Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Received: 10 April 2019 | Accepted: 23 August 2019

DOI: 10.1002/jcp.29173

REVIEW ARTICLE

Modulation of autophagy in traumatic brain injury

Zhiqing Zeng1* | Yao Zhang2* | Weiping Jiang1 | Lu He1 | Hongtao Qu1

1
Department of Neurosurgery, First Affiliated
Hospital, University of South China, Abstract
Hengyang, China Traumatic brain injury (TBI) is defined as a traumatically induced structural injury or
2
Department of Clinical Laboratory, The First
physiological disruption of brain function as a result of external forces, leading to
People’s Hospital of Changde City, Changde
City, Hunan Province, China adult disability and death. A growing body of evidence reveals that alterations in
autophagy‐related proteins exist extensively in both experimentally and clinically
Correspondence
Lu He and Hongtao Qu, Department of after TBI. Of note, the autophagy pathway plays an essential role in pathophysio-
Neurosurgery, First Affiliated Hospital,
logical processes, such as oxidative stress, inflammatory response, and apoptosis, thus
University of South China, 421001
Hengyang, China. contributing to neurological properties of TBI. With this in mind, this review
Email: luhe1989@126.com (L. H.) and
summarizes a comprehensive overview on the beneficial and detrimental effects of
qht959801@sina.com (H. Q.)
autophagy in pathophysiological conditions and how these activities are linked to the
Funding information
pathogenesis of TBI. Moreover, the relationship between oxidative stress, inflamma-
Heath and Family Planning Commission
Science Foundation Projects of Hunan tion, apoptosis, and autophagy occur TBI. Ultimately, multiple compounds and various
Province of China, Grant/Award Number:
drugs targeting the autophagy pathway are well described in TBI. Therefore,
C20190121; National Natural Science
Foundation of Hunan, Grant/Award Number: autophagy flux represents a potential clinical therapeutic value for the treatment of
2018JJ6070; Technology and Innovation
TBI and its complications.
Guiding Program of Hunan, Grant/Award
Number: 2017SK50207; National Natural
Science Foundation of China, Grant/Award KEYWORDS
Number: 81803535 apoptosis, autophagy, inflammatory response, oxidative stress, traumatic brain injury

1 | INTRODUCTION patients’ outcomes. TBI triggers a cascade of pathophysiological


actions by secondary brain injury, such as oxidative stress,
Traumatic brain injury (TBI) remains a significant worldwide medical inflammatory response, and apoptosis, resulting in neuronal cell
concern as a considerable cause of death and permanent disability death, brain edema, and neurological deficits. The development of
among adults, imposing public burden in modern society (Langlois, neurological deficits includes learning and memory impairments,
Rutland‐Brown, & Wald, 2006). TBI is recognized as penetrating or brain damage, and motor behavioral functions (Saatman et al., 2008).
blunt trauma and nonaccidental head injury with traumatic etiology. But no effective therapies for patients with TBI in clinical interven-
The pathological process of TBI is very complicated and is generally tion currently exist. Thus, understanding the pathophysiological
divided into two phases, primary brain injury and secondary brain processes after TBI, and identifying potential therapeutic approaches
injury. The primary brain injury is the major factor in determining the are urgently needed and are of great importance.

Abbreviations: 17‐AAG, 17‐Allylamino‐demethoxygeldanamycin; 3‐MA, 3‐Methyladenine; 3‐MST, 3‐Mercaptopyruvate sulfurtransferase; ALI, Acute lung injury; ALS, Amyotrophic lateral
sclerosis; ANDRO, Andrographolide; BBB, Blood–brain barrier; BMSCs, Bone marrow stromal cells; BNIP3L, Bcl‐2/E1B‐19K‐interacting protein 3‐like; CCI, Controlled cortical impact; CL,
Cardiolipin; CQ, Chloroquine; CR, Caloric restriction; CTX, Ceftriaxone; CXs, Connexins; DHA, Docosahexaenoic acid; EPPS, 4‐(2‐Hydroxyethyl)‐1‐piperazinepropanesulphonic acid; ERK,
Extracellular signal‐regulated kinase; FGF2, Fibroblast growth factor‐2; FoxO, Forkhead box; GCEE, c‐Glutamylcysteinyl ethyl ester; H2S, Hydrogen sulfide; HO‐1, heme oxygenase‐1; IL‐33,
Interleukin‐33; JNK, c‐Jun NH2‐terminal kinase; LC3, Microtubule‐associated protein light chain 3; LKE, Lanthionine ketimine ethyl ester; Lut, Luteolin; MB, Methylene blue; Mdivi‐1,
Mitochondrial division inhibitor 1; Melatonin, N‐acetyl‐5‐methoxytryptamine; miRNAs, MicroRNAs; MSCs‐IL‐10, Mesenchymal stem cells overexpressing interleukin10; NEC‐1, Necrostatin‐1;
NLRP3, NOD‐like receptor protein‐3; NR2B, N‐methyl‐d‐aspartate receptor 2B; Nrf2, Nuclear factor‐erythroid2‐like2; P188, Poloxamer 188; P2X7R, P2X7 receptor; p62, SQSTM1/p62; PEA,
Palmitoylethanolamide; PG, Progesterone; PQQ, Pyrroloquinoline quinone; RSG, Rosiglitazone; RV, Resveratrol; STING, Stimulator of interferon genes; TAK‐242, Resatorvid; TAT, Trans‐
activating transduction protein; TBI, Traumatic brain injury; THC, Tetrahydrocurcumin; TLR4, Toll‐like receptor 4; TPEN, Zinc chelator N,N,N’,N’‐tetrakis‐ (2‐pyridylmethyl) ethylenediamine;
UCH‐L1, Ubiquitin carboxy‐terminal hydrolase L1; VPA, Valproic acid; ω‐3 PUFA, Omega‐3 polyunsaturated fatty acids.

*Zhiqing Zeng and Yao Zhang contributed equally to this work.

J Cell Physiol. 2019;1–13. wileyonlinelibrary.com/journal/jcp © 2019 Wiley Periodicals, Inc. | 1


2 | ZENG ET AL.

The term autophagy referred to as “self‐eating,” is an evolutio- Administration of docosahexaenoic acid (DHA) activates autop-
narily conserved process in which cells mediate the degradation of hagic flux, and subsequently improving cognitive function recovery
damaged organelles, protein aggregates, and invading pathogens and reducing brain damage upon TBI (Yin et al., 2018). Short term
through a lysosome‐dependent pathway. The process plays a crucial caloric restriction (CR) prevents cognitive impairment after mild TBI,
role in various biological functions in maintaining cellular home- which is accompanied by increasing autophagy and suppressing
ostasis (Kim & Klionsky, 2000; Klionsky & Emr, 2000). Recently, astrocyte activation (Liu, Wang et al., 2017). Posttreatment with
autophagy pathway takes participate in the pathogenesis of brain Morin improves memory impair via enhanced autophagy upon mild
injury after TBI (Feldmann et al., 2019). In a controlled cortical TBI (El‐Gazar, Soubh, Mohamed, Awad, & El‐Abhar, 2019). NIX3, also
impact (CCI) system model of TBI in vitro and in vivo, autophagy called BNIP3L (Bcl‐2/E1B‐19K‐interacting protein 3‐like), amelio-
markers such as microtubule‐associated protein light chain 3 (LC3)‐II rates brain water content and neurological deficits after TBI via the
and beclin‐1 are significantly increased, sequestosome 1 (SQSTM1/ induction of autophagy (Ma et al., 2019). Mesenchymal stem cells
p62) are remarkably decreased, indicating that autophagic activity is overexpressing interleukin10 (MSCs‐IL‐10) transplantation induces
persistently activated after TBI (Au et al., 2017; Erlich, Shohami, & autophagy to protect against TBI‐induced neuronal damage (Maiti
Pinkas‐Kramarski, 2006; Liu, Chen, Dietrich, & Hu, 2008; Sadasivan, et al., 2019). Taken together, the stimulation of autophagy pathway
Dunn, Hayes, & Wang, 2008; Sebastiani et al., 2017). Conversely, confers pivotal neuroprotective effects following TBI.
autophagy inhibitors 3‐methyladenine (3‐MA) and chloroquine (CQ)
have precisely the opposite effect on TBI (Luo et al., 2011; Zeng et al.,
2.2 | The detrimental effect of autophagy
2018). Here, N‐methyl‐d‐aspartate receptor 2B (NR2B) and polox-
amer 188 (P188) stimulate autophagy following TBI both in vivo and On the contrary, various cellular factors and molecules regulate
in vitro (Bao et al., 2016; Bigford, Alonso, Dietrich, & Keane, 2009). autophagic activity that play a detrimental role in the progression of
The c‐Jun NH2‐terminal kinase (JNK) also provokes neuron TBI. The peptide apelin‐13, an endogenous ligand for the APJ
autophagy via p53 phosphorylation in response to TBI (Hong et al., receptor, is abundantly observed in the nervous systems. For
2012). However, administration of bone marrow stromal cells instance, apelin‐13 significantly mitigates autophagy, and it may
(BMSCs) diminishes autophagy after TBI (Sun et al., 2014). These contribute to ameliorate TBI‐induced brain damage (Bao et al., 2016;
observations imply that the involvement of autophagy contributes to Bao, Zhang, Han, & Dai, 2015). Interestingly, neurons transactivating
the pathophysiological responses following TBI. transduction protein with ubiquitin carboxy‐terminal hydrolase L1
In this review, we mainly focus on the recent discoveries of the (TAT‐UCH‐L1) and Wnt/β‐catenin reduce autophagy to protect brain
beneficial and detrimental roles of autophagy concerning neurologi- injury against CCI, a well documented TBI model (Liu et al., 2017;
cal properties, which may be involved in the pathogenesis of TBI. Zhang et al., 2018). The specific necroptosis inhibitor necrostatin‐1
Furthermore, we summarize the correlation between oxidative (NEC‐1) and p53 antagonist pifithrin‐α (PFT‐α) decrease autophagy
stress, inflammation, apoptosis, and autophagy after TBI. Finally, to attenuate brain damage suffering from TBI (Huang et al., 2018;
compounds and drugs targeting the autophagy pathway are well Wang et al., 2012).
elaborated in the literature. Therefore, autophagy provides a Hydrogen sulfide (H2S), a novel gaseous neuromodulator, has
promising pharmacological intervention for the treatment of TBI been reported to exert neuroprotectant following TBI. Indeed, NaHS
and its complicated management. (a H2S donor) restrains brain edema and improves behavioral
symptoms in TBI via the suppression of autophagy (K. Xu et al.,
2018; Zhang et al., 2014). Administration of H2S‐synthesizing
2 | T H E PR O P E R T I E S O F AU T O P H A G Y
enzyme 3‐mercaptopyruvate sulfurtransferase (3‐MST) reverses
AF TER TBI
these neuroprotective agents by enhancing autophagy after TBI
(Zhang et al., 2017). The zinc chelator N,N,N’,N’‐tetrakis‐(2‐pyridyl-
2.1 | Beneficial role of autophagy
methyl) ethylenediamine (TPEN) and (Gly14)‐humanin inhibit autop-
Accumulating evidence has demonstrated that autophagy, the chief hagy in injured hippocampal neurons, thus improving morphological
machinery pathway for bulk elimination of aberrant cell components, and functional outcomes after TBI (Wang et al., 2013; Zhao, Liu, Ma,
is persistently increased after both experimental and clinical TBI Zhang, & Liang, 2018). Vitamin D active metabolite calcitriol
models (Zhang et al., 2008). Herein, connexins (CXs, also termed gap attenuates neurological deficits in the brain cortex region following
junctions) are membrane proteins with dual‐channel functions that TBI, which is highly associated with suppressing autophagy (Cui et al.,
permit the intercellular exchange of metabolites, ions, and small 2017). Furthermore, knockdown of forkhead box (foxO) 3a improves
molecules (Bennett, Contreras, Bukauskas, & Sáez, 2003). Astrocytic neurological dysfunctions in the hippocampus, via the downregula-
p‐CX43 promotes autophagy in the hippocampal by the activation of tion of autophagic flux (Sun, Zhao et al., 2018).
P2X7 receptor (P2X7R), contributing to TBI‐induced cognitive MicroRNAs (miRNAs) dysfunction are aberrantly expressed in
deficits repair (Sun et al., 2014; Sun et al., 2015). In turn, autophagy the hippocampus after the experimental model of TBI. For
is an important regulator of p‐Cx43 and CX40 degradation in the example, overexpression of miR‐23b prevents cognitive impair-
hippocampal following TBI (Chen et al., 2017; Sun et al., 2015). ments by directly inhibiting ATG12‐mediated autophagic activity
ZENG ET AL. | 3

