Abstract
It is established that increased autophagy is readily detectable after various types of acute brain injury, including trauma, focal and global cerebral ischemia. What remains controversial, however, is whether this heightened detection of autophagy in brain represents a homeostatic or pathologic process, or an epiphenomenon. The ultimate role of autophagy after acute brain injury likely depends upon: (1) the degree of brain injury and the overall autophagic burden; (2) the capacity of individual cell types to ramp up autophagic flux; (3) the local redox state and signaling of parallel cell death pathways; (4) the capacity to eliminate damage associated molecular patterns and toxic proteins and metabolites both intra- and extracellularly; and (5) the timing of the pro- or anti-autophagic intervention. In this review, we attempt to reconcile conflicting studies that support both a beneficial and detrimental role for autophagy in models of acute brain injury.
Keywords: Autophagic flux, cerebral ischemia, hypoxia-ischemic brain injury, traumatic brain injury
Introduction
Autophagy is vital to cellular homeostasis in brain and is tightly regulated under normal conditions. In conditions of stress (e.g. nutrient deprivation, starvation) or acute cellular injury (e.g. trauma, ischemia-reperfusion), autophagy is increased and appears to be proportional to the degree of stress/injury. Increased detection of autophagy has been reported after various types of acute brain injury, including traumatic brain injury (TBI), stroke, global cerebral ischemia, and seizures. However, whether this heightened detection of autophagy in brain represents a protective response, pathologic process, or an epiphenomenon remains controversial, with reports in experimental models showing beneficial effects of both promoting and inhibiting autophagy (1). Indeed, since the review by Smith et al. (2) there have been over 400 new citations listed on PubMed related to autophagy after acute brain injury promulgating mechanistic insights, but without further clarifying the role of autophagy in brain after injury. Our objective is to review recent studies and novel mechanistic discoveries, and attempt to provide a speculative synthesis as to the role of autophagy after acute brain injury.
Autophagy after brain injury
The presence of autophagosomes in the mature brain normally and during starvation is lower compared with other organs (heart, liver, pancreas, kidney, skeletal muscle) (3), a finding that could be related to reduced autophagy and/or enhanced turnover (4). However, after acute brain injury, there is compelling evidence that autophagy is induced, observed in multiple experimental models by ultrastructural identification of autophagosomes and autolysosomes, and/or the presence of autophagy-related microtubule-associated protein 1 light chain 3-II (LC3-II) detected using western blot or immunohistochemistry (1). Increased detection of autophagosomes may be a consequence of increased autophagy itself, or impaired autophagic flux—the process of fusion with lysosomes to form autolysosomes, and subsequent degradation (Figure 1). During starvation-induced autophagy triggered in response to nutrient deprivation, or homeostatic autophagy recycling aging organelles and dysfunctional proteins, autophagic flux is balanced, i.e. can keep up with demand. Under conditions of injury or disease autophagic flux may be impaired and/or overwhelmed and unable to keep up with generation of autophagosomes. Impaired autophagic flux and lysosomal function have been suggested to contribute to Parkinson’s disease (5), Alzheimer’s disease (6), and TBI (7). Enhancement of autophagic flux by activating transcription factor EB is protective in experimental brain injury produced by cadmium (8).
Figure 1. A hypothetical view of the continuum from homeostatic autophagy to autosis after brain injury.
In homeostasis autophagosomes formation and autophagic flux are in equilibrium. After injury the cell’s capacity to turn over autophagosomes may be exceeded, perhaps leading to secretory autophagy. Exhaustion of the cell’s autophagic capacity may lead to autosis. Both secretory autophagy and cell disruption during autosis may lead to release of extracellular signals (DAMPs) and autophagic proteins that may regulate inflammation, apoptosis, and/or autophagy. These extracellular signals, neurotoxic proteins such as Aβ and tau, and other macromolecules can be eliminated via the glymphatic pathway, potentially reducing their impact. Therapies and/or interventions that (1) stimulate autophagic flux, (2) reduce autophagic burden, and/or (3) enhance clearance of DAMPs and neurotoxic proteins, may help restore cellular homeostasis. Representative electron micrographs from ultrathin sections taken from the CA1 region of the hippocampus. (Left to right: naïve 20 day old female rat, bar = 2 µm; 20 day old male rat 72 hours after a 9 min asphyxia cardiac arrest, bar = 500 nm; 20 day old female rat 72 hours after a 9 minute asphyxial cardiac arrest, bar = 2 µm (nm = nuclear membrane; pm = plasma membrane). Images courtesy of Mara Sullivan, Center for Biologic Imaging, University of Pittsburgh.
After acute brain injury, several initiators of autophagy have been observed. These include classic pathways regulated by phosphatidylinositol-3-kinase (PI3K) and AMP-dependent protein kinase (AMPK), and recent discoveries focusing on selective clearance of mitochondria using autophagic components, termed mitophagy (9). Mitochondrial damage/dysfunction is a common denominator in acute brain injury and in aging mitochondria. Upstream redox-related pathways regulated by extracellular signal-regulated protein kinases (10) and Nrf2/Keap1 (11) link oxidative stress and mitophagy. In addition, other mitochondria-specific signaling pathways have been reported involving phosphatase and tensin homolog-induced putative kinase 1 (PINK1)/parkin RING-between-RING E3 ubiquitin protein ligase (PARK2) (12) and cardiolipin externalization to the outer mitochondrial membrane (13). Under conditions of overwhelming mitochondrial damage complete cellular failure accompanied by exhaustion of recyclable energy stores and plasma membrane rupture can be observed, culminating in a form of regulated cell death termed “autosis” (14), which may be thought of as “terminal autophagy”.
Autophagy after TBI
The presence of autophagosomes and/or an increase in LC3-II in injured brain have been observed in several experimental models of TBI including fluid-percussion injury (FPI) (15), controlled cortical impact (CCI) (16), and weight drop injury (17). Autophagosomes and LC3-II have also been reported in human brain specimens from adult patients after TBI (18). In children after severe TBI, p62/sequestosome 1 (SQSTM1) and Beclin 1 are increased in cerebrospinal fluid (CSF) compared with CSF from control patients, with p62 concentration associated with unfavorable outcome, consistent with increased overwhelmed and/or impaired autophagic flux (19).
Using a weight-drop model which produces diffuse brain injury, Liu et al. (20) recently showed that caloric restriction after mild TBI results in increased autophagy markers in the CA3 region of the hippocampus, with a twofold increase in Beclin 1, a threefold increase in LC3, as well as decreased mammalian target of rapamycin (mTOR) compared to injured animals with normal diets. Calorie-restricted mice also demonstrated better cognitive performance, measured by escape latency on Morris-water maze testing. Wang et al. (21) more directly evaluated the mTOR pathway’s involvement in TBI-related autophagy in a weight drop model. After repetitive mild TBI, rats given rapamycin via intraperitoneal injection four hours after injury showed decreased mTOR, increased Beclin 1, as well as increased mitophagy-related protein PINK1. Rapamycin-treated rats also had decreased hippocampal neuron apoptosis and improved performance in beam walking and open field tasks.
