Res 61, 146–150. [DOI] [PubMed] [Google Scholar]
  85. Shields DC, Tyor WR, Deibler GE, Hogan EL, Banik NL, 1998. Increased calpain expression in activated glial and inflammatory cells in experimental allergic encephalomyelitis. Proc Natl Acad Sci U S A 95, 5768–5772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M, 2003. Signaling at the gliovascular interface. J Neurosci 23, 9254–9262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Sofroniew MV, Vinters HV, 2010. Astrocytes: biology and pathology. Acta Neuropathol 119, 7–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Sompol P, Furman JL, Pleiss MM, Kraner SD, Artiushin IA, Batten SR, Quintero JE, Simmerman LA, Beckett TL, Lovell MA, Murphy MP, Gerhardt GA, Norris CM, 2017. Calcineurin/NFAT Signaling in Activated Astrocytes Drives Network Hyperexcitability in Abeta-Bearing Mice. J Neurosci 37, 6132–6148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Stephen TL, Higgs NF, Sheehan DF, Al Awabdh S, Lopez-Domenech G, Arancibia-Carcamo IL, Kittler JT, 2015. Miro1 Regulates Activity-Driven Positioning of Mitochondria within Astrocytic Processes Apposed to Synapses to Regulate Intracellular Calcium Signaling. J Neurosci 35, 15996–16011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, Sher A, Litke AM, Lambris JD, Smith SJ, John SW, Barres BA, 2007. The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178. [DOI] [PubMed] [Google Scholar]
  91. Stobart JL, Ferrari KD, Barrett MJP, Gluck C, Stobart MJ, Zuend M, Weber B, 2018. Cortical Circuit Activity Evokes Rapid Astrocyte Calcium Signals on a Similar Timescale to Neurons. Neuron 98, 726–735 e724. [DOI] [PubMed] [Google Scholar]
  92. Sun C, L X, Wang B, Wang Z, Liu Y, Di C, Si J, Li H, Wu Q, Xu D, Li J, Li G, Wang Y, Wang F, Zhang H, 2019. Endocytosis-mediated mitochondrial transplantation: Transferring normal human astrocytic mitochondria into glioma cells rescues aerobic respiration and enhances radiosensitivity. Theranostics 9, 3595–3607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Terashima T, Ogawa N, Nakae Y, Sato T, Katagi M, Okano J, Maegawa H, Kojima H, 2018. Gene Therapy for Neuropathic Pain through siRNA-IRF5 Gene Delivery with Homing Peptides to Microglia. Mol Ther Nucleic Acids 11, 203–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Ullian EM, Sapperstein SK, Christopherson KS, Barres BA, 2001. Control of synapse number by glia. Science 291, 657–661. [DOI] [PubMed] [Google Scholar]
  95. Vainchtein ID, Chin G, Cho FS, Kelley KW, Miller JG, Chien EC, Liddelow SA, Nguyen PT, Nakao-Inoue H, Dorman LC, Akil O, Joshita S, Barres BA, Paz JT, Molofsky AB, Molofsky AV, 2018. Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science 359, 1269–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Valiante S, Falanga A, Cigliano L, Iachetta G, Busiello RA, La Marca V, Galdiero M, Lombardi A, Galdiero S, 2015. Peptide gH625 enters into neuron and astrocyte cell lines and crosses the blood-brain barrier in rats. Int J Nanomedicine 10, 1885–1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Valori CF, Guidotti G, Brambilla L, Rossi D, 2019. Astrocytes: Emerging Therapeutic Targets in Neurological Disorders. Trends Mol Med 25, 750–759. [DOI] [PubMed] [Google Scholar]
  98. Verkhratsky A, 2019. Astroglial Calcium Signaling in Aging and Alzheimer's Disease. Cold Spring Harb Perspect Biol 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Voloboueva LA, Suh SW, Swanson RA, Giffard RG, 2007. Inhibition of mitochondrial function in astrocytes: implications for neuroprotection. J Neurochem 102, 1383–1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Waitt AE, Reed L, Ransom BR, Brown AM, 2017. Emerging Roles for Glycogen in the CNS. Front Mol Neurosci 10, 73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Wakida NM, Cruz GMS, Ro CC, Moncada EG, Khatibzadeh N, Flanagan LA, Berns MW, 2018. Phagocytic response of astrocytes to damaged neighboring cells. PloS one 13, e0196153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Wang H, Song G, Chuang H, Chiu C, Abdelmaksoud A, Ye Y, Zhao L, 2018. Portrait of glial scar in neurological diseases. Int J Immunopathol Pharmacol 31, 2058738418801406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Wilhelmsson U, Bushong EA, Price DL, Smarr BL, Phung V, Terada M, Ellisman MH, Pekny M, 2006. Redefining the concept of reactive astrocytes as cells that remain within their unique domains upon reaction to injury. Proc Natl Acad Sci U S A 103, 17513–17518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Wilhelmsson U, Li L, Pekna M, Berthold CH, Blom S, Eliasson C, Renner O, Bushong E, Ellisman M, Morgan TE, Pekny M, 2004. Absence of glial fibrillary acidic protein and vimentin prevents hypertrophy of astrocytic processes and improves post-traumatic regeneration. J Neurosci 24, 5016–5021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Wyss-Coray T, Loike JD, Brionne TC, Lu E, Anankov R, Yan F, Silverstein SC, Husemann J, 2003. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med 9, 453–457. [DOI] [PubMed] [Google Scholar]
  106. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O'Keeffe S, Phatnani HP, Guarnieri P, Caneda C, Ruderisch N, Deng S, Liddelow SA, Zhang C, Daneman R, Maniatis T, Barres BA, Wu JQ, 2014. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci 34, 11929–11947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Zhu D, Hu C, Sheng W, Tan KS, Haidekker MA, Sun AY, Sun GY, Lee JC, 2009. NAD(P)H oxidase-mediated reactive oxygen species production alters astrocyte membrane molecular order via phospholipase A2. Biochem J 421, 201–210. [DOI] [PubMed] [Google Scholar]

RESOURCES