upon TBI (Sun, Liu et al., 2018). miR‐27a protects against TBI‐ 2008). Of note, tetrahydrocurcumin (THC) has shown to induce
induced brain damage though a mechanism suppressing FoxO3a‐ antioxidant activity in vitro and in vivo. However, administration of
mediated autophagy (Sun et al., 2017). Notably, increased both THC ameliorates cerebral edema, inhibits apoptosis, and improves
miR‐21‐5p and miR‐124‐3p in microglial exosomes attenuate neurological deficits after TBI. The neuroprotection of THC occurs
neuronal autophagy, thus abrogating TBI‐induced brain injury in via initiating autophagy (Gao et al., 2016, 2017). Consistently,
vitro and in vivo (Li, Huang, Yin et al., 2019; Li, Huang, Zhu et al., fucoxanthin with antioxidant properties confers neuroprotective
2019). Therefore, multiple miRNAs take participate in the role in models of TBI, which is closely associated with enhancing
pathophysiological outcomes of TBI via the inhibition of autop- Nrf2‐autophagy in vitro and in vivo (Zhang et al., 2017). Altogether,
hagy. Long noncoding RNA CRNDE (lncRNA CRNDE) silencing oxidative stress contributes to neuropathology following TBI by
reduces autophagic flux to promote the nerve repair following TBI influencing autophagy.
(Yi et al., 2019). Collectively, targeting autophagy by suppressors
serves as a valuable therapeutic strategy for TBI.
4.2 | Autophagy modulates inflammatory response
Several studies have demonstrated that interleukin‐33 (IL‐33)
3 | DUAL ROLES O F M ITOP HAGY improves neurological function and ameliorates cerebral edema in
F O L L O W I N G TB I model of TBI via partly reducing autophagy and neuroinflammation,
revealing that autophagy and inflammatory cascades are an essential
Mitophagy plays a prominent role in the selective degradation of events in brain injury after TBI (Fairlie‐Clarke et al., 2018). However,
damaged mitochondria by autophagy, therefore maintaining mito- the correlation between autophagy and inflammation remains poorly
chondrial quality control (Gustafsson & Dorn, 2019). Some studies understood. Multiple inflammatory factors, such as stimulator of
have demonstrated that dysfunction of mitophagy is closely interferon genes (STING; Nazmi et al., 2019), toll‐like receptor 4
associated with the pathophysiology of TBI (Chen et al., 2016). (TLR4; Zhou, Andonegui, Wong, & Kubes, 2009), and so forth, are
Recently, it has been observed that the reduction of mitochondrial known to be critical mediators of the inflammatory reaction in the
fission Drp1 with mitochondrial division inhibitor 1 (mdivi‐1) central nervous systems. Accordingly, STING mediates TBI‐induced
prevents TBI‐triggered blood–brain barrier (BBB) and cell death in neuroinflammation, thus contributing to autophagy dysfunction
vitro via blocking mitophagy (Wu et al., 2018). However, Niu, Dong, (Abdullah et al., 2018). Genetic ablation of TLR4 delays neuroin-
Xu, Zhang, and Liu (2019) strongly argue that mdivi‐1 represses flammatory response and subsequently aggravates brain damage
mitophagy to aggravate neurological manifestations in a rat model of after TBI by inhibiting autophagy (Jiang et al., 2018). Simultaneously,
mild TBI. Therefore, it is remain unclear that how mitophagy affects TLR4 antagonist resatorvid (TAK‐242) diminishes neuronal autop-
the pathological process of TBI. Of note, melatonin (N‐acetyl‐5‐ hagy in the hippocampus upon TBI by MyD88/TRIF‐NF‐κB signaling
methoxytryptamine) and cardiolipin (CL) decrease neuronal cell (Feng, Gao et al., 2017). In particularly, rosiglitazone (RSG) and
death and alleviate behavioral deficits following TBI via enhancing ketamine improve the behavioral and histopathological outcomes
mitophagy, and these neuroprotecive effects abolish by inhibiting after TBI, carrying out these anti‐inflammatory properties via the
mitophagy with 3‐MA (Chao et al., 2019; Lin et al., 2016). In vivo CCI‐ downregulation of autophagy (Wang et al., 2017; Yao, Zheng, &
induced TBI model, mitophagy induced by extracellular signal‐ Zhang, 2015). However, the antiepileptic drug valproic acid (VPA)
regulated kinase (ERK)/nuclear factor‐erythroid2‐like2 (Nrf2)/heme inhibits TBI‐induced inflammatory response in vivo due to increased
oxygenase‐1 (HO‐1) diminishes intestinal mucosal damage and Nrf2/ARE‐mediated autophagy (Chen, Wang et al., 2018). These
epithelial barrier dysfunction, thus leading to TBI‐evoked gastro- finding indicate that inhibition of inflammatory process acts as a
intestinal diseases (Liu, Bao et al., 2017). Taken together, mitophagy useful tool for TBI via the modulation of autophagy.
plays a dual role in pathological condition of TBI and its complication.

4.3 | The dual role of autophagy on apoptosis


4 | RELATIONSHIP BE TWEE N O XIDATIVE
It is widely known that various forms of cell death, especially in
S T R E S S , I N F L A M M A T IO N , AP O P T O S I S , A N D
apoptosis, occurring following TBI is modulated through autophagy
AUTOPH AGY
signaling pathway (Jin, Wang, Yang, Zhang, & Dai, 2017; Sarkar et al.,
2014). Here, moderate hypothermia reduces microglial activation by
4.1 | Oxidative stress regulates autophagy
promoting apoptosis and inhibiting autophagy, indicating the
Currently, oxidative stress and autophagy are believed to play an connecting between autophagy and apoptosis in TBI (Zhang et al.,
essential role in brain injury after TBI. To determine the autophagic 2018). More importantly, moderate hypothermia significantly alle-
activity whether it can be regulated in response to oxidative stress, viates cell death both in ipsilateral hippocampus and in the injured
treatment of antioxidant c‐glutamylcysteinyl ethyl ester (GCEE) cortex after fluid‐percussion TBI through the activation of autophagy
partially blocks autophagy, thereby improving neurologic and (Jin et al., 2015a, 2015b). The suppression of autophagy with 3‐MA
behavioral outcome following TBI (Clark et al., 2008; Lai et al., deteriorates long‐term behavioral outcome upon post‐TBI moderate
4
|