After CCI, a contusion model of TBI, Ma et al. (22) tested the experimental cancer drug 17-allyamino-demethoxygeldanamycin (17-AAG). The drug showed beneficial effects, including reduced brain water content, improved functional assessment, and improved cortical neuron survival compared to injured animals treated with vehicle only. 17-AAG also resulted in a twofold increase in both LC3-II and Beclin 1. The effects of 17-AAG were amplified by inducing autophagy with the mTOR inhibitor rapamycin, and diminished by inhibiting autophagy with the PI3K inhibitor 3-methyladenine (3-MA) (22). An increase in the presence of autophagosomes, reduced neuronal death, and better behavioral outcome have also been reported in animals treated with methylene blue (23) and hydrogen sulfide after CCI (24). Mice treated with hydrogen sulfide after CCI had near-complete reversal of a TBI-induced decrease in cortical and hippocampal p62/SQSTM1 (24). Taken together, these studies are consistent with the possibility that enhancing autophagy improves neurological outcome after TBI.
Autophagy has also been linked to reduced neuroinflammation after TBI. After FPI in rats, Jin et al. (25) observed an increase in microglial activation, with increased expression of ionized calcium-binding adapter molecule 1 (Iba1) suggestive of microglial activation in the hippocampus ipsilateral to the injury site. Tumor necrosis factor α (TNFα) and interleukin-1β (IL-1β) expression also increased following injury. Inhibition of autophagy with 3-MA 24 hours after injury amplified the injury-induced increases in all three proteins, indicating a possible relationship between autophagy, microglial activation, and inflammation. Post-treatment with fucoxanthin, an antioxidant carotenoid compound, was associated with reduced lesion volume, edema, and less severe neurologic deficits after weight drop TBI in mice (26). Fucoxanthin was also associated with greater glutathione peroxidase (GPx) activity, an amplified TBI-associated increase in Beclin 1 and LC3-II and decrease in p62/SQSTM1, and attenuated cleavage of inflammasome-related caspase-1. Caution is in order, however, as neuroinflammation after TBI is highly complex and likely has multifaceted and temporally-dependent roles, and reduced inflammation in these studies may simply represent a proportional response to reduced cell death (27).
Since autophagy and apoptosis are interrelated, complex modeling is required to separate crosstalk and overlap versus distinctions between the two pathways (28). Using an open-skull weight-drop model of TBI in rats, the non-selective antioxidant tetrahydrocurcumin reduced apoptosis and neurologic severity, while increasing Beclin 1 and LC3-II abundance as well as GPx activity compared with vehicle-treated rats (29). Neuroprotection was partially reversed with 3-MA, suggesting that increased autophagy produced by tetrahydrocurcumin leads to a concomitant reduction in apoptosis after TBI; however, it is also possible that inhibition of apoptosis using non-selective antioxidant treatment reduces apoptotic cell death at the expense of increased autophagy (30).
In contrast to the studies discussed above, divergent studies have suggested potentially detrimental effects of autophagy after TBI. After CCI in mice, Zhang et al. recently reported reduced lesion volume, neuronal apoptosis, and behavioral deficits in mice treated with the signaling protein Wnt3a (31). An observed decrease in TBI-induced Beclin 1 and LC3-II abundance led to the authors to conclude that suppressing autophagy is beneficial after TBI. Multiple antioxidant compounds have been evaluated in experimental TBI models, with improved histologic and behavioral outcomes observed concomitantly with a reduction in biomarkers of autophagy (32–34). Given that antioxidants have the potential to attenuate many cellular processes associated with multiple cell death pathways, these findings may simply reflect a reduction in cell stress and therefore less provocation of autophagy. Alternatively, antioxidants may increase autophagic flux or capacity after TBI. Liu et al. (35) induced expression of ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) in rats subjected to CCI. UCH-L1 overexpression improved survival of hippocampal neurons, reduced accumulation of amyloid-precursor protein, and decreased Beclin 1 and LC3-II in injured rats. Given a role for UCH-L1 in proteasomal clearance of misfolded proteins, these results may reflect increased autophagic flux and/or decreased intracellular demand for autophagy. Similarly, neuroprotection and decreased abundance of Beclin 1 and LC3-II observed when rats are treated with rosiglitazone after CCI may reflect anti-oxidant and other salutary effects of the drug and may not represent cause and effect (36). However, these studies are consistent with effective autophagic flux corresponding with improved neurological outcome after TBI.
Clinically used sedative medications have also been shown to exert neuroprotective effects in experimental TBI. A recent study using a weight-drop model demonstrated improved behavioral outcome and reduced Beclin 1 and LC3-II abundance following treatment with the NMDA receptor antagonist ketamine (37). Ketamine treatment resulted in partial preservation of cellular ATP stores in addition to attenuation of TNFα and interleukin-6 (IL-6) compared with vehicle. One could speculate that ketamine reduces neuronal excitation and therefore metabolic demands in brain, thereby reducing autophagic pressure.
Autophagic flux after TBI
Interpretation of Beclin 1, p62/SQSTM1, and LC3-II abundance as a reflection of autophagy as a whole is likely an oversimplification, as measuring snapshots of autophagy biomarkers do not reflect the dynamic process of initiation, nucleation, elongation, autophagosome formation, fusion with lysosomes to form autolysosomes, and degradation. Yin et al. recently reported TBI-induced increases in autophagy-related protein expression in the hippocampus, as well as the lysosome-associated membrane protein 1 (LAMP-1) and the lysosomal protease cathepsin D (38). TBI impaired cleavage of cathepsin D to its active form, indicating impaired lysosomal function. Treatment with docosahexanoic acid restored lysosomal function, attenuated hippocampal and white matter damage, and improved performance in the Morris-water maze. Cui et al. recently evaluated calcitriol treatment after CCI, finding evidence of enhanced autophagic flux determined by LC3-II/LC3-I ratio and improved neurological function with treatment (39). Inhibiting lysosomal function and endosomal acidification with chloroquine reversed the effects of calcitriol. Similar findings were reported when chloroquine was administered with lanthionine-ketamine ethyl ester after FPI in mice (40). Taken together, these studies suggest that enhancing autophagic flux in brain may be beneficial after TBI.
