You are on page 1of 14

Autophagy

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/kaup20

BNIP3L/NIX degradation leads to mitophagy


deficiency in ischemic brains

Xiaoli Wu, Yanrong Zheng, Mengru Liu, Yue Li, Shijia Ma, Weidong Tang,
Wenping Yan, Ming Cao, Wanqing Zheng, Lei Jiang, Jiaying Wu, Feng Han,
Zhenghong Qin, Liang Fang, Weiwei Hu, Zhong Chen & Xiangnan Zhang

To cite this article: Xiaoli Wu, Yanrong Zheng, Mengru Liu, Yue Li, Shijia Ma, Weidong Tang,
Wenping Yan, Ming Cao, Wanqing Zheng, Lei Jiang, Jiaying Wu, Feng Han, Zhenghong Qin,
Liang Fang, Weiwei Hu, Zhong Chen & Xiangnan Zhang (2021) BNIP3L/NIX degradation
leads to mitophagy deficiency in ischemic brains, Autophagy, 17:8, 1934-1946, DOI:
10.1080/15548627.2020.1802089

To link to this article: https://doi.org/10.1080/15548627.2020.1802089

View supplementary material

Published online: 12 Aug 2020.

Submit your article to this journal

Article views: 8504

View related articles

View Crossmark data

Citing articles: 56 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=kaup20
AUTOPHAGY
2021, VOL. 17, NO. 8, 1934–1946
https://doi.org/10.1080/15548627.2020.1802089

RESEARCH PAPER

BNIP3L/NIX degradation leads to mitophagy deficiency in ischemic brains


Xiaoli Wua*, Yanrong Zhenga*, Mengru Liua, Yue Lia, Shijia Ma a, Weidong Tanga, Wenping Yanb, Ming Caoa,
Wanqing Zhenga, Lei Jianga, Jiaying Wub, Feng Hanc, Zhenghong Qind, Liang Fang e, Weiwei Hua, Zhong Chen a
,
and Xiangnan Zhang a
a
Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of
China, Zhejiang University, Hangzhou, China; bThe First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; cKey
Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China; dDepartment of
Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-
Diseases, Soochow University School of Pharmaceutical Sciences, Suzhou, China; eAcademy for Advanced Interdisciplinary Studies and Department
of Biology, Southern University of Science and Technology, Shenzhen, China

ABSTRACT ARTICLE HISTORY


Mitophagy, the elimination of damaged mitochondria through autophagy, promotes neuronal survival Received 5 September 2019
in cerebral ischemia. Previous studies found deficient mitophagy in ischemic neurons, but the mechan­ Revised 17 July 2020
isms are still largely unknown. We determined that BNIP3L/NIX, a mitophagy receptor, was degraded by Accepted 22 July 2020
proteasomes, which led to mitophagy deficiency in both ischemic neurons and brains. BNIP3L exists as KEYWORDS
a monomer and homodimer in mammalian cells, but the effects of homodimer and monomer on BNIP3L/NIX; carfilzomib;
mitophagy are unclear. Site-specific mutations in the transmembrane domain of BNIP3L (S195A and cerebral ischemia;
G203A) only formed the BNIP3L monomer and failed to induce mitophagy. Moreover, overexpression of mitophagy; ubiquitin-
wild-type BNIP3L, in contrast to the monomeric BNIP3L, rescued the mitophagy deficiency and pro­ proteasome pathway
tected against cerebral ischemic injury. The macroautophagy/autophagy inhibitor 3-MA and the protea­
some inhibitor MG132 were used in cerebral ischemic brains to identify how BNIP3L was reduced. We
found that MG132 blocked the loss of BNIP3L and subsequently promoted mitophagy in ischemic
brains. In addition, the dimeric form of BNIP3L was more prone to be degraded than its monomeric
form. Carfilzomib, a drug for multiple myeloma therapy that inhibits proteasomes, reversed the BNIP3L
degradation and restored mitophagy in ischemic brains. This treatment protected against either acute or
chronic ischemic brain injury. Remarkably, these effects of carfilzomib were abolished in bnip3l-/- mice.
Taken together, the present study linked BNIP3L degradation by proteasomes with mitophagy defi­
ciency in cerebral ischemia. We propose carfilzomib as a novel therapy to rescue ischemic brain injury by
preventing BNIP3L degradation.
Abbreviations: 3-MA: 3-methyladenine; AAV: adeno-associated virus; ATG7: autophagy related 7;
BCL2L13: BCL2-like 13 (apoptosis facilitator); BNIP3L/NIX: BCL2/adenovirus E1B interacting protein
3-like; CCCP: carbonyl cyanide 3-chlorophenylhydrazone; CFZ: carfilzomib; COX4I1: cytochrome
c oxidase subunit 4I1; CQ: chloroquine; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP:
green fluorescent protein; I-R: ischemia-reperfusion; MAP1LC3A/LC3A: microtube-associated protein 1
light chain 3 alpha; MAP1LC3B/LC3B: microtube-associated protein 1 light chain 3 beta; O-R: oxygen and
glucose deprivation-reperfusion; OGD: oxygen and glucose deprivation; PHB2: prohibitin 2; pMCAO:
permanent middle cerebral artery occlusion; PRKN/PARK2: parkin RBR E3 ubiquitin protein ligase; PT:
photothrombosis; SQSTM1: sequestosome 1; tMCAO: transient middle cerebral artery occlusion;
TOMM20: translocase of outer mitochondrial membrane 20; TTC: 2,3,5-triphenyltetrazolium
hydrochloride.

Introduction mitophagy, is essential in controlling mitochondrial quality


[5]. Autophagic machinery eliminates damaged mitochon­
Cerebral ischemia results in high mortality and disability
dria and prevents mitochondrial-dependent apoptosis in
and remains one of the most refractory human diseases
ischemic neurons [6]. Enhanced mitophagy has thus been
[1,2]. Besides being the source of bioenergy, mitochondria
proposed as a therapeutic strategy to protect neurons from
function directly in regulating programmed cell death [3]
ischemic insults [7–9]. Unfortunately, few drugs can mod­
and mitochondrial damage has been well-documented as
ulate mitophagy in ischemic brains [10]. Mitophagy defi­
a cause of neuronal death in ischemic neurons [4].
ciency leads to accumulation of damaged mitochondria in
Mitochondrial elimination by autophagy, termed
brains, causing bioenergetic dysfunction and neuronal