T A B L E 1 Roles of autophagy following TBI


Molecules TBI models Pathways Properties Ref.
NR2B Fluid‐percussion injury Promotes autophagy ? Chen et al. (2017)
P188 Weight‐drop, PC‐12 Promotes autophagy ? Chen et al. (2018)
JNK Marmarou’s weight‐drop Promotes autophagy ? Chen et al. (2018)
BMSCs Weight‐drop device Inhibits autophagy ? Chen et al. (2019)
CX43 Marmarou’s weight‐drop Promotes autophagy via P27R Improves cognitive deficits Cui et al. (2017); Cui et al. (2014)
Autophagy CCI, marmarou’s weight‐drop CX40 and CX43 degradation ? Cui et al. (2015); Ding et al. (2015)
DHA CCI Promotes autophagy flux Improves cognitive functions and reduces brain damage Du et al. (2016)
CR Marmarou’s weight‐drop Promotes autophagy Improves cognitive function Du et al. (2018)
Morin Marmarou’s weight‐drop Promotes autophagy Improves impair memory El‐Gazar et al. (2019)
NIX3/BNIP3L Weight‐drop device Promotes autophagy Ameliorates brain water content and neurological deficits Erlich et al. (2007)
MSCs‐IL‐10 CCI Promotes autophagy Protect against TBI‐induced neuronal damage Erlich et al. (2006)
Apelin‐13 Marmarou’s weight‐drop Inhibits autophagy Ameliorates brain edema and apoptosis Fairlie‐Clarke et al. (2018); Fang et al. (2019)
TAT‐UCH‐L1 CCI Inhibits autophagy Protect against brain injury Feldmann et al. (2019)
Wnt3a CCI Inhibits autophagy Improves functional recovery Feng et al. (2017)
NEC‐1 Weight‐drop model Inhibits autophagy Attenuates brain damage Feng et al. (2016)
PFT‐α CCI Inhibits autophagy Attenuates brain damage Feng et al. (2016)
H2S Marmarou’s weight‐drop Inhibits autophagy Mitigates brain edema and improves behavioral symptoms Feng et al. (2017); Gao et al. (2016)
3‐MST CCI Promotes autophagy ? Gao et al. (2017)
TPEN CCI Inhibits autophagy Improves morphological and functional outcomes Gustafsson and Dorn (2019)
[Gly14]‐humanin CCI Inhibits autophagy Improves morphological and functional outcomes He et al. (2018)
Calcitriol CCI Inhibits autophagic activity Attenuates neurological deficits Hensley et al. (2016)
FoxO3a Marmarou’s weight‐drop Promotes autophagy Aggravates neurobehavioral deficits Hong et al. (2012)
MiR‐23b Marmarou’s weight‐drop Inhibits autophagic activity Prevents cognitive impairments Huang et al. (2018)
MiR‐27a Marmarou’s weight‐drop Inhibits autophagy Ameliorates brain damage Jiang et al. (2018)
MiR‐21‐5p CCI Inhibits rab11a‐mediated autophagy Attenuates brain injury Jin et al. (2016)
MiR‐124‐3p Marmarou’s weight‐drop Inhibits autophagy via CX43 Protect against brain injury Jin et al. (2015a)
lncR‐CRNDE CCI Promotes autophagy Nerve damage Jin et al. (2015b)
(Continues)
ZENG
ET AL.
ZENG ET AL. | 5

Abbreviations: BBB, blood–brain barrier; BMSCs, bone marrow stromal cells; CX43, connexin43; CR, caloric restriction; DHA, docosahexaenoic acid; FoxO3, forkhead box 3; H2S, hydrogen sulfide; MSCs‐IL‐
10, mesenchymal stem cells overexpressing interleukin10; miR, microRNA; 3‐MST, 3‐mercaptopyruvate sulfurtransferase; Mdivi‐1, mitochondrial division inhibitor 1; NR2B, N‐methyl‐d‐aspartate receptor 2B;
hypothermia, as a result of increased apoptosis (Jin, Lei, Lin, Gao, &

NEC‐1, necrostatin‐1; NIX3/BNIP3L, Bcl‐2/E1B‐19K‐interacting protein 3‐like; P188,poloxamer 188; PFT‐α, pifithrin‐α; TAT, transactivating transduction protein; TBI, traumatic brain injury; TPEN,zinc
Jiang, 2016). Poly‐arginine R18 attenuates apoptosis and activates

Kim and Klionsky (2000); Klionsky and


neurocyte cell growth by promoting autophagy in TBI (Batulu et al.,
2019). Melatonin and omega‐3 polyunsaturated fatty acids (ω‐3
PUFA) promote autophagy to inhibit neuronal apoptotic pathway,
thus protecting brain damage after TBI (Chen, Pan et al., 2018; Ding
Langlois et al. (2006) et al., 2015). These findings suggesting the neuroprotective role of
autophagy in apoptosis after TBI.
Lai et al. (2008)

However, growing evidence has revealed that autophagic flux is


Emr (2000)

a detrimental effect of apoptosis after TBI. Here, FTY720 (also


termed as fingolimod) decreases TBI‐induced apoptosis though
Ref.

enhancement of autophagy (Zhang, Ding, Wang, Wu, & Xu, 2016).


The neurotrophic factor fibroblast growth factor‐2 (FGF2) attenu-
Ameliorates neuronal death and improves behavioral deficits

ates apoptosis, and thus improving functional recovery in the early


Attenuates BBB or aggravate neurological manifestations

stage of mild TBI through the inhibition of autophagy. Treatment


with autophagy agonist rapamycin abolishes these protective
effects of FGF2 (Tang et al., 2017). The selective p‐eIF2α
phosphatase inhibitor salubrinal improves TBI‐caused neurological
outcome, which is linked with the downregulation of apoptosis via
suppressing autophagy (Wang et al., 2019). Postconditioning of
sevoflurane delays neuronal apoptosis after TBI by activating PI3K/
AKT‐mediated autophagy (He et al., 2018). Recently, PLA2G4A‐
Ameliorates inflammation

mediated lysosomal membrane damage is involved in apoptosis by


autophagy inhibition following CCI‐induced TBI (Sarkar et al., 2019).
chelator N,N,N’,N’‐tetrakis‐ (2‐pyridylmethyl) ethylenediamine; UCH‐L1, ubiquitin carboxy‐terminal hydrolase L1.

Taken together, apoptosis induced by autophagy pathway con-


tributes to the pathogenesis of TBI.
Properties

5 | A U T O P HA G Y I N T B I‐I N D U C E D
Inhibits autophagy and mitophagy

COMPL I CATION S

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegen-


Promotes autophagy
Promotes autophagy

erative disease characterized by upper and lower motor neuron cell


death. It has been reported that TBI exacerbates ALS‐related
TDP43 mutation in cortical neurons following TBI, indicating TBI is
Pathways

considered as a predisposing factor for the pathology of ALS


(Wiesner et al., 2018). Interestingly, traumatic injury causes stress
granule formation, thus leading to motor dysfunctions in ALS
(Anderson et al., 2018). In G93A‐SOD1 transgenic mice ALS model,
the female sex hormone progesterone (PG) promotes autophagy
flux in the spinal cord to protect against motoneuronal death.
Moreover, autophagy inhibitor 3‐MA reverses these PG effects,
revealing that PG delays moto neuron degeneration in ALS
TBI models

progression via initiating autophagy in vitro and in vivo (Kim, Kim,


Cho, Kim, & Koh, 2013). In addition, TBI‐induced enhancement of
CCI

CCI
CCI

autophagy via ERK1/2/mTOR/Stat3 pathway mitigates apoptosis,


(Continued)

inflammation, and oxidative stress in lung tissues, improving


pulmonary barrier function, oxygenation function, and static
compliance in acute lung injury (ALI), whereas the interruption of
Cardiolipin
Molecules

Melatonin
TABLE 1

autophagy aggravates these protective effects (Xu et al., 2018),


Mdivi‐1

indicating that ERK1/2/mTOR/Stat3‐mediated autophagy may be


valuable in the treatment of TBI‐triggered ALI. Taken together,
6
|

T A B L E 2 Summary about compounds and drugs targeting the autophagy pathway following TBI

Forms Compounds/drugs TBI models Mechanisms Functions Ref.