Zeng et al. recently examined induction of autophagy and autophagic flux after CCI of varying injury severity (41). LC3-II abundance was increased in ipsilateral cortical tissue at 1 and 3 days in an injury severity-dependent manner. After mild CCI increased induction of autophagy-related proteins Beclin 1, Atg5 and Atg12 concomitant with a reduction in p62/SQSTM1 and autophagosomes, and an increase in autolysosomes was seen, consistent with enhanced autophagic flux. Treatment with chloroquine after mild CCI resulted in sustained LC3-II abundance (vs. sham), increased apoptosis, and worse neurological outcome (vs. vehicle treatment). After moderate or severe CCI a lack of conversion from autophagosomes to autolysosomes was seen, consistent with an impairment in autophagic flux. These data suggest that the autophagic flux capacity may be injury-severity dependent.
Autophagy after ischemia-reperfusion
As with TBI, divergent outcomes in in vivo and in vitro studies have led to conflicting insights regarding the role of autophagy after ischemia-reperfusion in brain. These divergent outcomes may in part be explained by cell-type and brain region-specific differences in response to ischemia-reperfusion. In astrocyte and neuron co-cultures subjected to oxygen-glucose deprivation (OGD), enhancement of autophagy in astrocytes with rapamycin reduced apoptotic neuronal death, whereas inhibiting autophagy in astrocytes with 3-MA or small interfering RNA (siRNA) against Atg5 increased apoptotic neuronal death (42). In astrocyte monocultures, Kasprowska et al. observed that inhibiting autophagy with 3-MA increased apoptosis at 1 and 4 hours, and necrosis at 8 and 24 hours after OGD (43). In contrast, other studies have shown that 3-MA reduces neuronal death after OGD (44), and that inhibiting induction of autophagy using siRNA targeting Atg7 reduces neuronal death after nutrient deprivation (45). Interestingly, in the study by Du et al., while Atg7 siRNA protected primary neurons it exacerbated cell death in fibroblasts, suggesting that the role of autophagy in response to nutrient deprivation and/or ischemia may differ in non-dividing (primary neurons) versus dividing (fibroblasts, astrocytes) cells (45).
Multiple studies in experimental models of ischemia-reperfusion injury suggest that augmenting autophagy using rapamycin is neuroprotective, including studies by Carloni et al. using a neonatal model of hypoxia-ischemia (46) and by Buckley et al. using both transient and permanent middle cerebral artery occlusion (MCAO) (47). In a more recent study in 10 day old rats after unilateral carotid artery ligation and hypoxemia, preventing microRNA (miR)-30d-5p associated Beclin 1 suppression using antagomir enhanced autophagy, inhibited apoptosis, reduced infarct volume, and improved neurological outcome (48). In contrast to studies showing that pharmacologically augmenting autophagy is protective after cerebral ischemia-reperfusion in vivo, are reports such as those by Puyal et al. showing that 3-MA reduces lesion volume in 12 day old rats after MCAO (49). More recently, Dong et al. observed that rapamycin exacerbates neuronal death in rats after forebrain ischemia (50) and Wang et al. observed that 3-MA reduced infarct volume after ischemia-reperfusion (51). In a model of chronic cerebral ischemia, the fatty acid amide hydrolase URB597 reduced Beclin 1 and LC3-II abundance and attenuated cognitive impairment, an effect was reversed with co-administration of 3-MA (52).
Caveats related to pharmacologic modulation of autophagy include off-target effects of the drug(s). However, more selective approaches targeting autophagy have also generated conflicting results. In Atg7 deficient mice, unilateral carotid artery ligation followed by hypoxemia in 7 or 9 day old mice results in reduced autophagy and hippocampal neuronal death (53, 54). Studies in Atg7 deficient mice are confounded by the fact that inhibition of autophagy throughout development leads to neurodegeneration (55). In a 9-minute asphyxial cardiac arrest model in juvenile rats, targeted inhibition of Atg7 using siRNA in the cerebellum reduced LC3-II and improved motor function and Purkinje cell survival (56). In contrast, targeted overexpression of Atg7 in astrocytes using adeno-associated virus (AAV)-GFAP-Atg7 reduced infarct volume in mice after MCAO (42). It is interesting to note that inhibiting neuronal Atg7 using Atg7flox/+;Nes-Cre (54), and over-expressing astrocyte Atg7 using AAV-GFAP-Atg7 both appear neuroprotective (42).
Autophagic flux after ischemia-reperfusion
Evaluating the protective effects of sodium hydrosulfide after transient MCAO in mice, Zhu et al. co-administered bafilomycin A1 to inhibit formation of autolysosomes (57). Co-administration of bafilomycin A1 reversed the pro-autophagic and neuroprotective effects of sodium hydrosulfide. Following MCAO in diabetic rats, administration of chloroquine exacerbates brain microvascular endothelial cell injury presumably via inhibiting lysosomal function (58). In a model of transient forebrain ischemia, Zhan et al. observed ischemia-induced increases in LC3-II and p62 in CA1 hippocampal neurons (59). LAMP2 was decreased in neurons but increased in glial cells. Inhibiting autophagic flux with chloroquine decreased cell survival and increased LC3 and p62, and hypoxic preconditioning partially reversed injury these effects. These findings indicate that preventing lysosomal fusion and subsequent formation of autolysosomes and clearance of autophagosomes exacerbates brain injury. However, in contrast, after asphyxia cardiac arrest in rats treatment with bafilomycin A1 (or 3-MA) reduces hippocampal neuronal death (60).
Recently, Zhou et al. evaluated autophagic flux in hippocampal neurons in vitro (61). OGD resulted in decreased lysosomal number and acidity, and decreased LAMP2. Using a GFP-RFP-tf-LC3 assay, the investigators quantified autolysosomes and autophagosomes. OGD decreased the ratio of autolysosomes to autophagosomes, indicating impaired flux. Notably, mild hypothermia in this study enhanced autophagic flux after OGD in neurons. A reduction in LAMP2 and an increase in LC3-II is also reported after permanent MCAO in rats (62). The experimental compound pseudoginsenoside-F11, which has been reported to have wide-ranging effects, improved lysosomal function and reduced infarct volume, but not with co-administration of chloroquine.
Secretory autophagy and brain injury
Secretory autophagy refers to a specialized variation of autophagy, which begins with canonical initiation of autophagy and culminates in autophagosome fusion with the plasma membrane and expulsion of contents, rather than lysosomal fusion, formation of autolysosomes, and degradation (63, 64). In contrast to conventional secretory pathways, secretory autophagy allows cells to clear proteins lacking N-terminal signal peptides, and aggregation-prone proteins including amyloid β (Aβ) (65, 66). When the degradative final steps of autophagic flux are inhibited, secretory autophagy can be induced to rid cells of dysfunctional proteins. For example, in an in vitro model of Parkinson’s disease, Ejlerskov et al. observed that inhibiting autophagosome mobility hindered their fusion with lysosomes and increased extracellular secretion of α-synuclein (67). Neurodegenerative disorders including Alzheimer’s disease and Parkinson’s disease are characterized by impaired autophagy and both intracellular and extracellular accumulation of abnormal proteins (68–70). Extracellular accumulation of these same proteins are implicated in chronic traumatic encephalopathy (71, 72).