CONTACT Xiangnan Zhang xiangnan_zhang@zju.edu.cn; Zhong Chen chenzhong@zju.edu.cn Institute of Pharmacology & Toxicology, College of
Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou 310058, China
*These authors contributed equally to this work
Supplemental data for this article can be accessed here.
© 2020 Informa UK Limited, trading as Taylor & Francis Group
AUTOPHAGY 1935

inflammation [11]. Notably, mitochondria accumulate in were in line with previous observations [4,7,12,14] and
a variety of hypoxic and/or ischemic neuronal injury mod­ implied deficiency in neuronal mitophagy with ischemia
els [12,13], suggesting the insufficiency of mitophagy in alone. To confirm this phenotype, primary cultured cortical
ischemia [7]. In particular, our previous studies showed neurons were subjected to oxygen and glucose deprivation
that mitophagy can be activated by ischemia-reperfusion, (OGD) or OGD-reperfusion (O-R) treatment. Similarly,
while ischemia alone did not trigger mitophagy in spite of both O-R and OGD alone caused LC3B lipidation and
autophagy activation [7,14]. These results implied that reduced SQSTM1. However, OGD alone failed to cause
ischemic brains without reperfusion might be rescued by mitochondrial loss as in O-R treated neurons. We con­
restoring mitophagy. However, the molecular mechanisms firmed the autophagic mitochondrial loss because of chlor­
underlying neuronal mitophagy deficiency in ischemic neu­ oquine treatment resulted in further accumulation of LC3B-
rons remain largely unclear. II and reversed the degradation of SQSTM1, TOMM20, and
BNIP3L/NIX is located on the mitochondrial outer COX4I1 [28] (Figure 1C,D).
membrane, where it serves as a mitophagy receptor in To further identify mitophagy deficiency, atg7 (autophagy
either cell development or pathological conditions [15,16]. related 7) knockout primary cortical neurons (Atg7fl/fl; hSyn-
We previously found that Bnip3l gene deletion significantly Cre) [29] were used. atg7 knockout blocked the mitophagy in
blocked ischemia-reperfusion-induced neuronal mitophagy O-R neurons but not neurons after OGD alone (Figure 1E,F).
and consequently exacerbated ischemic brain injury. These We transfected neurons with mCherry-LC3B and Mito-GFP,
results highlighted BNIP3L as a potential target of mito­ which labeled autophagosomes and mitochondria, respec­
phagy for stroke intervention [6,8,17]. Nevertheless, it tively. Both O-R and OGD treatment increased the number
remained unclear how to manipulate mitophagy in of autophagosomes. OGD alone did not promote the overlap
ischemic neurons by regulating BNIP3L. Intriguingly, of autophagosomes with mitochondria (Fig. S1A and S1B). As
BNIP3L was reduced along with the differentiation of either an alternative approach to identify mitophagy, primary cul­
erythroblast [16] or cardiac progenitor cells [18]. Likewise, tured neurons were transfected with Mito-QC, which is
reduced BNIP3L was found in several disorders with insuf­ widely used to monitor mitophagy [30]. Consistent with the
ficient mitophagy, e.g., traumatic brain injury [19], chronic western blot data, O-R treatment enhanced the mitochondria
obstructive pulmonary disease [20] and acute myeloid leu­ ratio in mCherry-alone. In contrast, OGD alone reduced
kemia [21]. These findings implied the potential association mitochondria fusion with lysosomes (Fig. S1C and S1D).
of reduced BNIP3L with lower mitophagy activity. Together, these data revealed that ischemia alone compro­
However, whether the abundance of BNIP3L controls the mised neuronal mitophagy despite the activation of
duration of mitophagy remains unclear, and how BNIP3L autophagy.
was reduced under pathological conditions is also ambig­
uous. BNIP3L forms either a monomer or homodimer in
BNIP3L relates to mitophagy deficiency in cerebral
mammalian cells [22,23] and it seems that a homodimer of
ischemia
BNIP3L is required for its proapoptotic effects in tumor
cells [24,25]. However, the contributions of homodimer and Since ischemia did not compromise autophagy activation either
monomer of BNIP3L to mitophagy have not been clarified in vivo or in vitro, we thus assumed a mitophagy-specific
[26,27]. Therefore, further characterization of the molecular mechanism deficiency. We examined the protein levels of
regulation of BNIP3L in ischemic brains may provide novel PRKN/PARK2, BNIP3L, FUNDC1, BCL2L13, PHB2 (prohibi­
therapies for cerebral ischemia. tin 2) and BNIP3 in pMCAO brains [6,31–34]. The results
In the present study, we aimed to explore how neuronal showed significant BNIP3L reduction with pMCAO treatment
BNIP3L was regulated in ischemic neurons. We unexpectedly while the expression of PRKN, FUNDC1, BCL2L13, PHB2 and
found that BNIP3L was degraded by proteasomes, which led BNIP3 remained intact. These results suggested that these
to mitophagy deficiency. Carfilzomib, a proteasome inhibitor mitophagy receptors, excepting BNIP3L, may have little impact
for multiple myeloma therapy, protected brains from ischemic on mitophagy deficiency in ischemic brains (Fig. S2). To con­
injury by rescuing BNIP3L degradation. firm the involvement of BNIP3L loss in mitophagy deficiency,
we infected the mice brain cortex and striatum with adeno-
associated viruses carrying the cDNA of GFP-BNIP3L (AAV-
Results GFP-Bnip3l) two weeks prior to pMCAO. The mitophagy
deficiency was rescued by ectopic expression of BNIP3L as
Ischemia alone leads to neuronal mitophagy deficiency
revealed by loss of mitochondrial markers COX4I1 and
To identify mitophagy induction in ischemic brains, we TOMM20 (Figure 2A,B).
determined the expression of mitophagy-related proteins In line with these findings, we found that endogenous
in mice subjected to either transient middle cerebral artery BNIP3L decreased after OGD treatment in primary cul­
occlusion (tMCAO) or permanent MCAO (pMCAO). Both tured neurons, while the mitochondrial content was not
tMCAO and pMCAO led to increased LC3B-II and reduced significantly affected (Figure 2C). These phenotypes were
SQSTM1, indicating autophagy activation. The mitochon­ rescued by overexpression of Flag-BNIP3L (Figure 2D). The
drial proteins COX4I1 and TOMM20, which reflect mito­ immunostaining showed the same results (Fig. S3). Because
chondrial content, decreased in tMCAO- but not in the abundance of mitophagy receptors cannot reflect the
pMCAO-treated mice brains (Figure 1A,B). These data mitochondrial mass [35–37], we determined the COX4I1,
1936 X. WU ET AL.