Diterpenoid lactone ANDRO Hypoxia‐injured Promotes autophagy Protects against hypoxia injury Wu et al. (2018)
astrocytes
Isaria sinclairii metabolite FTY720 Weight‐drop Increases autophagy Reduces brain edema, improves Sun et al. (2014)
neurobehavioral function
Fatty acid metabolite ω‐3 PUFA Feeney Increases SIRT1‐mediated Attenuates neuronal apoptosis Sun et al. (2015)
deacetylation of autophagy
Flavonoids Luteolin Mamadou’s weight‐drop Enhances autophagy Reduces neuronal degeneration, alleviates Xu et al. (2014)
brain edema, and BBB disruption
Flavonoids Baicalin Weight‐drop Enhances autophagy Improves motor functions, reduces cerebral Xu et al. (2018)
edema, and BBB disruption
Flavonoids Quercetin Marmarou's weight‐drop Inhibits autophagy Attenuates neurological impairment and Xu et al. (2018)
improves cognitive function
Picrohiza kurroa extractive Apocynin CCI Suppresses autophagy Ameliorates neurobehavioral deficits, brain Yao et al. (2015)
edema, and neuronal damage
Polyphenols RV Astrocyte and CCI Suppresses autophagy Improves cognitive function, neurological Ye et al. (2017); Yi et al. (2019);
impairment, and brain edema Yin et al. (2018); Zeng et al.
(2018)
Oxidoreductase coenzyme PQQ Astrocyte and Feeney Inhibits autophagy Improves brain function and enhances cell Zhang et al. (2018); Zhang et al.
viability (2018)
Sulfur amino acid metabolite LKE Central fluid‐percussion Reduces autophagy Improves long‐term behavioral outcome and Zhang et al. (2016)
neuropathology
Flavonoids Co‐ultraPEALut CCI Reduces autophagy Improve neurobehavioral functions and Zhang et al. (2017)
neuropathology
Buffering agent EPPS CCI Ameliorates autophagic flux Improves motor, cognitive functions, and Zhang et al. (2017)
reduces axonal injury
Anticancer 17‐AAG CCI Promotes autophagy Attenuates brain edema, decreases neuronal Zhang et al. (2014)
death, and improves motor function
Antidote MB CCI Promotes autophagy Attenuates brain edema, improves Zhang et al. (2017)
neurological deficit, and neuronal death
Volatile anesthetics Sevoflurane Feeney Promotes PI3K/Akt‐mediated Attenuates neuronal apoptosis and alleviates Wang et al. (2017)
autophagy brain edema
Epilepsy VPA Feeney Promotes Nrf2/ARE‐mediated Promote functional recovery Ma et al. (2019)
autophagy
Macrolide antibiotics Rapamycin Weight‐drop Promotes autophagy and mitophagy Improves cognitive functions Zhang et al. (2008); Zhang et al.
via repressing NLRP3 inflammasome (2019); Zhao et al. (2018)
β‐Lactam antibiotics CTX Mamadou’s weight‐drop Suppresses autophagy Reduces brain edema and improves cognitive Zhao et al. (2016)
function deficits
ZENG

(Continues)
ET AL.
ZENG ET AL. | 7

nesulphonic acid; LKE, lanthionine ketimine ethyl ester; MB, methylene blue; PQQ, pyrroloquinoline quinone; RSG, rosiglitazone; RV, resveratrol; ω‐3 PUFA, omega‐3 polyunsaturated fatty acids; VPA, valproic
autophagy pathway may represent a novel therapeutic strategy for

Abbreviations: 17‐AAG, 17‐allylamino‐demethoxygeldanamycin; ANDRO, andrographolide; CCI, controlled cortical impact; CTX, ceftriaxone; CQ, chloroquine; EPPS, 4‐(2‐hydroxyethyl)‐1‐piperazinepropa-
the treatment of TBI‐induced complications.

Nazmi et al. (2019)


Zhou et al. (2009)

6 | CO MPOU NDS TA RGET THE

Lin et al. (2014)


AUTO PHA GY P ATH WAY A FTER TBI

To date, various compounds have been used to enhance or weaken


Ref.

autophagic activity following TBI. Accordingly, diterpenoid lactone


andrographolide (ANDRO) is isolated from Andrographis paniculata
that protects against hypoxia‐injured astrocytes by promoting
Attenuates brain edema and improves

autophagy (Du et al., 2018). The synthetic compound FTY720 is also


derived from metabolite modification of Isaria sinclairii. The neuro-
protective role of FTY720 is accompanied by the induction of
Promotes functional recovery
Alleviates functional deficit

autophagy (Zhang et al., 2016). Flavonoids such as baicalin, luteolin


(Lut), and quercetin are ubiquitously exist in various fruits and
neurological function

vegetable. Postinjury treatment with Lut and baicalin protect against


brain damage following TBI via enhanced autophagy (Fang et al.,
2019; Xu et al., 2014).
Functions

Nevertheless, the neuroprotection of quercetin in the hippo-


campus following TBI is closely associated with the suppression of
autophagy via activating PI3K/Akt pathway (Du et al., 2016).
Suppresses autophagy via PI3K/Akt/

Apocynin, a natural organic compound derived from the root


extract of Picrohiza kurroa, alleviates neurobehavioral deficits,
reduces cerebral edema, and BBB disruption by the inactivation
of autophagy pathway (Feng, Cui et al., 2017). The natural
Suppresses autophagy

Suppresses autophagy

polyphenolic compound resveratrol (3,5,4′‑trihydroxystilbene, RV)


mTOR pathway

improves cognitive function and neurological impairment as well as


Mechanisms

attenuates brain edema occurring TBI by suppressing autophagy


(Feng, Cui, Gao, Li, Jiang et al., 2016; Lin, Chen, Yang, & Shih, 2014).
Strikingly, RV reduces neuronal autophagy to exert neuroprotective
agent via the downregulation of TLR4/NF‐κB and the upregulation
of Sirt1 (Feng, Cui, Gao, Li, Li et al., 2016; Zhang, Li, Xu, & Li, 2019).
Modified weight drop

More importantly, pyrroloquinoline quinone (PQQ) is an anionic,


water‐soluble compound that provides neuroprotection following
TBI, this effect is linked to the suppression of autophagy (Ye, Zhang,
TBI models

Qian, Yin, & Yan, 2017; Zhang et al., 2017). Lanthionine ketimine
ethyl ester (LKE) and palmitoylethanolamide with luteolin (Co‐
CCI
CCI

ultraPEALut) compounds improve neurobehavioral functions and


Compounds/drugs

neuropathology in central fluid‐percussion injury after TBI accom-


panied by reducing autophagic flux (Cordaro et al., 2016; Hensley
et al., 2016). The buffering compound 4‐(2‐hydroxyethyl)‐1‐piper-
Ketamine

azinepropanesulphonic acid (EPPS) is significantly associated with


RSG
CQ

ameliorated autophagy, ultimately improving the cortex‐dependent


motor and hippocampal‐dependent cognitive deficits after TBI
(Anthony Jalin, Jin, Wang, & Li, 2019).
Multiple attractive potential drugs targeting autophagy pathway
(Continued)

after TBI have been well described in the documents. 17‐Allylamino‐


demethoxygeldanamycin (17‐AAG) has long been in the treatment of
PPAR‐γ agonist
Antimalarial

cancer in clinical trials. Intriguingly, the neuroprotective action of 17‐


Anesthetics
TABLE 2

AAG is closely linked with enhanced autophagy, as the consequence


Forms

of attenuating brain edema, decreasing neuronal death as well as


acid.

improving motor function following TBI (Ma et al., 2015). The


8 | ZENG ET AL.

F I G U R E 1 The role of autophagy in TBI


autophagy plays an essential role in
pathophysiological processes, such as
oxidative stress, inflammatory response,
and apoptosis, thus contributing to
neurological properties of TBI. The
development of neurological functions
includes neuronal cell death, brain edema
and learning and memory impairments,
brain damage, and motor behavioral
function. Therefore, autophagy pathway
represents a potential clinical therapeutic
value for treatment of TBI. TBI, traumatic
brain injury [Color figure can be viewed at
wileyonlinelibrary.com]

inhibitory effect of antidote methylene blue (MB) and volatile suppression of autophagy occurring TBI (Cui et al., 2014; Cui et al.,
anesthetics sevoflurane on brain edema exhibits closely related to 2015). In a rat model of TBI, ketamine is able to alleviate functional
the activation of PI3K/AKT‐mediated autophagy upon TBI (He et al., deficits though the reduction of autophagy (Wang et al., 2017).
2018; Zhao, Liang, Xu, Yan, & Zhang, 2016). The antiepileptic drug Consistently, RSG treatment reduces neuronal autophagy to promote
VPA attenuates TBI‐induced brain injury due to increased Nrf2/ARE‐ functional recovery after TBI (Yao et al., 2015). These findings
mediated autophagy after TBI (Chen, Wang et al., 2018). Rapamycin suggest that targeting autophagy pathway might provide a novel
is a macrolide antibiotic first developed as an antifungal agent, dues clinical efficacy on TBI.
to its anti‐immunosuppressive properties. Currently, accumulating
evidence has indicated that rapamycin aggravates both autophagy
and mitophagy following TBI by repressing NOD‐like receptor 7 | CO NCL UDING REMA RKS AND FUT URE
protein‐3 (NLRP3) inflammasome, leading to neuroprotection (Chen DI R ECT ION S
et al., 2019; Erlich, Alexandrovich, Shohami, & Pinkas‐Kramarski,
2007; Wang et al., 2017). However, the β‐lactam antibiotics In summary, we mainly focus on the key contribution of autophagy and
ceftriaxone (CTX) and antimalarial drug chloroquine (CQ) curb TBI‐ its properties in the pathogenesis of TBI. Accumulating evidence has
induced brain edema and improve cognitive function deficits by the revealed that autophagy, a very well‐characterized cellular degradation
ZENG ET AL. | 9