Extracellular macromolecules, including misfolded proteins such as Aβ, are carried through the brain interstitial space via bulk flow of fluid, and removed from the brain via paravascular channels (termed the glymphatic system due to their delineation by astrocyte foot processes) that eventually drain into the lymphatic system (73). Impaired flow through the glymphatic system preceded reduced Aβ clearance in a mouse model of Alzheimer’s disease (74). Glymphatic flow depends on movement of water through aquaporin channels. Genetic knockout of aquaporin-4 in a mouse model of impairs glymphatic flow and promotes pathologic accumulation of phosphorylated tau (75). These studies may highlight a mechanistic link between a cell’s autophagic capacity, overwhelmed autophagic flux, secretory autophagy, extracellular accumulation of neurotoxic proteins, and glymphatic elimination (Figure 1). In a repetitive mild TBI model in mice, Saykally et al. reported decreased LC3-II, LAMP2, and Atg7 at 3 days, and a subsequent increased aggregation of transactivation response DNA protein 43, a protein associated with neurodegeneration (76).
Secretory autophagy and the efficiency of glymphatic elimination may also effect regulation of inflammation and cell death (Figure 1). Contents of secreted autophagosomes are known to contain a number of damage-associated molecular patterns (DAMPs), including high-mobility group box 1 (HMGB1), ATP, IL-1β, and DNA (77). HMGB1, IL-1β, and mitochondrial DNA have all been shown to increase in CSF from patients after severe TBI (78–80). In the case of HMGB1, its release has been shown to regulate Beclin 1-dependent autophagy in cancer cells (81). A recent study suggests that the protective effects of ischemic preconditioning may be associated with concomitant reduction in plasma HMGB1 and autophagy (82). Interestingly, reducible HMGB1 induces autophagy and promotes tumor resistance to chemotherapeutic agents, whereas oxidized HMGB1 increases apoptosis and sensitizes tumor cells. Oxidative stress is universally observed in all types of acute brain injury, and is thought to regulate autophagy (11). Autosis, mediated by the Na+, K+-ATPase pump, displays numerous autophagosomes and autolysosomes without late stage secondary lysosomes, and plasma membrane rupture suggesting osmotic disequilibrium (83). Intriguingly, there is also a link between HMGB1 and the water channel aquaporin (84). It is tempting to speculate that secretory autophagy of DAMPs and autophagy-related proteins may regulate inflammation, autophagy, and/or apoptosis in neighboring cells, perhaps acting as “survival signals”, whereas overwhelming autophagy exceeding the capacity of autophagic flux may lead to toxic accumulation of autophagosomes and autosis, the consequence of which would be large-scale release of DAMPs and other “surrender signals”. Extracellular signaling of autophagy by DAMPs would also be affected by the rate of glymphatic flux.
Where does the field stand now?
Taken together, studies of autophagy after acute brain injury are consistent in the following: 1) autophagosomes and autophagy-related proteins are more readily detectable after acute brain injury and this may be due to increased autophagy itself and/or impaired autophagic flux; 2) just as autophagy is a vital homeostatic process, some degree of autophagy represents a homeostatic response to cellular insults; 3) feedback regulation of autophagy may be controlled by both intracellular and extracellular autophagic flux; and 4) glymphatic flow may work in concert with secretory autophagy to clear potentially toxic proteins and metabolic waste products from the brain.
What remains less clear is whether inhibition or augmentation of autophagy after acute TBI or cerebral ischemia produces detrimental or beneficial effects, since all combinations have reported in various experimental models. Definitive studies have been hampered by the lack of highly selective pharmacological tools to manipulate autophagy. Case in point, the most commonly used drugs in studies of autophagy, rapamycin to stimulate autophagy and 3-MA to inhibit it, each have multiple off-target effects. Rapamycin is a clinically used immunosuppressive drug that has effects on inflammation, cell cycle arrest and cell proliferation. 3-MA also inhibits non-class III PI3-kinases and promotes glycogenolysis. Furthermore, knockout studies targeting autophagy-related genes to-date have not utilized inducible transgenic mice. Further study using more autophagy-selective pharmacological interventions, inducible, cell type-specific transgenic animals, and/or genome editing in models of acute brain injury would assist greatly in this regard.
In the meantime, we have organized recently published interventions where their effect on autophagy-related protein abundance, autophagosome formation, and/or autophagic flux was evaluated, along with at least one neurological outcome, after experimental acute brain injury in vivo (Figure 2). Interventions were further organized based on the stage of the autophagic process targeted, i.e. initiation, elongation, fusion, or degradation. Most publications report benefit of the intervention whether the effect is to promote or inhibit autophagy. The most generalizable inference is that inhibiting autolysosome formation or lysosome function without reducing autophagosome formation appears detrimental after acute brain injury. Taken together, interventions that stimulate autophagic flux, reduce autophagosome abundance, and enhance autophagosome clearance may afford neuroprotection in experimental models. The clinical corollary is the observation that higher CSF p62/SQSTM1 is associated with poor neurological outcome after severe TBI in children (19).
Figure 2. Recently published interventions organized based on the stage of the autophagic process targeted.
Interventions are separated based on whether they enhance (green box) or inhibit (pink box) autophagy after acute brain injury (T = TBI; I = cerebral ischemia) in vivo. Interventions that produce detrimental effects are typed in red font (note that some have been reported to have both beneficial and detrimental effects).
Conclusion
Recent studies of autophagy in acute brain injury have provided new mechanistic insights, while at the same time highlighting the complexity of the autophagic process from autophagosome formation to intracellular and extracellular clearance of macromolecules, related to interconnected signaling mechanisms and crosstalk between cell death pathways. The role of autophagy after acute brain injury likely depends upon: the degree of brain injury and the overall autophagic burden; the capacity of individual cell types to ramp up autophagic flux; the local redox state and signaling of parallel cell death pathways; the capacity to eliminate DAMPs and toxic proteins and metabolites both intra- and extracellularly; and the specific component of the autophagic process targeted. What appears consistent is that interventions that offload the intracellular burden of autophagosomes (reducing formation, stimulating autophagic flux, enhancing clearance) may afford neuroprotection after acute brain injury in experimental models. However, while much has been learned since the review by Smith et al. (2), understanding of autophagy’s role after acute brain injury remains in a state of flux.