Figure 1. Ischemia alone leads to neuronal mitophagy deficiency. (A) Mice were subjected to 1 h of occlusion followed by 3 h of transient middle cerebral artery
occlusion (tMCAO) or 4 h of permanent middle cerebral artery occlusion (pMCAO). The expression of SQSTM1, LC3B, COX4I1, and TOMM20 in ischemic penumbra was
detected by western blot. Duplicate lanes are shown for each grou(B) Semi-quantitative analyses are shown (n = 5 mice for each group). (C) Primary cultured mice
cortical neurons were subjected to 1 h of oxygen and glucose deprivation (OGD) followed by 1 h of reperfusion (O-R) or 2 h of OGD. The neurons were incubated
with 20 μM chloroquine for 4 h prior to the OGD procedure. The protein expression of SQSTM1, LC3B, COX4I1, and TOMM20 was detected by western blot, and (D)
relative protein markers were analyzed (n = 3 from independent experiments). Ctrl, control; Veh, vehicle; CQ, chloroquine. (E) Primary cultured cortical neurons of WT
and Atg7fl/fl; hSyn-Cre were subjected to 1 h of OGD followed by 1 h of reperfusion (O-R) or 2 h of OGD. The protein levels of SQSTM1, LC3B, COX4I1, TOMM20, and
ATG7 were detected by western blot, and (F) relative protein markers were analyzed (n = 3 from independent experiments). Data are expressed as mean ± SEM.
Statistical comparisons were performed as follows: one-way ANOVA for (B, D, and F). *P < 0.05; **P < 0.01; n.s. vs. the indicated group.

TOMM20, and expression of BNIP3L along with the dura­ BNIP3L rescues mitophagy deficiency and protects
tion of pMCAO (Figure 2E). Results showed mitochondrial against cerebral ischemia
accumulation (mitophagy deficiency) and BNIP3L loss
The aforementioned results raised questions about the
along with pMCAO duration. These data indicated an
requirement of BNIP3L dimer and monomer in mitophagy
increasing duration of mitophagy deficiency. The expres­
activation. To investigate this, we constructed the BNIP3L
sions of PRKN, FUNDC1, BCL2L13 and PHB2 were not
monomer by mutating the transmembrane (TM) domain
significantly altered after pMCAO. Abundance analysis of
of BNIP3L (S195A, G203A) [39] (Figure 3A). Constructs
the correlations of PRKN, FUNDC1, BCL2L13 and PHB2
of the single site-specific BNIP3L mutants (S195A, G203A)
with mitochondrial marker COX4I1 showed no significant
showed only a monomer form in the immunoblot (Figure
changes (Figure 2F).
3B). Moreover, these mutants remained localized to the
BNIP3L can be detected at 40 and 80 kDa, represent­
mitochondria in COS7 cells and primary neurons while
ing the monomer and homodimer of BNIP3L, respec­
the TM domain deletion mutant (ΔTM) lost mitochondrial
tively [38]. The quantification of opti-density of
distribution [25] (Fig. S4A and S4B). We then transfected
immunoblots showed that the BNIP3L dimer decreased
HeLa cells with 3× Flag-fused BNIP3L or its mutants
significantly and the monomeric form decreased at
(S195A, G203A and ΔTM). Results showed that the mito­
a slower rate along with the duration of pMCAO
chondrial contents (COX4I1 and TOMM20) were signifi­
(Figure 2G). We then determined the correlation of
cantly reduced after wild-type BNIP3L overexpression,
BNIP3L dimer/monomer with mitochondrial content
while treatment of S195A and G203A mutants had
in each individual sample with distinct pMCAO dura­
a minor effect in inducing mitochondrial loss in normal
tion. The results clearly indicated a higher correlation
conditions (Figure 3C,D). The wild-type BNIP3L still
of BNIP3L dimer loss than monomer loss with the
induced mitochondrial reduction under CCCP treatment.
increased mitochondrial content (Figure 2H,I). Overall,
In contrast, both S195A and G203A mutation failed to
we found that BNIP3L dimer loss was related to mito­
reinforce CCCP-induced mitochondrial loss (Figure 3E,F).
phagy deficiency in cerebral ischemia.
AUTOPHAGY 1937

Figure 2. BNIP3L relates to mitophagy deficiency in cerebral ischemia. (A) Mice were infected with AAV-GFP-Bnip3l or AAV-GFThe mice were subjected to pMCAO for
4 h, then the protein levels of GFP, SQSTM1, LC3B, COX4I1 and TOMM20 in brain tissues were determined by western blot. Empty arrows indicate exogenous BNIP3L.
Duplicate lanes are shown for each grou(B) Semi-quantitative analyses of SQSTM1, LC3B, COX4I1 and TOMM20 protein levels are shown (n = 3 mice for each group).
(C) Primary neurons were subjected to OGD for 1 h. The expression of BNIP3L and TOMM20 were detected by western blot. Semi-quantitative analyses are shown
(n = 3 from independent experiments). (D) The neurons overexpression Flag-BNIP3L were subjected to OGD for 1 h, and the expression of Flag-BNIP3L and TOMM20
were detected by western blot (n = 3 from independent experiments). (E) Mice were subjected to 1, 3 or 6 h of pMCAO. The BNIP3L, PRKN, FUNDC1, BCL2L13, PHB2,
SQSTM1, LC3B, COX4I1 and TOMM20 protein levels were determined by western blot. Black arrows indicate endogenous BNIP3L. Duplicate lanes are shown for each
grou(F-I) Analyses of the correlations between BNIP3L, PRKN, FUNDC1, BLC2L13, PHB2, and mitochondrial proteins are shown (n = 3 mice for each group). Data are
expressed as mean ± SEM. Statistical comparisons were performed as follows: one-way ANOVA for (B and F-I) and t-test for (C and D). *P < 0.05; **P < 0.01;
***P < 0.001; n.s. vs. the indicated group.

Furthermore, exogenous BNIP3L reduced COX4I1 and with Flag-BNIP3L significantly reduced mitochondrial con­
TOMM20 levels, which was reversed by chloroquine, indicat­ tent by measuring COX4I1 and TOMM20 immunoblots.
ing mitophagic degradation. However, the BNIP3L monomer The BNIP3LG203A mutant construct, however, failed to
mutants were not able to reduce the mitochondrial contents reduce mitochondrial mass in OGD-treated neurons
(Figure 3C-F). MitoTracker Red was used to visualize the (Figure 4A,B). The neuron immunostaining showed the
mitochondrial mass after BNIP3L and mutant transfection same results (Fig. S5). These data provided evidence sup­
in HeLa cells (Fig. S4 C). Compared with BNIP3L, the mono­ porting the involvement of wild-type BNIP3L in mitophagy
mer mutants failed to decrease the mitochondrial area (Fig. activation in ischemia. This notion was further verified in
S4D). BNIP3L interacts with ATG8-family proteins (LC3 and ischemic mice brains by determining mitophagy following
GABARAP subfamilies) and thus induces mitophagy [15]. By virus-mediated expression. Results showed that GFP-
carrying out immunoprecipitation in HeLa cells with BNIP3L BNIP3L transduction to brains reversed the accumulation
gene deletion by CRISPR-Cas9, we recapitulated the interac­ of mitochondrial markers COX4I1 and TOMM20 after
tion of wild-type BNIP3L with either lipidated forms of LC3A pMCAO treatment. In contrast, GFP-BNIP3LG203A did not
or LC3B. However, the interaction of mutant BNIP3Ls with alter the mitochondrial mass in ischemic brains (Figure 4C,
unlipidated LC3s was weaker (Figure 3G,H). Taken together, D). These data from in vitro and in vivo ischemic models
these data indicated the incompetency of BNIP3 L monomer indicated that wild-type BNIP3L rescued mitophagy defi­
for inducing mitophagy and suggested the requirement of ciency in ischemic brains.
BNIP3L dimer to activate mitophagy. Our previous studies indicated the neuroprotective effects
We next asked whether wild-type BNIP3L may rescue of mitophagy against ischemic insults [6,31]. To further test
neuronal mitophagy deficiency in cerebral ischemia. After whether mitophagy rescued by wild-type BNIP3L may also
being subjected to OGD, the primary neurons transfected produce neuroprotective effects, AAV-GFP-Bnip3l and AAV-
1938 X. WU ET AL.