or recycling process, has been implicated in human and animal models ORCI D
of TBI. Multiple cellular factors and molecules are considered to confer
Zhiqing Zeng http://orcid.org/0000-0002-5374-608X
beneficial and detrimental effect concerning autophagy, thus contribut-
Lu He http://orcid.org/0000-0002-7367-2452
ing to neurological functions of TBI (Table 1). Moreover, the correlation
between inflammation, oxidative stress, apoptosis, and autophagy are
completely described in TBI in vitro and in vivo. Intriguingly, various R E F E R E N CE S
compounds and multiple drugs target autophagic activity, as a
Abdullah, A., Zhang, M., Frugier, T., Bedoui, S., Taylor, J. M., & Crack, P. J.
consequence of TBI progression (Table 2). Taken together, these
(2018). STING‐mediated type‐I interferons contribute to the neuroin-
findings support the viewpoint that autophagy pathway is of great flammatory process and detrimental effects following traumatic brain
importance in TBI and the modulation of this pathway provides injury. Journal of Neuroinflammation, 15(1), 323. https://doi.org/10.
pharmacological approach for TBI and its complications (Figure 1). 1186/s12974‐018‐1354‐7
From this point of view, knowledge gaps remains to be elucidated Anderson, E. N., Gochenaur, L., Singh, A., Grant, R., Patel, K., Watkins, S., …
Pandey, U. B. (2018). Traumatic injury induces stress granule formation
detail mechanisms that cellular factors and contents determine
and enhances motor dysfunctions in ALS/FTD models. Human Molecular
promoting or inhibiting autophagy pathway, ultimately contribute to Genetics, 27(8), 1366–1381. https://doi.org/10.1093/hmg/ddy047
the progression of TBI. Among these, the connection between Anthony Jalin, A. M. A., Jin, R., Wang, M., & Li, G. (2019). EPPS treatment
oxidative stress, inflammation, apoptosis, and autophagy following attenuates traumatic brain injury in mice by reducing Aβ burden and
ameliorating neuronal autophagic flux. Experimental Neurology, 314,
TBI need to be further investigated. It is worthy of comprehensive
20–33. https://doi.org/10.1016/j.expneurol.2019.01.002
understanding of the potential role of mitophagy, to lead to the Au, A. K., Aneja, R. K., Bayır, H., Bell, M. J., Janesko‐Feldman, K., Kochanek, P.
manifestation of TBI. Much of existing literature about compounds M., & Clark, R. S. B. (2017). Autophagy biomarkers Beclin 1 and p62 are
and drugs directly and indirectly affecting autophagy pathway on increased in cerebrospinal fluid after traumatic brain injury. Neurocritical
Care, 26(3), 348–355. https://doi.org/10.1007/s12028‐016‐0351‐x
accounts of cell experiments in vitro and CCI‐triggered TBI in vivo. In
Bao, H., Qiu, H., Kuai, J., Song, C., Wang, S., Wang, C., … Wu, Y. (2016).
further, researchers should pay close attention in various animals and Apelin‐13 as a novel target for intervention in secondary injury after
even human experiment in clinical in vivo. The effects of agonists and traumatic brain injury. Neural Regeneration Research, 11(7), 1128–1133.
antagonists modulate the autophagy pathway upon TBI remain https://doi.org/10.4103/1673‐5374.187049
Bao, H., Yang, X., Zhuang, Y., Huang, Y., Wang, T., Zhang, M., … Tao, L.
elusive and need to be further explored. The optimization of
(2016). The effects of poloxamer 188 on the autophagy induced by
pharmacological inhibitors or the development of specific drugs to traumatic brain injury. Neuroscience Letters, 634, 7–12. https://doi.org/
help design of agent with more specificity and less side effects than 10.1016/j.neulet.2016.09.052
those currently in clinical. Therefore, targeting autophagy could be a Bao, H. J., Zhang, L., Han, W. C., & Dai, D. K. (2015). Apelin‐13 attenuates
traumatic brain injury‐induced damage by suppressing autophagy.
potential approach for TBI and its complications.
Neurochemical Research, 40(1), 89–97. https://doi.org/10.1007/
s11064‐014‐1469‐x
Batulu, H., Du, G. J., Li, D. Z., Sailike, D., Fan, Y. H., & Geng, D. (2019).
A C K N O W L E D GM E N T
Effect of poly‐arginine R18 on neurocyte cell growth via autophagy in
This study was supported by the National Natural Science Founda- traumatic brain injury. Experimental and Therapeutic Medicine, 17(5),
4109–4115. https://doi.org/10.3892/etm.2019.7423
tion of China (81803535), the National Natural Science Foundation
Bennett, M. V. L., Contreras, J. E., Bukauskas, F. F., & Sáez, J. C. (2003).
of Hunan (2018JJ6070), the Technology and Innovation Guiding New roles for astrocytes: Gap junction hemichannels have something
Program of Hunan (2017SK50207), and the Heath and Family to communicate. Trends in Neurosciences, 26(11), 610–617. https://doi.
Planning Commission Science Foundation Projects of Hunan Pro- org/10.1016/j.tins.2003.09.008
Bigford, G. E., Alonso, O. F., Dietrich, W. D., & Keane, R. W. (2009). A novel
vince of China (C20190121).
protein complex in membrane rafts linking the NR2B glutamate
receptor and autophagy is disrupted following traumatic brain injury.
Journal of Neurotrauma, 26(5), 703–720. https://doi.org/10.1089/neu.
CO NFLICT OF I NTERE STS 2008.0783
Chao, H., Lin, C., Zuo, Q., Liu, Y., Xiao, M., Xu, X., … Ji, J. (2019). Cardiolipin‐
The authors declare that there are no conflict of interests.
dependent mitophagy guides outcome after traumatic brain injury.
Journal of Neuroscience, 39(10), 1930–1943. https://doi.org/10.1523/
jneurosci.3415‐17.2018
A UT HO R C ONT RI BU TIO NS
Chen, H., Chan, Y. L., Nguyen, L. T., Mao, Y., de Rosa, A., Beh, I. T., … Gorrie,
C. (2016). Moderate traumatic brain injury is linked to acute
L. H. and H. Q. conceived the manuscript. Z. Z. and Y. Z. consulted the
behaviour deficits and long term mitochondrial alterations. Clinical
literature. Z. Z. and Y. Z. taken participate in writing the initial draft and Experimental Pharmacology and Physiology, 43(11), 1107–1114.
of the manuscript. L. H. and H. Q. revised the whole manuscript. All https://doi.org/10.1111/1440‐1681.12650
authors reviewed the manuscript. Chen, W., Guo, Y., Yang, W., Zheng, P., Zeng, J., & Tong, W. (2017).
Involvement of autophagy in connexin 40 reduction in the late phase
of traumatic brain injury in rats. Brain Research Bulletin, 131, 100–106.
D A T A A V A I L A B I LI TY S TA T E ME N T https://doi.org/10.1016/j.brainresbull.2017.03.014
Chen, X., Pan, Z., Fang, Z., Lin, W., Wu, S., Yang, F., … Li, S. (2018). Omega‐3
Research data not shared. polyunsaturated fatty acid attenuates traumatic brain injury‐induced
10 | ZENG ET AL.