Acknowledgments
Supported by NIH T32 HD040686 (MSW) and the Children’s Hospital of Pittsburgh Scientific Program. We thank Mara Sullivan for technical support in obtaining the electron micrographs in Figure 2.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Galluzzi L, Bravo-San Pedro JM, Blomgren K, Kroemer G. Autophagy in acute brain injury. Nat Rev Neurosci. 2016;17(8):467–484. doi: 10.1038/nrn.2016.51. [DOI] [PubMed] [Google Scholar]
- 2.Smith CM, Chen Y, Sullivan ML, Kochanek PM, Clark RS. Autophagy in acute brain injury: feast, famine, or folly? Neurobiol Dis. 2011;43(1):52–59. doi: 10.1016/j.nbd.2010.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell. 2004;15(3):1101–1111. doi: 10.1091/mbc.E03-09-0704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Boland B, Nixon RA. Neuronal macroautophagy: from development to degeneration. Mol Aspects Med. 2006;27(5–6):503–519. doi: 10.1016/j.mam.2006.08.009. [DOI] [PubMed] [Google Scholar]
- 5.Su CJ, Feng Y, Liu TT, Liu X, Bao JJ, Shi AM, Hu DM, Liu T, Yu YL. Thioredoxin-interacting protein induced alpha-synuclein accumulation via inhibition of autophagic flux: Implications for Parkinson's disease. CNS Neurosci Ther. 2017;23(9):717–723. doi: 10.1111/cns.12721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bordi M, Berg MJ, Mohan PS, Peterhoff CM, Alldred MJ, Che S, Ginsberg SD, Nixon RA. Autophagy flux in CA1 neurons of Alzheimer hippocampus: Increased induction overburdens failing lysosomes to propel neuritic dystrophy. Autophagy. 2016;12(12):2467–2483. doi: 10.1080/15548627.2016.1239003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sarkar C, Zhao Z, Aungst S, Sabirzhanov B, Faden AI, Lipinski MM. Impaired autophagy flux is associated with neuronal cell death after traumatic brain injury. Autophagy. 2014;10(12):2208–2222. doi: 10.4161/15548627.2014.981787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pi H, Li M, Tian L, Yang Z, Yu Z, Zhou Z. Enhancing lysosomal biogenesis and autophagic flux by activating the transcription factor EB protects against cadmium-induced neurotoxicity. Sci Rep. 2017;7:43466. doi: 10.1038/srep43466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zimmermann M, Reichert AS. How to get rid of mitochondria: crosstalk and regulation of multiple mitophagy pathways. Biol Chem. 2017;399(1):29–45. doi: 10.1515/hsz-2017-0206. [DOI] [PubMed] [Google Scholar]
- 10.Zhu JH, Horbinski C, Guo F, Watkins S, Uchiyama Y, Chu CT. Regulation of autophagy by extracellular signal-regulated protein kinases during 1-methyl-4-phenylpyridinium-induced cell death. Am J Pathol. 2007;170(1):75–86. doi: 10.2353/ajpath.2007.060524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hensley K, Harris-White ME. Redox regulation of autophagy in healthy brain and neurodegeneration. Neurobiol Dis. 2015;84:50–59. doi: 10.1016/j.nbd.2015.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, Sideris DP, Fogel AI, Youle RJ. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature. 2015;524(7565):309–314. doi: 10.1038/nature14893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, Tyurin VA, Yanamala N, Shrivastava IH, Mohammadyani D, et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol. 2013;15(10):1197–1205. doi: 10.1038/ncb2837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Liu Y, Shoji-Kawata S, Sumpter RM, Jr, Wei Y, Ginet V, Zhang L, Posner B, Tran KA, Green DR, Xavier RJ, et al. Autosis is a Na+,K+-ATPase-regulated form of cell death triggered by autophagy-inducing peptides, starvation, and hypoxia-ischemia. Proc Natl Acad Sci U S A. 2013;110(51):20364–20371. doi: 10.1073/pnas.1319661110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Liu CL, Chen S, Dietrich D, Hu BR. Changes in autophagy after traumatic brain injury. J Cereb Blood Flow Metab. 2008;28(4):674–683. doi: 10.1038/sj.jcbfm.9600587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lai Y, Hickey RW, Chen Y, Bayir H, Sullivan ML, Chu CT, Kochanek PM, Dixon CE, Jenkins LW, Graham SH, et al. Autophagy is increased after traumatic brain injury in mice and is partially inhibited by the antioxidant gamma-glutamylcysteinyl ethyl ester. J Cereb Blood Flow Metab. 2008;28(3):540–550. doi: 10.1038/sj.jcbfm.9600551. [DOI] [PubMed] [Google Scholar]
- 17.Sun L, Gao J, Zhao M, Jing X, Cui Y, Xu X, Wang K, Zhang W, Cui J. The effects of BMSCs transplantation on autophagy by CX43 in the hippocampus following traumatic brain injury in rats. Neurol Sci. 2014;35(5):677–682. doi: 10.1007/s10072-013-1575-6. [DOI] [PubMed] [Google Scholar]
- 18.Clark RS, Bayir H, Chu CT, Alber SM, Kochanek PM, Watkins SC. Autophagy is increased in mice after traumatic brain injury and is detectable in human brain after trauma and critical illness. Autophagy. 2008;4(1):88–90. doi: 10.4161/auto.5173. [DOI] [PubMed] [Google Scholar]
- 19.Au AK, Aneja RK, Bayir H, Bell MJ, Janesko-Feldman K, Kochanek PM, Clark RSB. Autophagy Biomarkers Beclin 1 and p62 are Increased in Cerebrospinal Fluid after Traumatic Brain Injury. Neurocrit Care. 2017;26(3):348–355. doi: 10.1007/s12028-016-0351-x. [DOI] [PubMed] [Google Scholar]
- 20.Liu Y, Wang R, Zhao Z, Dong W, Zhang X, Chen X, Ma L. Short-term caloric restriction exerts neuroprotective effects following mild traumatic brain injury by promoting autophagy and inhibiting astrocyte activation. Behav Brain Res. 2017;331:135–142. doi: 10.1016/j.bbr.2017.04.024. [DOI] [PubMed] [Google Scholar]
- 21.Wang C, Hu Z, Zou Y, Xiang M, Jiang Y, Botchway BOA, Huo X, Du X, Fang M. The post-therapeutic effect of rapamycin in mild traumatic brain-injured rats ensuing in the upregulation of autophagy and mitophagy. Cell Biol Int. 2017;41(9):1039–1047. doi: 10.1002/cbin.10820. [DOI] [PubMed] [Google Scholar]