Figure 3. BNIP3L monomeric mutants failed to induce mitophagy in HeLa cells. (A) Structural diagram of mouse BNIP3L and mutants of the transmembrane (TM)
domain were shown. (B) HeLa cells were transfected with Flag, Flag-Bnip3l, Flag-Bnip3lS195A, Flag-Bnip3lG203A, or Flag-Bnip3lΔTM for 24 h. The relative protein levels in
HeLa cells were determined by western blot. Empty arrows indicate exogenous BNIP3L. (C) Hela cells were transfected with Flag, Flag-Bnip3l, Flag-Bnip3lS195A, Flag-
Bnip3lG203A, or Flag-Bnip3lΔTM for 24 h. Cells were treated with PBS as a control or 20 μM chloroquine for 4 h. The expression of Flag, COX4I1, TOMM20 and GAPDH
were determined by western blot. (D) Semi-quantitative analyses of COX4I1 and TOMM20 protein levels are shown (n = 3 from independent experiments). (E) HeLa
cells were transfected with Flag, Flag-Bnip3l, Flag-Bnip3lS195A, or Flag-Bnip3lG203A, Flag-Bnip3lΔTM for 24 h. Cells were treated with PBS as a control or 20 μM
chloroquine for 4 h and then incubated with CCCP (10 μM) for 6 h. The exogenous expression of BNIP3L and mitochondrial markers in HeLa cells were assessed by
western blot. (F) Semi-quantitative analyses of COX4I1 and TOMM20 protein levels are shown (n = 3 from independent experiments). (G-H) Protein-protein
interactions of MYC-LC3 with Flag, Flag-BNIP3L, Flag-BNIP3LS195A, Flag-BNIP3LG203A, Flag-BNIP3LΔLIR were confirmed by immunoprecipitation in BNIP3L KO HeLa
cells. Plasmids were co-transfected for 24 h. After 6 h of 10 μM CCCP incubation, the expression of LC3s and BNIP3L were assessed by western blot. Empty arrows
indicate exogenous BNIP3L. Semi-quantitative analyses of IP: MYC-LC3-I/IP: Flag are shown (n = 3 from independent experiments). Data are expressed as mean ±
SEM. Statistical comparisons were performed as follows: one-way ANOVA for (D, F, G and H). *P < 0.05; **P < 0.01; ***P < 0.001; n.s. vs. the indicated group.

GFP-Bnip3lG203A were separately injected into wild-type mice concluded that wild-type BNIP3L was required for its neuro­
brains. The ectopic BNIP3L expression produced significant protective effect against ischemic injury.
protection by reducing the brain infarct volumes from 51.9%
± 2.2% to 35.1% ± 2.5% (Figure 4E,F). This neuroprotection
was also reflected by improved neurological deficit score BNIP3L is degraded by ubiquitin-proteasome pathway
(NDS) (3.67 ± 0.21 vs. 2.57 ± 0.20, P < 0.01, Figure 4G)
24 h after ischemia onset. As a comparison, the Our data supported an intimate link of BNIP3L loss with
BNIP3LG203A failed to rescue the infarct volumes (51.9% ± mitophagy deficiency in cerebral ischemia. Multiple lines of
2.2% vs. 47.8% ± 2.5%, n.s., Figure 4F) as well as NDS evidence indicated that both autophagy-lysosome pathway
(3.67 ± 0.21 vs. 3.57 ± 0.20, n.s., Figure 4G). We thus and ubiquitin-proteasome pathway were activated in
AUTOPHAGY 1939

Figure 4. BNIP3L rescues mitophagy deficiency and protects against cerebral ischemia. (A) Primary cultured cortical neurons transfected with Flag, Flag-Bnip3l, or
Flag-Bnip3lG203A were subjected to 1 h of OGD. The exogenous expression of Flag, COX4I1, TOMM20 and GAPDH were determined by western blot. (B) Semi-
quantitative analyses of COX4I1 and TOMM20 are shown (n = 3 from independent experiments). (C) Mice were infected with AAV-GFP, AAV-GFP-Bnip3l or AAV-GFP-
Bnip3lG203A for at least two weeks and then subjected to 6 h of pMCAO. The AAVs, COX4I1 and TOMM20 protein levels in brain tissues were determined by western
blot. Empty arrows indicate exogenous BNIP3L. Duplicate lanes are shown for each grou(D) Semi-quantitative analyses of COX4I1 and TOMM20 are shown (n = 3
mice for each group). (E) Mice brains were infected with AAVs expressing GFP, GFP-Bnip3l, or GFP-Bnip3lG203A and then subjected to 24 h of pMCAO. The
representative TTC-stained brain slices from each group are shown. (F) The brain infarct volumes and (G) neurological deficit scores of each group were determined
(n = 6–7 mice for each group). Data are expressed as mean ± SEM. Statistical comparisons were performed as follows: one-way ANOVA for (B, D, F and G). **P < 0.01;
***P < 0.001; n.s. vs. the indicated group.

ischemic brains. We thus blocked these two degradation being ubiquitinated. Further studies are needed to address
pathways by 3-MA and MG132, respectively. The LC3B-II this issue.
protein levels were inhibited by 3-MA. MG132 treatment led We next determined whether wild-type BNIP3L was ubi­
to an increase in level of TRP53/p53 [40], suggesting protea­ quitylated in ischemic brains. To this end, mice brains were
some inhibition (Figure 5A). The proteasome inhibitor injected with viruses to express the wild-type or monomer
MG132, rather than autophagy inhibitor 3-MA, rescued BNIP3L in bnip3l-/- mice. After being subjected to pMCAO,
wild-type BNIP3L degradation. This result suggested that ubiquitination of BNIP3Ls was assayed using immunopreci­
that proteasomal activation was involved in wild-type pitation. Both BNIP3L dimer and monomer were ubiquiti­
BNIP3L loss. To investigate this, we determined the ubiqui­ nated, and the ubiquitylation was further increased with
tination of wild-type BNIP3L and its mutants by immuno­ MG132 treatment (Figure 5D). Additionally, MG132 pre­
precipitation in BNIP3L-/- HeLa cells. Wild-type BNIP3L vented BNIP3L loss in ischemic mice brains when adminis­
was more ubiquitylated compared with its monomer tered systemically. Rescue of BNIP3L loss was accompanied
mutants (Figure 5B,C). We cannot exclude the possibility by enhanced mitophagy as revealed by decreased levels of
that the smear of ubiquitin chains on wild-type BNIP3L COX4I1 and TOMM20 (Figure 5E). Taken together, these
comes from other proteins interacting with BNIP3L and data indicated that BNIP3L was degraded by the ubiquitin-
1940 X. WU ET AL.