neuronal apoptosis by inducing autophagy through the upregulation Fang, J., Zhu, Y., Wang, H., Cao, B., Fei, M., Niu, W., … Zhou, M. (2019).
of SIRT1‐mediated deacetylation of Beclin‐1. Journal of Neuroinflam- Baicalin protects mice brain from apoptosis in traumatic brain injury
mation, 15(1), 310. https://doi.org/10.1186/s12974‐018‐1345‐8 model through activation of autophagy. Frontiers in Neuroscience, 12,
Chen, X., Wang, H., Zhou, M., Li, X., Fang, Z., Gao, H., … Hu, W. (2018). 1006. https://doi.org/10.3389/fnins.2018.01006
Valproic acid attenuates traumatic brain injury‐induced inflammation in Feldmann, L. K., Le Prieult, P., Felzen, V., Thal, S. C., Engelhard, K., Behl, C., &
vivo: Involvement of autophagy and the Nrf2/ARE signaling pathway. Mittmann, T. (2019). Proteasome and autophagy‐mediated impairment
Frontiers in Molecular Neuroscience, 11, 117. https://doi.org/10.3389/ of late long‐term potentiation (l‐LTP) after traumatic brain injury in the
fnmol.2018.00117 somatosensory cortex of mice. International Journal of Molecular
Chen, Y., Meng, J., Xu, Q., Long, T., Bi, F., Chang, C., & Liu, W. (2019). Sciences, 20(12), 3048. https://doi.org/10.3390/ijms20123048
Rapamycin improves the neuroprotection effect of inhibition of Feng, Y., Cui, C., Liu, X., Wu, Q., Hu, F., Zhang, H., … Wang, L. (2017).
NLRP3 inflammasome activation after TBI. Brain Research, 1710, Protective role of apocynin via suppression of neuronal autophagy
163–172. https://doi.org/10.1016/j.brainres.2019.01.005 and TLR4/NF‐κB signaling pathway in a rat model of traumatic brain
Clark, R. S. B., Bayir, H., Chu, C. T., Alber, S. M., Kochanek, P. M., & injury. Neurochemical Research, 42(11), 3296–3309. https://doi.org/10.
Watkins, S. C. (2008). Autophagy is increased in mice after traumatic 1007/s11064‐017‐2372‐z
brain injury and is detectable in human brain after trauma and critical Feng, Y., Cui, Y., Gao, J. L., Li, M. H., Li, R., Jiang, X. H., … CUI, J. Z. (2016).
illness. Autophagy, 4(1), 88–90. Resveratrol attenuates neuronal autophagy and inflammatory injury
Cordaro, M., Impellizzeri, D., Paterniti, I., Bruschetta, G., Siracusa, R., De by inhibiting the TLR4/NF‐κB signaling pathway in experimental
Stefano, D., … Esposito, E. (2016). Neuroprotective effects of co‐ traumatic brain injury. International Journal of Molecular Medicine,
UltraPEALut on secondary inflammatory process and autophagy 37(4), 921–930. https://doi.org/10.3892/ijmm.2016.2495
involved in traumatic brain injury. Journal of Neurotrauma, 33(1), Feng, Y., Cui, Y., Gao, J. L., Li, R., Jiang, X. H., Tian, Y. X., … CUI, J. Z. (2016).
132–146. https://doi.org/10.1089/neu.2014.3460 Neuroprotective effects of resveratrol against traumatic brain injury
Cui, C., Cui, J., Jin, F., Cui, Y., Li, R., Jiang, X., … Gao, J. (2017). Induction of in rats: Involvement of synaptic proteins and neuronal autophagy.
the vitamin D receptor attenuates autophagy dysfunction‐mediated Molecular Medicine Reports, 13(6), 5248–5254. https://doi.org/10.
cell death following traumatic brain injury. Cellular Physiology and 3892/mmr.2016.5201
Biochemistry, 42(5), 1888–1896. https://doi.org/10.1159/000479571 Feng, Y., Gao, J., Cui, Y., Li, M., Li, R., Cui, C., & Cui, J. (2017).
Cui, C., Cui, Y., Gao, J., Sun, L., Wang, Y., Wang, K., … Cui, J. (2014). Neuroprotective effects of resatorvid against traumatic brain injury
Neuroprotective effect of ceftriaxone in a rat model of traumatic brain in rat: Involvement of neuronal autophagy and TLR4 signaling
injury. Neurological Science, 35(5), 695–700. https://doi.org/10.1007/ pathway. Cellular and Molecular Neurobiology, 37(1), 155–168.
s10072‐013‐1585‐4 https://doi.org/10.1007/s10571‐016‐0356‐1
Cui, C. M., Gao, J. L., Cui, Y., Sun, L. Q., Wang, Y. C., Wang, K. J., … CUI, J. Z. Gao, Y., Li, J., Wu, L., Zhou, C., Wang, Q., Li, X., … Wang, H. (2016).
(2015). Chloroquine exerts neuroprotection following traumatic brain Tetrahydrocurcumin provides neuroprotection in rats after traumatic
injury via suppression of inflammation and neuronal autophagic death. brain injury: Autophagy and the PI3K/AKT pathways as a potential
Molecular Medicine Reports, 12(2), 2323–2328. https://doi.org/10. mechanism. Journal of Surgical Research, 206(1), 67–76. https://doi.org/
3892/mmr.2015.3611 10.1016/j.jss.2016.07.014
Ding, K., Xu, J., Wang, H., Zhang, L., Wu, Y., & Li, T. (2015). Melatonin Gao, Y., Zhuang, Z., Gao, S., Li, X., Zhang, Z., Ye, Z., & Li, J. (2017).
protects the brain from apoptosis by enhancement of autophagy after Tetrahydrocurcumin reduces oxidative stress‐induced apoptosis via
traumatic brain injury in mice. Neurochemistry International, 91, 46–54. the mitochondrial apoptotic pathway by modulating autophagy in rats
https://doi.org/10.1016/j.neuint.2015.10.008 after traumatic brain injury. American Journal of Translational Research,
Du, G., Zhao, Z., Chen, Y., Li, Z., Tian, Y., Liu, Z., … Song, J. (2016). 9(3), 887–899.
Quercetin attenuates neuronal autophagy and apoptosis in rat Gustafsson, Å. B., & Dorn, G. W., 2nd (2019). Evolving and expanding the roles
traumatic brain injury model via activation of PI3K/Akt signaling of mitophagy as a homeostatic and pathogenic process. Physiological
pathway. Neurological Research, 38(11), 1012–1019. https://doi.org/ Reviews, 99(1), 853–892. https://doi.org/10.1152/physrev.00005.2018
10.1080/01616412.2016.1240393 He, H., Liu, W., Zhou, Y., Liu, Y., Weng, P., Li, Y., & Fu, H. (2018).
Du, J., Zhang, C., Na, X., Li, A., Zhang, Q., Li, K., & Ding, Y. (2018). Sevoflurane post‐conditioning attenuates traumatic brain injury‐
Andrographolide protects mouse astrocytes against hypoxia injury by induced neuronal apoptosis by promoting autophagy via the PI3K/
promoting autophagy and S100B expression. Brazilian Journal of AKT signaling pathway. Drug Design, Development and Therapy, 12,
Medical and Biological Research, 51(6), e7061. https://doi.org/10.1590/ 629–638. https://doi.org/10.2147/dddt.S158313
1414‐431x20177061 Hensley, K., Poteshkina, A., Johnson, M. F., Eslami, P., Gabbita, S. P.,
El‐Gazar, A. A., Soubh, A. A., Mohamed, E. A., Awad, A. S., & El‐Abhar, H. S. Hristov, A. M., … Harris‐White, M. E. (2016). Autophagy modulation by
(2019). Morin post‐treatment confers neuroprotection in a novel rat lanthionine ketimine ethyl ester improves long‐term outcome after
model of mild repetitive traumatic brain injury by targeting dementia central fluid percussion injury in the mouse. Journal of Neurotrauma,
markers, APOE, autophagy and Wnt/β‐catenin signaling pathway. 33(16), 1501–1513. https://doi.org/10.1089/neu.2015.4196
Brain Research, 1717, 104–116. https://doi.org/10.1016/j.brainres. Hong, M. Y., Gao, J. L., Cui, J. Z., Wang, K. J., Tian, Y. X., Li, R., & Wang, H.
2019.04.003 (2012). Effect of c‐Jun NH(2)‐terminal kinase‐mediated p53 expres-
Erlich, S., Alexandrovich, A., Shohami, E., & Pinkas‐Kramarski, R. (2007). sion on neuron autophagy following traumatic brain injury in rats.
Rapamycin is a neuroprotective treatment for traumatic brain injury. Chinese Medical Journal (England), 125(11), 2019–2024.
Neurobiology of Disease, 26(1), 86–93. https://doi.org/10.1016/j.nbd. Huang, Y. N., Yang, L. Y., Greig, N. H., Wang, Y. C., Lai, C. C., & Wang, J. Y.
2006.12.003 (2018). Neuroprotective effects of pifithrin‐α against traumatic brain
Erlich, S., Shohami, E., & Pinkas‐Kramarski, R. (2006). Neurodegeneration injury in the striatum through suppression of neuroinflammation,
induces upregulation of Beclin 1. Autophagy, 2(1), 49–51. oxidative stress, autophagy, and apoptosis. Scientific Reports, 8(1),
Fairlie‐Clarke, K., Barbour, M., Wilson, C., Hridi, S. U., Allan, D., & Jiang, H. R. 2368. https://doi.org/10.1038/s41598‐018‐19654‐x
(2018). Expression and function of IL‐33/ST2 axis in the central Jiang, H., Wang, Y., Liang, X., Xing, X., Xu, X., & Zhou, C. (2018). Toll‐like
nervous system under normal and diseased conditions. Frontiers in receptor 4 knockdown attenuates brain damage and neuroinflammation
Immunology, 9, 2596. https://doi.org/10.3389/fimmu.2018.02596 after traumatic brain injury via inhibiting neuronal autophagy and
ZENG ET AL. | 11