- 22.Ma L, Li Z, Liu Z, Li M, Sui D, Liu Y, Shao W, Wang B, Liu P, Li G. 17AAG improves histological and functional outcomes in a rat CCI model through autophagy activation and apoptosis attenuation. Neurosci Lett. 2015;599:1–6. doi: 10.1016/j.neulet.2015.05.004. [DOI] [PubMed] [Google Scholar]
- 23.Zhao M, Liang F, Xu H, Yan W, Zhang J. Methylene blue exerts a neuroprotective effect against traumatic brain injury by promoting autophagy and inhibiting microglial activation. Mol Med Rep. 2016;13(1):13–20. doi: 10.3892/mmr.2015.4551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zhang M, Shan H, Chang P, Wang T, Dong W, Chen X, Tao L. Hydrogen sulfide offers neuroprotection on traumatic brain injury in parallel with reduced apoptosis and autophagy in mice. PLoS One. 2014;9(1):e87241. doi: 10.1371/journal.pone.0087241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Jin Y, Wang R, Yang S, Zhang X, Dai J. Role of Microglia Autophagy in Microglia Activation After Traumatic Brain Injury. World Neurosurg. 2017;100:351–360. doi: 10.1016/j.wneu.2017.01.033. [DOI] [PubMed] [Google Scholar]
- 26.Zhang L, Wang H, Fan Y, Gao Y, Li X, Hu Z, Ding K, Wang Y, Wang X. Fucoxanthin provides neuroprotection in models of traumatic brain injury via the Nrf2-ARE and Nrf2-autophagy pathways. Sci Rep. 2017;7:46763. doi: 10.1038/srep46763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Simon DW, McGeachy MJ, Bayir H, Clark RS, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol. 2017;13(3):171–191. doi: 10.1038/nrneurol.2017.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Liu B, Oltvai ZN, Bayir H, Silverman GA, Pak SC, Perlmutter DH, Bahar I. Quantitative assessment of cell fate decision between autophagy and apoptosis. Sci Rep. 2017;7(1):17605. doi: 10.1038/s41598-017-18001-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Gao Y, Zhuang Z, Gao S, Li X, Zhang Z, Ye Z, Li L, Tang C, Zhou M, Han X, et al. Tetrahydrocurcumin reduces oxidative stress-induced apoptosis via the mitochondrial apoptotic pathway by modulating autophagy in rats after traumatic brain injury. Am J Transl Res. 2017;9(3):887–899. [PMC free article] [PubMed] [Google Scholar]
- 30.Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 2007;8(9):741–752. doi: 10.1038/nrm2239. [DOI] [PubMed] [Google Scholar]
- 31.Zhang JY, Lee J, Gu X, Wei Z, Harris MJ, Yu SPP, Wei L. Intranasally Delivered Wnt3a Improves Functional Recovery after Traumatic Brain Injury by Modulating Autophagic, Apoptotic and Regenerative Pathways in the Mouse Brain. J Neurotrauma. 2017 doi: 10.1089/neu.2016.4871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cordaro M, Impellizzeri D, Paterniti I, Bruschetta G, Siracusa R, De Stefano D, Cuzzocrea S, Esposito E. Neuroprotective Effects of Co-UltraPEALut on Secondary Inflammatory Process and Autophagy Involved in Traumatic Brain Injury. J Neurotrauma. 2016;33(1):132–146. doi: 10.1089/neu.2014.3460. [DOI] [PubMed] [Google Scholar]
- 33.Feng Y, Cui C, Liu X, Wu Q, Hu F, Zhang H, Ma Z, Wang L. Protective Role of Apocynin via Suppression of Neuronal Autophagy and TLR4/NF-kappaB Signaling Pathway in a Rat Model of Traumatic Brain Injury. Neurochem Res. 2017;42(11):3296–3309. doi: 10.1007/s11064-017-2372-z. [DOI] [PubMed] [Google Scholar]
- 34.Feng Y, Cui Y, Gao JL, Li R, Jiang XH, Tian YX, Wang KJ, Li MH, Zhang HA, Cui JZ. Neuroprotective effects of resveratrol against traumatic brain injury in rats: Involvement of synaptic proteins and neuronal autophagy. Mol Med Rep. 2016;13(6):5248–5254. doi: 10.3892/mmr.2016.5201. [DOI] [PubMed] [Google Scholar]
- 35.Liu H, Rose ME, Ma X, Culver S, Dixon CE, Graham SH. In vivo transduction of neurons with TAT-UCH-L1 protects brain against controlled cortical impact injury. PLoS One. 2017;12(5):e0178049. doi: 10.1371/journal.pone.0178049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yao J, Zheng K, Zhang X. Rosiglitazone exerts neuroprotective effects via the suppression of neuronal autophagy and apoptosis in the cortex following traumatic brain injury. Mol Med Rep. 2015;12(5):6591–6597. doi: 10.3892/mmr.2015.4292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang CQ, Ye Y, Chen F, Han WC, Sun JM, Lu X, Guo R, Cao K, Zheng MJ, Liao LC. Posttraumatic administration of a sub-anesthetic dose of ketamine exerts neuroprotection via attenuating inflammation and autophagy. Neuroscience. 2017;343:30–38. doi: 10.1016/j.neuroscience.2016.11.029. [DOI] [PubMed] [Google Scholar]
- 38.Yin Y, Li E, Sun G, Yan HQ, Foley LM, Andrzejczuk LA, Attarwala IY, Hitchens TK, Kiselyov K, Dixon CE, et al. Effects of DHA on Hippocampal Autophagy and Lysosome Function After Traumatic Brain Injury. Mol Neurobiol. 2017 doi: 10.1007/s12035-017-0504-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Cui C, Cui J, Jin F, Cui Y, Li R, Jiang X, Tian Y, Wang K, Jiang P, Gao J. Induction of the Vitamin D Receptor Attenuates Autophagy Dysfunction-Mediated Cell Death Following Traumatic Brain Injury. Cell Physiol Biochem. 2017;42(5):1888–1896. doi: 10.1159/000479571. [DOI] [PubMed] [Google Scholar]
- 40.Hensley K, Poteshkina A, Johnson MF, Eslami P, Gabbita SP, Hristov AM, Venkova-Hristova KM, Harris-White ME. Autophagy Modulation by Lanthionine Ketimine Ethyl Ester Improves Long-Term Outcome after Central Fluid Percussion Injury in the Mouse. J Neurotrauma. 2016;33(16):1501–1513. doi: 10.1089/neu.2015.4196. [DOI] [PubMed] [Google Scholar]