Figure 5. BNIP3L is degraded by the ubiquitin-proteasome pathway. (A) The mice were injected (i.c.v.) with 3-MA (7.5 μg) or MG132 (4 mg/kg) at the onset of
occlusion and then subjected to 6 h of pMCAO. The BNIP3L, LC3 and TP53 protein levels were determined by western blot. (B-C) Protein-protein interactions of MYC-
Ub with Flag-BNIP3L, Flag-BNIP3LS195A, Flag-BNIP3LG203A, or Flag-BNIP3LΔTM were confirmed by immunoprecipitation in BNIP3L KO HeLa cells. Plasmids were co-
transfected for 24 h. CCCP (10 μM) and MG132 (10 μM) were incubated for 6 h before harvesting. The protein levels of ubiquitin and exogenous expression of BNIP3L
were detected by using anti-Flag and anti-MYC antibodies, respectively. Empty arrows indicate exogenous BNIP3L. (D) The bnip3l-/- mice were infected with AAVs
expressing GFP, GFP-Bnip3l, or GFP-Bnip3lG203A for two weeks and then subjected to 6 h of pMCAO. The protein-protein interactions of ubiquitin and overexpression
of BNIP3L in vivo were determined by immunoprecipitation. The ubiquitylation levels of exogenous BNIP3L were detected by using anti-ubiquitin and anti-BNIP3L
antibodies. A ratio t-test was applied (n = 3 from independent experiments). (E) The wild-type mice treated with MG132 (4 mg/kg) at the onset of occlusion were
subjected to 6 h of pMCAO. Duplicate lanes are shown for each group. The BNIP3L, LC3B, COX4I1 and TOMM20 protein levels were assessed by western blot. Black
arrows indicate endogenous BNIP3L. Data are expressed as mean ± SEM. Statistical comparisons were performed as follows: t-test for (D). **P < 0.01 vs. the indicated
group.

proteasomal pathway and thus led to mitophagy deficiency in ubiquitylated BNIP3L (Figure 6B). These data strengthened
ischemic brains. the notion that BNIP3L was ubiquitylated in ischemic brains.
We next determined the neuroprotective effect of CFZ in the
pMCAO model. CFZ (2 mg/kg) was injected intraperitoneally
at the onset of ischemia. The infarct volumes and NDS were
Carfilzomib rescues BNIP3L degradation and protects
determined 24 h after injury. Treatment with CFZ signifi­
against ischemic injury
cantly reduced the brain infarct volumes (50.6% ± 1.6% to
The fact that BNIP3L degradation by proteasomes contributed 31.2% ± 3.9%, P < 0.001) and NDS (3.62 ± 0.18 vs. 2.50 ± 0.19,
to mitophagy deficiency in ischemic brains led us to hypothe­ P < 0.05, Figure 6C,D). Importantly, neuroprotection from
size that proteasome inhibitors may serve to prevent BNIP3L CFZ treatment was almost abolished in bnip3l-/- mice, sug­
loss and thus alleviate ischemic brain injury. Some protea­ gesting that BNIP3 L was required for the therapeutic effects
some inhibitors are used in multiple myeloma therapy [41]. of CFZ.
Among these drugs, carfilzomib (CFZ) showed advantages in Short therapeutic time-window is a major limitation of
pharmacokinetics and safety [42]. We confirmed that BNIP3L anti-stroke drugs. To address the time-window of CFZ for
degradation caused by pMCAO was blocked by CFZ treat­ acute stroke therapy, CFZ was administered to mice at 3 or
ment. As a consequence, the administration of CFZ reduced 6 h after ischemia onset. Our results showed that administra­
the COX4l1 and TOMM20 protein levels, indicating the tion of CFZ at 3 h after pMCAO was still competent to
restoration of mitophagy deficiency (Figure 6A). alleviate brain infarct volumes (49.3% ± 1.1% to 28.0% ±
CFZ also led to accumulation of total ubiquitin in ischemic 3.4%, P < 0.001) as well as NDS (3.56 ± 0.20 vs. 2.71 ± 0.28,
brains. In particular, after pull-down of BNIP3L in ischemic P < 0.05, Figure 6E,F), but delayed administration at 6 h
brains, we found that CFZ treatment increased the level of abolished the benefits of CFZ treatment. These results
AUTOPHAGY 1941

Figure 6. Carfilzomib (CFZ) rescues BNIP3L degradation and protects against ischemic injury. (A) Mice treated with 2 mg/kg carfilzomib at the onset of occlusion were
subjected to 6 h of pMCAO. The protein levels of BNIP3L, SQSTM1, LC3B, COX4I1 and TOMM20 were detected by western blot. Duplicate lanes are shown for each
group. (B) Wild-type or bnip3l-/- mice were injected with CFZ at the onset of occlusion and then subjected to 6 h of pMCAO. The protein-protein interactions of
BNIP3L and ubiquitin after CFZ treatment were detected by immunoprecipitation in vivo. The ubiquitylation levels of endogenous BNIP3L were detected using anti-
ubiquitin and anti-BNIP3L antibodies. A ratio t-test was applied (n = 3 from independent experiments). (C) Wild-type or bnip3l-/- mice were given CFZ at the onset of
occlusion and subjected to pMCAO for 24 h. The brain slices were stained by TTC and representative slices from each group are shown. (D) The brain infarct volumes
and neurological deficit scores of each group were determined (n = 6–8 mice for each group). (E) The mice were treated with CFZ at 3 or 6 h after the onset of
occlusion. Representative brain slices are shown. (F) After 24 h of occlusion, the infarct volumes were determined by TTC staining and neurological deficit scores were
evaluated (n = 6–7 mice for each group). (G) The protein levels of BNIP3L, SQSTM1, LC3B, COX4I1 and TOMM20 were determined by western blot. Duplicate lanes are
shown for each grouBlack arrows indicate endogenous BNIP3L. Data are expressed as mean ± SEM. Statistical comparisons were performed as follows: t-test for (B);
one-way ANOVA for (D and F). *P < 0.05; **P < 0.01; ***P < 0.001; n.s. vs. the indicated group.