astrocyte activation. Cellular and Molecular Neurobiology, 38(5), oxygenase‐1 pathway‐mediated mitophagy alleviates traumatic brain
1009–1019. https://doi.org/10.1007/s10571‐017‐0570‐5 injury‐induced intestinal mucosa damage and epithelial barrier
Jin, Y., Lei, J., Lin, Y., Gao, G., & Jiang, J. (2016). Autophagy inhibitor 3‐MA dysfunction. Journal of Neurotrauma, 34(13), 2119–2131. https://doi.
weakens neuroprotective effects of posttraumatic brain injury org/10.1089/neu.2016.4764
moderate hypothermia. World Neurosurgery, 88, 433–446. https:// Liu, Y., Wang, R., Zhao, Z., Dong, W., Zhang, X., Chen, X., & Ma, L. (2017).
doi.org/10.1016/j.wneu.2015.10.055 Short‐term caloric restriction exerts neuroprotective effects following
Jin, Y., Lin, Y., Feng, J., Jia, F., Gao, G., & Jiang, J. (2015a). Attenuation of mild traumatic brain injury by promoting autophagy and inhibiting
cell death in injured cortex after post‐traumatic brain injury moderate astrocyte activation. Behavioural Brain Research, 331, 135–142.
hypothermia: Possible involvement of autophagy pathway. World https://doi.org/10.1016/j.bbr.2017.04.024
Neurosurgery, 84(2), 420–430. https://doi.org/10.1016/j.wneu.2015. Luo, C. L., Li, B. X., Li, Q. Q., Chen, X. P., Sun, Y. X., Bao, H. J., … Zhao, Z. Q.
03.039 (2011). Autophagy is involved in traumatic brain injury‐induced cell
Jin, Y., Lin, Y., Feng, J., Jia, F., Gao, G., & Jiang, J. (2015b). Moderate death and contributes to functional outcome deficits in mice.
hypothermia significantly decreases hippocampal cell death invol- Neuroscience, 184, 54–63. https://doi.org/10.1016/j.neuroscience.
ving autophagy pathway after moderate traumatic brain injury. 2011.03.021
Journal of Neurotrauma, 32(14), 1090–1100. https://doi.org/10.1089/ Ma, J., Ni, H., Rui, Q., Liu, H., Jiang, F., Gao, R., … Chen, G. (2019).
neu.2014.3649 Potential roles of NIX/BNIP3L pathway in rat traumatic brain injury.
Jin, Y., Wang, R., Yang, S., Zhang, X., & Dai, J. (2017). Role of microglia Cell Transplantation, 28, 585–595. https://doi.org/10.1177/096368
autophagy in microglia activation after traumatic brain injury. World 9719840353
Neurosurgery, 100, 351–360. https://doi.org/10.1016/j.wneu.2017. Ma, L., Li, Z., Liu, Z., Li, M., Sui, D., Liu, Y., … Li, G. (2015). 17AAG improves
01.033 histological and functional outcomes in a rat CCI model through
Kim, J., Kim, T. Y., Cho, K. S., Kim, H. N., & Koh, J. Y. (2013). Autophagy autophagy activation and apoptosis attenuation. Neuroscience Letters,
activation and neuroprotection by progesterone in the G93A‐SOD1 599, 1–6. https://doi.org/10.1016/j.neulet.2015.05.004
transgenic mouse model of amyotrophic lateral sclerosis. Neurobiology Maiti, P., Peruzzaro, S., Kolli, N., Andrews, M., Al‐Gharaibeh, A., Rossignol, J.,
of Disease, 59, 80–85. https://doi.org/10.1016/j.nbd.2013.07.011 & Dunbar, G. L. (2019). Transplantation of mesenchymal stem cells
Kim, J., & Klionsky, D. J. (2000). Autophagy, cytoplasm‐to‐vacuole overexpressing interleukin‐10 induces autophagy response and pro-
targeting pathway, and pexophagy in yeast and mammalian cells. motes neuroprotection in a rat model of TBI. Journal of Cellular and
Annual Review of Biochemistry, 69, 303–342. https://doi.org/10.1146/ Molecular Medicine, 23, 5211–5224. https://doi.org/10.1111/jcmm.
annurev.biochem.69.1.303 14396
Klionsky, D. J., & Emr, S. D. (2000). Autophagy as a regulated pathway of Nazmi, A., Field, R. H., Griffin, E. W., Haugh, O., Hennessy, E., Cox, D., …
cellular degradation. Science, 290(5497), 1717–1721. Cunningham, C. (2019). Chronic neurodegeneration induces type I
Lai, Y., Hickey, R. W., Chen, Y., Bayιr, H., Sullivan, M. L., Chu, C. T., … Clark, interferon synthesis via STING, shaping microglial phenotype and
R. S. (2008). Autophagy is increased after traumatic brain injury in accelerating disease progression. Glia, 67, 1254–1276. https://doi.org/
mice and is partially inhibited by the antioxidant γ‐glutamylcysteinyl 10.1002/glia.23592
ethyl ester. Journal of Cerebral Blood Flow and Metabolism, 28(3), Niu, F., Dong, J., Xu, X., Zhang, B., & Liu, B. (2019). Mitochondrial Division
540–550. https://doi.org/10.1038/sj.jcbfm.9600551 inhibitor 1 prevents early‐stage induction of mitophagy and acceler-
Langlois, J. A., Rutland‐Brown, W., & Wald, M. M. (2006). The ated cell death in a rat model of moderate controlled cortical impact
epidemiology and impact of traumatic brain injury: A brief overview. brain injury. World Neurosurgery, 122, e1090–e1101. https://doi.org/
Journal of Head Trauma Rehabilitation, 21(5), 375–378. 10.1016/j.wneu.2018.10.236
Li, D., Huang, S., Yin, Z., Zhu, J., Ge, X., Han, Z., … Lei, P. (2019). Increases in Saatman, K. E., Duhaime, A. C., Bullock, R., Maas, A. I. R., Valadka, A., &
miR‐124‐3p in microglial exosomes confer neuroprotective effects by Manley, G. T. (2008). Classification of traumatic brain injury for
targeting FIP200‐mediated neuronal autophagy following traumatic targeted therapies. Journal of Neurotrauma, 25(7), 719–738. https://
brain injury. Neurochemical Research, 44, 1903–1923. https://doi.org/ doi.org/10.1089/neu.2008.0586
10.1007/s11064‐019‐02825‐1 Sadasivan, S., Dunn, W. A., Jr., Hayes, R. L., & Wang, K. K. W. (2008).
Li, D., Huang, S., Zhu, J., Hu, T., Han, Z., Zhang, S., … Lei, P. (2019). Changes in autophagy proteins in a rat model of controlled cortical
Exosomes from MiR‐21‐5p‐increased neurons play a role in neuro- impact induced brain injury. Biochemical and Biophysical Research
protection by suppressing Rab11a‐mediated neuronal autophagy in Communications, 373(4), 478–481. https://doi.org/10.1016/j.bbrc.
vitro after traumatic brain injury. Medical Science Monitor, 25, 2008.05.031
1871–1885. https://doi.org/10.12659/msm.915727 Sarkar, C., Jones, J. W., Hegdekar, N., Thayer, J. A., Kumar, A., Faden, A. I., …
Lin, C., Chao, H., Li, Z., Xu, X., Liu, Y., Hou, L., … Ji, J. (2016). Melatonin Lipinski, M. M. (2019). PLA2G4A/cPLA2‐mediated lysosomal mem-
attenuates traumatic brain injury‐induced inflammation: A possible brane damage leads to inhibition of autophagy and neurodegeneration
role for mitophagy. Journal of Pineal Research, 61(2), 177–186. https:// after brain trauma. Autophagy, 1–20. https://doi.org/10.1080/155486
doi.org/10.1111/jpi.12337 27.2019.1628538
Lin, C. J., Chen, T. H., Yang, L. Y., & Shih, C. M. (2014). Resveratrol protects Sarkar, C., Zhao, Z., Aungst, S., Sabirzhanov, B., Faden, A. I., & Lipinski, M. M.
astrocytes against traumatic brain injury through inhibiting apoptotic (2014). Impaired autophagy flux is associated with neuronal cell death
and autophagic cell death. Cell Death & Disease, 5, e1147. https://doi. after traumatic brain injury. Autophagy, 10(12), 2208–2222. https://doi.
org/10.1038/cddis.2014.123 org/10.4161/15548627.2014.981787
Liu, C. L., Chen, S., Dietrich, D., & Hu, B. R. (2008). Changes in autophagy after Sebastiani, A., Gölz, C., Sebastiani, P. G., Bobkiewicz, W., Behl, C.,
traumatic brain injury. Journal of Cerebral Blood Flow and Metabolism, Mittmann, T., … Engelhard, K. (2017). Sequestosome 1 deficiency
28(4), 674–683. https://doi.org/10.1038/sj.jcbfm.9600587 delays, but does not prevent brain damage formation following acute
Liu, H., Rose, M. E., Ma, X., Culver, S., Dixon, C. E., & Graham, S. H. (2017). brain injury in adult mice. Frontiers in Neuroscience, 11, 678. https://
In vivo transduction of neurons with TAT‐UCH‐L1 protects brain doi.org/10.3389/fnins.2017.00678
against controlled cortical impact injury. PLOS One, 12(5), e0178049. Sun, L., Gao, J., Zhao, M., Cui, J., Li, Y., Yang, X., … Wu, Z. (2015). A novel
https://doi.org/10.1371/journal.pone.0178049 cognitive impairment mechanism that astrocytic p‐connexin 43
Liu, Y., Bao, Z., Xu, X., Chao, H., Lin, C., Li, Z., … Ji, J. (2017). Extracellular promotes neuronic autophagy via activation of P2X7R and down‐
signal‐regulated kinase/nuclear factor‐erythroid2‐like2/heme regulation of GLT‐1 expression in the hippocampus following
12 | ZENG ET AL.