- 41.Zeng XJ, Li P, Ning YL, Zhao Y, Peng Y, Yang N, Zhao ZA, Chen JF, Zhou YG. Impaired autophagic flux is associated with the severity of trauma and the role of A2AR in brain cells after traumatic brain injury. Cell Death Dis. 2018;9(2):252. doi: 10.1038/s41419-018-0316-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Liu X, Tian F, Wang S, Wang F, Xiong L. Astrocyte Autophagy Flux Protects Neurons Against Oxygen-Glucose Deprivation and Ischemic/Reperfusion Injury. Rejuvenation Res. 2017 doi: 10.1089/rej.2017.1999. [DOI] [PubMed] [Google Scholar]
- 43.Kasprowska D, Machnik G, Kost A, Gabryel B. Time-Dependent Changes in Apoptosis Upon Autophagy Inhibition in Astrocytes Exposed to Oxygen and Glucose Deprivation. Cell Mol Neurobiol. 2017;37(2):223–234. doi: 10.1007/s10571-016-0363-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shi R, Weng J, Zhao L, Li XM, Gao TM, Kong J. Excessive autophagy contributes to neuron death in cerebral ischemia. CNS Neurosci Ther. 2012;18(3):250–260. doi: 10.1111/j.1755-5949.2012.00295.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Du L, Hickey RW, Bayir H, Watkins SC, Tyurin VA, Guo F, Kochanek PM, Jenkins LW, Ren J, Gibson G, et al. Starving neurons show sex difference in autophagy. J Biol Chem. 2009;284(4):2383–2396. doi: 10.1074/jbc.M804396200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Carloni S, Buonocore G, Balduini W. Protective role of autophagy in neonatal hypoxia-ischemia induced brain injury. Neurobiol Dis. 2008 doi: 10.1016/j.nbd.2008.07.022. [DOI] [PubMed] [Google Scholar]
- 47.Buckley KM, Hess DL, Sazonova IY, Periyasamy-Thandavan S, Barrett JR, Kirks R, Grace H, Kondrikova G, Johnson MH, Hess DC, et al. Rapamycin up-regulation of autophagy reduces infarct size and improves outcomes in both permanent MCAL, and embolic MCAO, murine models of stroke. Exp Transl Stroke Med. 2014;6:8. doi: 10.1186/2040-7378-6-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhao F, Qu Y, Zhu J, Zhang L, Huang L, Liu H, Li S, Mu D. miR-30d-5p Plays an Important Role in Autophagy and Apoptosis in Developing Rat Brains After Hypoxic-Ischemic Injury. J Neuropathol Exp Neurol. 2017;76(8):709–719. doi: 10.1093/jnen/nlx052. [DOI] [PubMed] [Google Scholar]
- 49.Puyal J, Vaslin A, Mottier V, Clarke PG. Postischemic treatment of neonatal cerebral ischemia should target autophagy. Ann Neurol. 2009 doi: 10.1002/ana.21714. [DOI] [PubMed] [Google Scholar]
- 50.Dong F, Yao R, Yu H, Liu Y. Neuroprotection of Ro25-6981 Against Ischemia/Reperfusion-Induced Brain Injury via Inhibition of Autophagy. Cell Mol Neurobiol. 2017;37(4):743–752. doi: 10.1007/s10571-016-0409-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wang M, Wang J, Liu Z, Guo X, Wang N, Jia N, Zhang Y, Yuan J. Effects of intermedin on autophagy in cerebral ischemia/reperfusion injury. Neuropeptides. 2017 doi: 10.1016/j.npep.2017.10.004. [DOI] [PubMed] [Google Scholar]
- 52.Wang D, Lin Q, Su S, Liu K, Wu Y, Hai J. URB597 improves cognitive impairment induced by chronic cerebral hypoperfusion by inhibiting mTOR-dependent autophagy. Neuroscience. 2017;344:293–304. doi: 10.1016/j.neuroscience.2016.12.034. [DOI] [PubMed] [Google Scholar]
- 53.Koike M, Shibata M, Tadakoshi M, Gotoh K, Komatsu M, Waguri S, Kawahara N, Kuida K, Nagata S, Kominami E, et al. Inhibition of autophagy prevents hippocampal pyramidal neuron death after hypoxic-ischemic injury. Am J Pathol. 2008;172(2):454–469. doi: 10.2353/ajpath.2008.070876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Xie C, Ginet V, Sun Y, Koike M, Zhou K, Li T, Li H, Li Q, Wang X, Uchiyama Y, et al. Neuroprotection by selective neuronal deletion of Atg7 in neonatal brain injury. Autophagy. 2016;12(2):410–423. doi: 10.1080/15548627.2015.1132134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006;441(7095):880–884. doi: 10.1038/nature04723. [DOI] [PubMed] [Google Scholar]
- 56.Au AK, Chen Y, Du L, Smith CM, Manole MD, Baltagi SA, Chu CT, Aneja RK, Bayir H, Kochanek PM, et al. Ischemia-induced autophagy contributes to neurodegeneration in cerebellar Purkinje cells in the developing rat brain and in primary cortical neurons in vitro. Biochim Biophys Acta. 2015;1852(9):1902–1911. doi: 10.1016/j.bbadis.2015.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhu Y, Shui M, Liu X, Hu W, Wang Y. Increased autophagic degradation contributes to the neuroprotection of hydrogen sulfide against cerebral ischemia/reperfusion injury. Metab Brain Dis. 2017;32(5):1449–1458. doi: 10.1007/s11011-017-0014-4. [DOI] [PubMed] [Google Scholar]
- 58.Fang L, Li X, Zhong Y, Yu J, Yu L, Dai H, Yan M. Autophagy protects human brain microvascular endothelial cells against methylglyoxal-induced injuries, reproducible in a cerebral ischemic model in diabetic rats. J Neurochem. 2015;135(2):431–440. doi: 10.1111/jnc.13277. [DOI] [PubMed] [Google Scholar]
- 59.Zhan L, Chen S, Li K, Liang D, Zhu X, Liu L, Lu Z, Sun W, Xu E. Autophagosome maturation mediated by Rab7 contributes to neuroprotection of hypoxic preconditioning against global cerebral ischemia in rats. Cell Death Dis. 2017;8(7):e2949. doi: 10.1038/cddis.2017.330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cui D, Shang H, Zhang X, Jiang W, Jia X. Cardiac arrest triggers hippocampal neuronal death through autophagic and apoptotic pathways. Sci Rep. 2016;6:27642. doi: 10.1038/srep27642. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 61.Zhou T, Liang L, Liang Y, Yu T, Zeng C, Jiang L. Mild hypothermia protects hippocampal neurons against oxygen-glucose deprivation/reperfusion-induced injury by improving lysosomal function and autophagic flux. Exp Cell Res. 2017;358(2):147–160. doi: 10.1016/j.yexcr.2017.06.010. [DOI] [PubMed] [Google Scholar]