suggested that the effective time-window of CFZ for stroke after stroke. Taken together, these data suggested that CFZ
therapy may extend to at least 3 h post-ischemia. We identi­ could be a promising drug for stroke.
fied the BNIP3L in ischemic brains and found that delayed
administration of CFZ at 3 h, but not 6 h, rescued BNIP3L
Discussion
loss as well as mitophagy deficiency (Figure 6G). This obser­
vation further emphasized that BNIP3L is associated with In the present study, we identified a previously undetermined
effective mitophagy in ischemic brains. link between BNIP3L degradation and mitophagy deficiency
We further determined whether CFZ treatment may also in ischemic brains. BNIP3L dimer underwent a time-
attenuate chronic ischemic brain injury. To do this, mice were dependent decrease along with accumulation in ischemic
subjected to photothrombosis (PT) [43] and their motor func­ brains (Figure 2E). Moreover, ectopic expression of wild-
tion was measured every other day based on a grid-walking type BNIP3L, but not its monomer, reversed mitophagy defi­
task and forelimb symmetry in the cylinder task over the next ciency (Figures 3 and 4). Of note, neuronal autophagy was not
13 d. We found that CFZ treatment (2 mg/kg, i.p.) every impaired by ischemia (Figure 1), further suggesting that mito­
other day significantly reduced the number of foot faults phagy deficiency was not caused by defective autophagy
and the forelimb asymmetry during the 13 d after PT onset machinery. We previously found that mitophagy was acti­
(Fig. S6A and S6B). The brain infarct volumes were detected vated in brains that underwent ischemia-reperfusion (I-R) in
at 13 d after PT using toluidine blue stain. The brain infarct which BNIP3L dimer was not reduced [14]. Conversely,
volumes were significantly decreased (saline 0.85 ± 0.07 mm3 bnip3l-/- caused mitophagy deficiency in I-R [6]. Prior to
vs. CFZ 0.59 ± 0.04 mm3, P < 0.05, Fig. S6 C and S6D), this study, little was known about the mechanisms of neuro­
indicating that CFZ treatment could reduce infarct volumes nal mitophagy deficiency under ischemic conditions [44,45].
1942 X. WU ET AL.

Based on our results, we concluded that BNIP3L degradation present study emphasized the previously underestimated roles
played a causative role in mitophagy deficiency of ischemic of BNIP3L dimer in mitophagy activation and advanced our
brains. understanding of how BNIP3L serves to protect ischemic
Another finding of the present study is that BNIP3L mono­ brain injury [6]. Therefore, promoting BNIP3L dimer forma­
mer mutants failed to induce mitophagy. Although BNIP3L is tion in ischemic brains could be a promising strategy in
present as both a dimer and monomer in cells, their respective rescuing ischemic brain injury, which should be addressed
contributions to mitophagy remained largely unexplored. As in future studies.
a result, BNIP3L was unequivocally determined to be Ubiquitination has been shown to engage in mitophagy
a mitophagy receptor in most studies [46,47]. Previous regulation in ischemic neurons [55,56] but the detailed
research indicated that BNIP3, the homolog of BNIP3L [48], mechanisms have not been fully illustrated. Here we showed,
formed a homodimer to promote autophagic flux and cell for the first time, that BNIP3L underwent proteasome-
death [49]. Paradoxically, it remained undetermined whether dependent rather than autophagy-dependent degradation.
BNIP3L dimer would be indispensable for its biological func­ Ubiquitylated BNIP3L, particularly in its dimeric form, accu­
tions [24]. BNIP3L dimer loss showed a much stronger cor­ mulated after ischemia (Figure 5), but the ubiquitination sites
relation with mitophagy deficiency than its monomeric form and corresponding E3 ligases for BNIP3L degradation should
in ischemic brains (Figure 2H,I). Furthermore, the single site- be further addressed. Importantly, proteasomal inhibitors,
specific mutations (S195A, G203A) of the TM domain in MG132 and carfilzomib prevented BNIP3L degradation as
BNIP3L formed only monomer and were sufficient to abolish well as mitophagy deficiency (Figures 5E and 6A). The
the mitophagy activation (Figure 3C-F). BNIP3L monomer BNIP3L loss-induced mitophagy deficiency was significant
failed to rescue mitophagy deficiency in ischemic neurons and for ischemic neuronal injury, since restored BNIP3L attenu­
brains (Figure 4A-D). Our results highlighted that the mono­ ated brain infarct volumes and NDS (Figure 4E-G). We thus
mer mutants (S195A and G203A) were not sufficient to hypothesized that preventing BNIP3L degradation could be
induce mitophagy. Neurons prohibit mitophagy by degrading a novel therapy for cerebral ischemia. As expected, a single
BNIP3L and may restore mitophagy by allowing homodimer­ dosage of carfilzomib at the onset of ischemia showed signifi­
ization of the remaining BNIP3L monomers. This mechanism cant neuroprotection in the MCAO model.
may provide a way for ischemic neurons to promptly control Carfilzomib is a selective proteasomal inhibitor approved
mitophagy. In addition, although mutant BNIP3L (S195A and by the FDA for multiple myeloma [57]. Carfilzomib showed
G203A) showed only the monomeric form, we cannot exclude improved safety profiles over bortezomib, another proteaso­
the possibility that the mutants may have effects besides pre­ mal inhibitor with profound neurotoxicity [58]. Some other
venting dimerization. Moreover, a dramatic drop in intracel­ studies indicated the neuroprotection of proteasome inhibi­
lular ATP or glycolysis perturbation, as a stress caused by tors [59,60], but it was not fully understood how proteasome
ischemia, may also block mitophagy [50–52]. Although we inhibition attenuated ischemic injury [61,62]. We found that
found BNIP3L loss was attributed to proteasomal degrada­ BNIP3L deletion abolished the neuroprotection of carfilzomib
tion, it was not clear whether this degradation was triggered (Figure 6C,D), indicating that BNIP3L-mediated mitophagy
by energy crisis. It remains unclear whether and how glucose was the prominent mechanism, if not the only mechanism,
metabolism alterations and BNIP3L degradation work underlying the benefits of proteasomal inhibitors. We cannot
together to regulate neuronal mitophagy in ischemia-related exclude that other pathways induced by proteasomal blockage
scenarios. may also contribute to neuronal survival besides mitophagy
Detailed mechanisms of BNIP3L homodimer in triggering induction. For example, proteasomal inhibition altered neu­
mitophagy were not fully understood. An emerging study ronal glucose metabolism, which may also promote neuronal
added a mechanistic perspective to our findings that survival [63]. Surprisingly, we also found that the administra­
BNIP3L dimerization may associate with its phosphorylation tion of carfilzomib as late as 3 h after ischemia onset was still
in TM domain [53]. Mutations in the TM domain did not effective in rescuing brain injury. Prolonged administration of
alter their mitochondrial location (Fig. S4A and S4B). We carfilzomib to 6 h failed to alleviate brain injury (Figure 6E,F).
confirmed that the mitochondrial distribution of BNIP3L Coincidently, BNIP3L cannot be restored by carfilzomib at
was required (compared with the TM domain deletion muta­ 6 h after ischemia, which further strengthened the significance
tion) for mitophagy. However, here we showed that anchoring of BNIP3L-mediated mitophagy in ischemic brains. Finally,
to mitochondria was not sufficient for BNIP3L to serve as we identified the efficacy of carfilzomib in the chronic ische­
a mitophagy receptor. Remarkably, BNIP3L homodimer was mia model (Fig. S6). Although more complicated pathophy­
prone to interact with LC3A/B-II and the mutants also had siological mechanisms may be involved, a variety of studies
a weaker interaction with LC3A/B-I (Figure 3E,F). It is likely indicated the importance of neuronal mitochondrial quality in
that the homodimer of BNIP3L might alter the conformation the late phases after stroke [64]. Overall, this is the first
of its LIR motif, which faces LC3A/B-II proteins and thus demonstration of carfilzomib as a promising therapy for
facilitates recognition of mitochondria by autophagosomes. In diverse states of stroke.
addition to its function in inducing mitophagy, we revealed Taken together, our work has unraveled critical contribu­
the requirement of BNIP3L homodimer for its neuroprotec­ tions of BNIP3L degradation to compromised mitophagy in
tive effect in ischemic brains (Figure 4E-G). The neuropro­ ischemic brains. Preventing BNIP3L degradation by inhibiting
tective feature of BNIP3L was distinct from that of BNIP3, the ubiquitin-proteasome pathway rescued mitophagy defi­
which promotes cell death in its monomeric form [54]. The ciency and attenuated cerebral ischemic injury. We identified
AUTOPHAGY 1943