traumatic brain injury in rats. Behavioural Brain Research, 291, Biochemistry & Cell Biology, 94, 44–55. https://doi.org/10.1016/j.
315–324. https://doi.org/10.1016/j.bbr.2015.05.049 biocel.2017.11.007
Sun, L., Gao, J., Zhao, M., Jing, X., Cui, Y., Xu, X., … Cui, J. (2014). The Xu, J., Wang, H., Lu, X., Ding, K., Zhang, L., He, J., … Wu, Y. (2014).
effects of BMSCs transplantation on autophagy by CX43 in the Posttraumatic administration of luteolin protects mice from trau-
hippocampus following traumatic brain injury in rats. Neurological matic brain injury: Implication of autophagy and inflammation. Brain
Science, 35(5), 677–682. https://doi.org/10.1007/s10072‐013‐1575‐6 Research, 1582, 237–246. https://doi.org/10.1016/j.brainres.2014.
Sun, L., Liu, A., Zhang, J., Ji, W., Li, Y., Yang, X., … Guo, J. (2018). miR‐23b 07.042
improves cognitive impairments in traumatic brain injury by targeting Xu, K., Wu, F., Xu, K., Li, Z., Wei, X., Lu, Q., … Zhang, H. (2018). NaHS
ATG12‐mediated neuronal autophagy. Behavioural Brain Research, restores mitochondrial function and inhibits autophagy by activating
340, 126–136. https://doi.org/10.1016/j.bbr.2016.09.020 the PI3K/Akt/mTOR signalling pathway to improve functional
Sun, L., Zhao, M., Liu, M., Su, P., Zhang, J., Li, Y., … Wu, Z. (2018). recovery after traumatic brain injury. Chemico‐Biological Interactions,
Suppression of FoxO3a attenuates neurobehavioral deficits after 286, 96–105. https://doi.org/10.1016/j.cbi.2018.02.028
traumatic brain injury through inhibiting neuronal autophagy. Xu, X., Zhi, T., Chao, H., Jiang, K., Liu, Y., Bao, Z., … Ji, J. (2018). ERK1/2/
Behavioural Brain Research, 337, 271–279. https://doi.org/10.1016/j. mTOR/Stat3 pathway‐mediated autophagy alleviates traumatic brain
bbr.2017.08.042 injury‐induced acute lung injury. Biochimica et Biophysica Acta (BBA) ‐
Sun, L., Zhao, M., Wang, Y., Liu, A., Lv, M., Li, Y., … Wu, Z. (2017). Molecular Basis of Disease, 1864(5 Pt A), 1663–1674. https://doi.org/
Neuroprotective effects of miR‐27a against traumatic brain injury via 10.1016/j.bbadis.2018.02.011
suppressing FoxO3a‐mediated neuronal autophagy. Biochemical and Yao, J., Zheng, K., & Zhang, X. (2015). Rosiglitazone exerts neuroprotec-
Biophysical Research Communications, 482(4), 1141–1147. https://doi. tive effects via the suppression of neuronal autophagy and apoptosis
org/10.1016/j.bbrc.2016.12.001 in the cortex following traumatic brain injury. Molecular Medicine
Sun, L. Q., Gao, J. L., Cui, C. M., Cui, Y., Jing, X. B., Zhao, M. M., … CUI, J. Reports, 12(5), 6591–6597. https://doi.org/10.3892/mmr.2015.4292
Z. (2014). Astrocytic p‐connexin 43 regulates neuronal autophagy Ye, Y., Zhang, P., Qian, Y., Yin, B., & Yan, M. (2017). The effect of
in the hippocampus following traumatic brain injury in rats. pyrroloquinoline quinone on the expression of WISP1 in traumatic
Molecular Medicine Reports, 9(1), 77–82. https://doi.org/10.3892/ brain injury. Stem Cells International, 2017, 1–16. https://doi.org/10.
mmr.2013.1787 1155/2017/4782820
Sun, L. Q., Gao, J. L., Cui, Y., Zhao, M. M., Jing, X. B., Li, R., … WU, Z. X. Yi, M., Dai, X., Li, Q., Xu, X., Chen, Y., & Wang, D. (2019). Downregulated
(2015). Neuronic autophagy contributes to p‐connexin 43 degrada- lncRNA CRNDE contributes to the enhancement of nerve repair after
tion in hippocampal astrocytes following traumatic brain injury in rats. traumatic brain injury in rats. Cell Cycle, 18(18), 2332–2343. https://
Molecular Medicine Reports, 11(6), 4419–4423. https://doi.org/10. doi.org/10.1080/15384101.2019.1647024
3892/mmr.2015.3264 Yin, Y., Li, E., Sun, G., Yan, H. Q., Foley, L. M., Andrzejczuk, L. A., … Sun, D.
Tang, C., Shan, Y., Hu, Y., Fang, Z., Tong, Y., Chen, M., … Xu, X. (2017). (2018). Effects of DHA on hippocampal autophagy and lysosome
FGF2 attenuates neural cell death via suppressing autophagy after rat function after traumatic brain injury. Molecular Neurobiology, 55(3),
mild traumatic brain injury. Stem Cells International, 2017, 1–14. 2454–2470. https://doi.org/10.1007/s12035‐017‐0504‐8
https://doi.org/10.1155/2017/2923182 Zeng, X. J., Li, P., Ning, Y. L., Zhao, Y., Peng, Y., Yang, N., … Zhou, Y. G.
Wang, C., Hu, Z., Zou, Y., Xiang, M., Jiang, Y., Botchway, B. O. A., … Fang, M. (2018). Impaired autophagic flux is associated with the severity of
(2017). The post‐therapeutic effect of rapamycin in mild traumatic trauma and the role of A2AR in brain cells after traumatic brain injury.
brain‐injured rats ensuing in the upregulation of autophagy and Cell Death & Disease, 9(2), 252. https://doi.org/10.1038/s41419‐018‐
mitophagy. Cell Biology International, 41(9), 1039–1047. https://doi. 0316‐4
org/10.1002/cbin.10820 Zhang, F., Dong, H., Lv, T., Jin, K., Jin, Y., Zhang, X., & Jiang, J. (2018).
Wang, C. Q., Ye, Y., Chen, F., Han, W. C., Sun, J. M., Lu, X., … Liao, L. C. (2017). Moderate hypothermia inhibits microglial activation after traumatic
Posttraumatic administration of a sub‐anesthetic dose of ketamine brain injury by modulating autophagy/apoptosis and the MyD88‐
exerts neuroprotection via attenuating inflammation and autophagy. dependent TLR4 signaling pathway. Journal of Neuroinflammation,
Neuroscience, 343, 30–38. https://doi.org/10.1016/j.neuroscience.2016. 15(1), 273. https://doi.org/10.1186/s12974‐018‐1315‐1
11.029 Zhang, J. Y., Lee, J. H., Gu, X., Wei, Z. Z., Harris, M. J., Yu, S. P., & Wei, L.
Wang, T., Zhang, L., Zhang, M., Bao, H., Liu, W., Wang, Y., … Tao, L. (2013). (2018). Intranasally delivered Wnt3a improves functional recovery
[Gly14]‐Humanin reduces histopathology and improves functional after traumatic brain injury by modulating autophagic, apoptotic, and
outcome after traumatic brain injury in mice. Neuroscience, 231, regenerative pathways in the mouse brain. Journal of Neurotrauma,
70–81. https://doi.org/10.1016/j.neuroscience.2012.11.019 35(5), 802–813. https://doi.org/10.1089/neu.2016.4871
Wang, Y. Q., Wang, L., Zhang, M. Y., Wang, T., Bao, H. J., Liu, W. L., … Tao, Zhang, L., Ding, K., Wang, H., Wu, Y., & Xu, J. (2016). Traumatic brain
L. Y. (2012). Necrostatin‐1 suppresses autophagy and apoptosis in injury‐induced neuronal apoptosis is reduced through modulation of
mice traumatic brain injury model. Neurochemical Research, 37(9), PI3K and autophagy pathways in mouse by FTY720. Cellular and
1849–1858. https://doi.org/10.1007/s11064‐012‐0791‐4 Molecular Neurobiology, 36(1), 131–142. https://doi.org/10.1007/
Wang, Z., Gao, C., Chen, W., Gao, Y., Wang, H., Meng, Y., … Tao, L. (2019). s10571‐015‐0227‐1
Salubrinal offers neuroprotection through suppressing endoplasmic Zhang, L., Wang, H., Fan, Y., Gao, Y., Li, X., Hu, Z., … Wang, X. (2017).
reticulum stress, autophagy and apoptosis in a mouse traumatic brain Fucoxanthin provides neuroprotection in models of traumatic brain
injury model. Neurobiology of Learning and Memory, 161, 12–25. injury via the Nrf2‐ARE and Nrf2‐autophagy pathways. Scientific
https://doi.org/10.1016/j.nlm.2019.03.002 Reports, 7, 46763. https://doi.org/10.1038/srep46763
Wiesner, D., Tar, L., Linkus, B., Chandrasekar, A., Olde Heuvel, F., Dupuis, L., … Zhang, M., Shan, H., Chang, P., Ma, L., Chu, Y., Shen, X., … Tao, L. (2017).
Roselli, F. (2018). Reversible induction of TDP‐43 granules in cortical Upregulation of 3‐MST relates to neuronal autophagy after traumatic
neurons after traumatic injury. Experimental Neurology, 299(Pt A), 15–25. brain injury in mice. Cellular and Molecular Neurobiology, 37(2),
https://doi.org/10.1016/j.expneurol.2017.09.011 291–302. https://doi.org/10.1007/s10571‐016‐0369‐9
Wu, Q., Gao, C., Wang, H., Zhang, X., Li, Q., Gu, Z., … Tao, L. (2018). Zhang, M., Shan, H., Chang, P., Wang, T., Dong, W., Chen, X., & Tao, L.
Mdivi‐1 alleviates blood‐brain barrier disruption and cell death in (2014). Hydrogen sulfide offers neuroprotection on traumatic brain
experimental traumatic brain injury by mitigating autophagy injury in parallel with reduced apoptosis and autophagy in mice. PLOS
dysfunction and mitophagy activation. The International Journal of One, 9(1), e87241. https://doi.org/10.1371/journal.pone.0087241
ZENG ET AL. | 13

Zhang, P., Ye, Y., Qian, Y., Yin, B., Zhao, J., Zhu, S., … Yan, M. (2017). The Zhao, M., Liang, F., Xu, H., Yan, W., & Zhang, J. (2016). Methylene blue exerts
effect of pyrroloquinoline quinone on apoptosis and autophagy in a neuroprotective effect against traumatic brain injury by promoting
traumatic brain injury. CNS & Neurological Disorders ‐ Drug Targets, 16(6), autophagy and inhibiting microglial activation. Molecular Medicine
724–736. https://doi.org/10.2174/1871527316666170124164306 Reports, 13(1), 13–20. https://doi.org/10.3892/mmr.2015.4551
Zhang, Y. B., Li, S. X., Chen, X. P., Yang, L., Zhang, Y. G., Liu, R., & Tao, L. Y. Zhou, H., Andonegui, G., Wong, C. H. Y., & Kubes, P. (2009). Role of
(2008). Autophagy is activated and might protect neurons from endothelial TLR4 for neutrophil recruitment into central nervous
degeneration after traumatic brain injury. Neuroscience Bulletin, 24(3), system microvessels in systemic inflammation. Journal of Immunology,
143–149. https://doi.org/10.1007/s12264‐008‐1108‐0 183(8), 5244–5250. https://doi.org/10.4049/jimmunol.0901309
Zhang, Z., Li, D., Xu, L., & Li, H. P. (2019). Sirt1 improves functional
recovery by regulating autophagy of astrocyte and neuron after brain
injury. Brain Research Bulletin, 150, 42–49. https://doi.org/10.1016/j.
brainresbull.2019.05.005 How to cite this article: Zeng Z, Zhang Y, Jiang W, He L, Qu
Zhao, L., Liu, Q., Ma, S., Zhang, Y., & Liang, P. (2018). TPEN attenuates
H. Modulation of autophagy in traumatic brain injury. J Cell
neural autophagy induced by synaptically‐released zinc translocation
and improves histological outcomes after traumatic brain injury in Physiol. 2019;1–13. https://doi.org/10.1002/jcp.29173
rats. Annals of Clinical and Laboratory Science, 48(4), 446–452.

You might also like