- 62.Liu YY, Zhang TY, Xue X, Liu DM, Zhang HT, Yuan LL, Liu YL, Yang HL, Sun SB, Zhang C, et al. Pseudoginsenoside-F11 attenuates cerebral ischemic injury by alleviating autophagic/lysosomal defects. CNS Neurosci Ther. 2017;23(7):567–579. doi: 10.1111/cns.12702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Davis S, Wang J, Ferro-Novick S. Crosstalk between the Secretory and Autophagy Pathways Regulates Autophagosome Formation. Dev Cell. 2017;41(1):23–32. doi: 10.1016/j.devcel.2017.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Farhan H, Kundu M, Ferro-Novick S. The link between autophagy and secretion: a story of multitasking proteins. Mol Biol Cell. 2017;28(9):1161–1164. doi: 10.1091/mbc.E16-11-0762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ponpuak M, Mandell MA, Kimura T, Chauhan S, Cleyrat C, Deretic V. Secretory autophagy. Curr Opin Cell Biol. 2015;35:106–116. doi: 10.1016/j.ceb.2015.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ventruti A, Cuervo AM. Autophagy and neurodegeneration. Curr Neurol Neurosci Rep. 2007;7(5):443–451. doi: 10.1007/s11910-007-0068-5. [DOI] [PubMed] [Google Scholar]
- 67.Ejlerskov P, Rasmussen I, Nielsen TT, Bergstrom AL, Tohyama Y, Jensen PH, Vilhardt F. Tubulin polymerization-promoting protein (TPPP/p25alpha) promotes unconventional secretion of alpha-synuclein through exophagy by impairing autophagosome-lysosome fusion. J Biol Chem. 2013;288(24):17313–17335. doi: 10.1074/jbc.M112.401174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR, Mehrpour M, Christoffersson J, Chaabane W, Moghadam AR, Kashani HH, et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol. 2014;112:24–49. doi: 10.1016/j.pneurobio.2013.10.004. [DOI] [PubMed] [Google Scholar]
- 69.Nixon RA, Wegiel J, Kumar A, Yu WH, Peterhoff C, Cataldo A, Cuervo AM. Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J Neuropathol Exp Neurol. 2005;64(2):113–122. doi: 10.1093/jnen/64.2.113. [DOI] [PubMed] [Google Scholar]
- 70.Wong YC, Holzbaur EL. Autophagosome dynamics in neurodegeneration at a glance. J Cell Sci. 2015;128(7):1259–1267. doi: 10.1242/jcs.161216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Asken BM, Sullan MJ, DeKosky ST, Jaffee MS, Bauer RM. Research Gaps and Controversies in Chronic Traumatic Encephalopathy: A Review. JAMA Neurol. 2017;74(10):1255–1262. doi: 10.1001/jamaneurol.2017.2396. [DOI] [PubMed] [Google Scholar]
- 72.Ikonomovic MD, Uryu K, Abrahamson EE, Ciallella JR, Trojanowski JQ, Lee VM, Clark RS, Marion DW, Wisniewski SR, DeKosky ST. Alzheimer's pathology in human temporal cortex surgically excised after severe brain injury. Exp Neuro. 2004;190(1):192–203. doi: 10.1016/j.expneurol.2004.06.011. [DOI] [PubMed] [Google Scholar]
- 73.Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4(147):147ra111. doi: 10.1126/scitranslmed.3003748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Peng W, Achariyar TM, Li B, Liao Y, Mestre H, Hitomi E, Regan S, Kasper T, Peng S, Ding F, et al. Suppression of glymphatic fluid transport in a mouse model of Alzheimer's disease. Neurobiol Dis. 2016;93:215–225. doi: 10.1016/j.nbd.2016.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Iliff JJ, Chen MJ, Plog BA, Zeppenfeld DM, Soltero M, Yang L, Singh I, Deane R, Nedergaard M. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J Neurosci. 2014;34(49):16180–16193. doi: 10.1523/JNEUROSCI.3020-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Saykally JN, Ratliff WA, Keeley KL, Pick CG, Mervis RF, Citron BA. Repetitive Mild Closed Head Injury Alters Protein Expression and Dendritic Complexity in a Mouse Model. J Neurotrauma. 2018;35(1):139–148. doi: 10.1089/neu.2017.5070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhang Q, Kang R, Zeh HJ, 3rd, Lotze MT, Tang D. DAMPs and autophagy: cellular adaptation to injury and unscheduled cell death. Autophagy. 2013;9(4):451–458. doi: 10.4161/auto.23691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Au AK, Aneja RK, Bell MJ, Bayir H, Feldman K, Adelson PD, Fink EL, Kochanek PM, Clark RS. Cerebrospinal fluid levels of high-mobility group box 1 and cytochrome C predict outcome after pediatric traumatic brain injury. J Neurotrauma. 2012;29(11):2013–2021. doi: 10.1089/neu.2011.2171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Satchell MA, Lai Y, Kochanek PM, Wisniewski SR, Fink EL, Siedberg NA, Berger RP, DeKosky ST, Adelson PD, Clark RS. Cytochrome c, a biomarker of apoptosis, is increased in cerebrospinal fluid from infants with inflicted brain injury from child abuse. J Cereb Blood Flow Metab. 2005;25(7):919–927. doi: 10.1038/sj.jcbfm.9600088. [DOI] [PubMed] [Google Scholar]
- 80.Walko TD, 3rd, Bola RA, Hong JD, Au AK, Bell MJ, Kochanek PM, Clark RS, Aneja RK. Cerebrospinal fluid mitochondrial DNA: a novel DAMP in pediatric traumatic brain injury. Shock. 2014;41(6):499–503. doi: 10.1097/SHK.0000000000000160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tang D, Kang R, Cheh CW, Livesey KM, Liang X, Schapiro NE, Benschop R, Sparvero LJ, Amoscato AA, Tracey KJ, et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene. 2010;29(38):5299–5310. doi: 10.1038/onc.2010.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wang J, Han D, Sun M, Feng J. A Combination of Remote Ischemic Perconditioning and Cerebral Ischemic Postconditioning Inhibits Autophagy to Attenuate Plasma HMGB1 and Induce Neuroprotection Against Stroke in Rat. J Mol Neurosci. 2016;58(4):424–431. doi: 10.1007/s12031-016-0724-9. [DOI] [PubMed] [Google Scholar]
- 83.Liu Y, Levine B. Autosis and autophagic cell death: the dark side of autophagy. Cell Death Differ. 2015;22(3):367–376. doi: 10.1038/cdd.2014.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Laird MD, Shields JS, Sukumari-Ramesh S, Kimbler DE, Fessler RD, Shakir B, Youssef P, Yanasak N, Vender JR, Dhandapani KM. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia. 2014;62(1):26–38. doi: 10.1002/glia.22581. [DOI] [PMC free article] [PubMed] [Google Scholar]