carfilzomib, a proteasomal inhibitor, as a promising strategy Cell culture, OGD/O-R procedures


for stroke therapy.
For culturing primary cortical neurons, pregnant mice with
embryonic (E17) fetuses were used. The primary cortical
neuronal culture experiments were performed as described
Materials and methods previously [31]. Briefly, the cortical neurons of fetal mice
Animals were digested with 0.25% trypsin (Invitrogen, 25,200–056).
Approximately 2 × 105 cells/cm2 were seeded onto poly-
Male C57BL/6 mice and male BALB/c mice weighing 22–25 g L-lysine (10 μg/ml)-coated plates and dishes. The neurons
(8–10 weeks old) were used. Mice were raised on a 12 h light/ were cultured in Neurobasal medium (Invitrogen, 21,103–­
dark cycle with free access to water and food. The bnip3l-/- 049) containing 2% B27 (Invitrogen,17,504–044), 10 U/ml
mice (BALB/c strain background) were provided by Prof. Paul penicillin-streptomycin (Gibco, 15,140–122) and 0.5 mmol/L
Ney (St. Jude Children’s Research Hospital). The Atg7fl/fl mice glutamine (Gibco, 25,030,081) at 37°C in a humidified atmo­
were kindly provided by Masaaki Komatsu (Tokyo sphere with 5% CO2 + 95% N2. Cultures were maintained for
Metropolitan Institute of Medical Science, Tokyo, Japan). All 7 d before further treatment and were routinely observed
experiments were approved by and conducted in accordance under a phase-contrast inverted microscope. To produce
with the ethical guidelines of the Zhejiang University Animal atg7 knockout neurons, pregnant Atg7fl/fl mice with embryo­
Experimentation Committee and were in complete compli­ nic (E17) fetuses were used. At the fifth day, Atg7fl/fl cortical
ance with the National Institutes of Health Guide for the Care neurons (2 × 106) were transfected with hSyn-Cre virus (2 μl;
and Use of Laboratory Animals. Efforts were made to reduce Obio Technology Crop., H4942). Neurons were maintained
any pain and the number of animals required. for 2 d before further treatment.
For OGD treatment, cells were washed twice with glucose-
free DMEM (Invitrogen, 12,800–017), and then refreshed with
MCAO mouse models and drug administration O2- and glucose-free DMEM (pre-balanced in an O2-free
chamber at 37°C). Then, we put the neurons in a sealed
Mice were anesthetized with isoflurane during surgery.
chamber (Billups Rothenburg, MIC-101) ventilated with
Cerebral blood flow (CBF) was detected in the middle cerebral
mixed gas (5% CO2 + 95% N2) for 7 min at 25 L/min. The
artery (MCA) by laser Doppler flowmetry (Model Moor
chambers were sealed and incubated for 2 h at 37°C. OGD-
VMS-LDF2, Moor Instruments Ltd, UK). A fiber-optic
reperfusion (O-R) was performed by adding oxygen and glu­
probe was fixed to the skull from 2-mm caudal to bregma
cose to the culture medium for 1 h at 37°C.
and 6-mm lateral to midline over the cortex supplied by the
proximal part of the right MCA. A 6–0 nylon monofilament
suture, blunted at the tip and coated with 1% poly-l-lysine, BNIP3L gene knockout using CRISPR-Cas9 system in HeLa
was inserted 10 mm into the internal carotid to occlude the cells
origin of the MCA. Animals were excluded from the study if
the reduction in CBF was less than 80%. Body temperature To knockout BNIP3L, two gRNAs (5ʹ-
was maintained at 37°C by a heat lamp (FHC, Bowdoinham, CGGCGGCGGCTCGACTAGGT-3ʹ, 5ʹ-GCGGCGGCGGCTC
ME, USA) until the mice woke uFor transient MCAO, reper­ GACTAGG-3ʹ) targeting the adjacent downstream sequence
fusion was allowed 1 h after ischemia by gently removing the of its start codon were designed using http://crispr.mit.edu/
monofilament suture. Focal cerebral ischemia was induced by and inserted into a pSpCas9(BB)-2A-GFP plasmid (Addgene,
MCAO without reperfusion. 48,138; deposited by Liang Fang) according to a previously
Mice were given an intracerebroventricular (i.c.v.) injection described protocol [65]. HeLa cells (ATCC, CCL-2) were co-
of 7.5 μg 3-MA (Sigma, M9281) at the onset of ischemia in transfected with two plasmids using Lipofectamine 2000
pMCAO as we described previously [7]. MG132 (Selleckchem, (Thermo Fisher Scientific, 11,668,027), and 48 h after trans­
S2619) was injected (4 mg/kg, i.c.v.) at the onset of ischemia. fection, GFP+ cells were FAC-sorted into 96-well plates as
CFZ (Selleckchem, S2853) was also injected intraperitoneally single cells to generate mutant cell clones. The successful
(2 mg/kg, i.p.) at ischemia onset and at 3 h or 6 h after knockout was validated using western blot.
ischemia. Control mice were injected with the same volume
of saline.
Transfection
Neurological deficit scores were evaluated at 24 h after
MCAO as follows: 0, no deficit; 1, flexion of contralateral p3× Flag-Bnip3l, and p3× Flag-Bnip3lG203A were transfected
forelimb upon lifting the whole animal by the tail; 2, circling into primary cultured neurons using Amaxa electroporation
to the contralateral side; 3, falling to the contralateral side; and (Lonza, VPG-1001) as described previously [14]. Briefly, the
4, no spontaneous motor activity. primary neurons (5 × 106) were electroporated with 3 μg
Mice were sacrificed using a lethal dose of isoflurane at plasmids. The cuvette with cell/DNA suspension was inserted
24 h after surgery and brain sections were stained with 2,3,5, - into the Nucleofector Cuvette Holder. After electroporation,
triphenyltetrazolium chloride (TTC; 0.25%; Sigma, T8877). the cells were resuspended in the medium mentioned above
Brain infarct volumes were quantified with ImageJ software and then transferred into the prepared culture dishes. The
and determined by an indirect method that corrects for dishes were maintained in an incubator for 7 d at 37°C. The
edema [6]. plasmids were transfected into wild-type HeLa and BNIP3L
1944 X. WU ET AL.

KO HeLa according to the jet PRIME (Polyplus, nl14-15) A Sepharose (GE Healthcare, 17,078,001) was added for
protocol. 4 h at 4°C.
For endogenous BNIP3 L IP, bnip3l-/- mice were injected
with AAVs expressing GFP, GFP-Bnip3l, or GFP-Bnip3lG203A
Lentivirus injection in vivo
for at least two weeks and then were subjected to 6 h of
Mice were anesthetized with isoflurane and mounted in pMCAO. The brain tissues were homogenized in RIPA buffer
a stereotaxic apparatus (Stoelting, 512,600) and adeno- with protease inhibitor cocktail. The suspensions were cen­
associated virus (AAV, designed, produced and identified by trifuged at 12,000 g for 20 min. Part of the supernatant
Obio Technology Crop., Ltd., Shanghai, China) including (300 μg) was transferred to a new tube as input. In total,
AAV-GFP, AAV-GFP-Bnip3l, and AAV-GFP-Bnip3lG203A 20 μl Protein A Sepharose was added to the supernatant for
(1 μl) were injected into the cortex (AP + 0.02 mm; the preclear process for 4 h and was subsequently discarded.
L − 3.2 mm; V − 1.5 mm) and corpus striatum (AP + The supernatant (1.5–3 μg) was incubated with 4–8 μl anti-
0.5 mm; L − 2.0 mm; V − 3.0 mm) with a 5-μl syringe and BNIP3L (Cell Signaling Technology, 12396s) overnight at 4°
a 34 gauge needle at 100 nl/min using an injection pump C. Then 50 μl of Protein A Sepharose was added for 4 h at 4°
(Micro 4, WPI, Sarasota, Fl, USA). After each injection, the C. Subsequently, the immunoprecipitates were washed with
needle was left in place for an additional 10 min and then lysis buffer 3 times. The immunoprecipitates were then eluted
slowly withdrawn. The virus was allowed to express target by boiling for 5 min in 2 × Laemmli sample buffer (Sigma-
proteins for a minimum of 2 weeks. Aldrich, S3401) and subjected to western blot analysis.

Immunoblotting and immunoprecipitation Statistical analysis


Brain tissues and cells were homogenized in RIPA buffer All data were collected and analyzed in a blind manner. Data
(50 mM Tris, pH 7.4, 150 mM NaCl, 1% Triton X-100 are presented as mean ± SEM. The single comparisons were
[Diamond, A110694], 1% sodium deoxycholate [Sigma- determined by a two-tailed Student’s t-test. One-way ANOVA
Aldrich, 30,970], 0.1% SDS [Biofroxx, 3250GR500]) with (analysis of variance) with Dunnett’s T3 post-hoc test applied
protease inhibitor cocktail (Roche, 04693132001). The sam­ for multiple comparisons. Sample sizes were based on our
ples come from three independent experiments. A 40 μg ali­ previous study and prior literature to achieve reliable mea­
quot of protein from each sample was separated using SDS- surements. Statistical analyses were conducted using
PAGE. The following primary antibodies were used: ATG7 GraphPad Prism 5.0 (GraphPad Software, San Diego, CA,
(1:1,000; Cell Signaling Technology, 8558 T), BCL2L13 USA). P < 0.05 was considered statistically significant.
(1:1000; Proteintech; 16,612-1-AP), BNIP3L (1:1,000; Cell
Signaling Technology, 12396s), COX4I1 (1:1,000; Cell
Signaling Technology, 4850), Flag (1:1,000; Cell Signaling Acknowledgments
Technology, 14793s), FUNDC1 (1:1,000; Abcam, ab74834), We are grateful to Prof. Paul Ney from St. Jude Children’s Research
GAPDH (1:3,000; KangChen, KC-5G4), GFP (1:1,000; MBL; Hospital for offering the bnip3l-/- mice and Professor Masaaki Komatsu
598), LC3B (1:1,000; Sigma-Aldrich, L7543), MYC (1:1,000; of Tokyo Metropolitan Institute of Medical Science for offering the
MBL; M192-3), PHB2/prohibitin 2 (1:1000; Proteintech; Atg7fl/fl mice. We are grateful to the Core Facilities of Zhejiang
12,295-1-AP); PRKN/PARK2 (1:1,000; Cell Signaling University Institute of Neuroscience and Imaging Facilities.
Technology, 2132s), SQSTM1 (1:1,000; Cell Signaling
Technology, 5114), TOMM20 (1:1,000; Ambo Disclosure statement
Biotechnology, c16678), TP53 (1:1000; Cell Signaling
Technology, 2524 T) and ubiquitin (1:1000, Cell Signaling No potential conflicts of interest are disclosed.
Technology, 3936s). Secondary antibodies conjugated with
HRP against either rabbit or mouse IgG (1:3,000; Cell Funding
Signaling Technology, 7071 and 7072) were applied. Digital
images were quantified using densitometric measurement This work was funded by the National Natural Science Foundation of
with Quantity-One software (Bio-Rad). China (81822044, 81773703 and 81872844) and the Fundamental
Research Funds for the Central Universities (2019XZZX004-17)
For immunoprecipitation, BNIP3L KO HeLa cells were
transiently transfected. At 24 h post-transfection, the cells
were suspended with the immunoprecipitation (IP) lysis ORCID
buffer (50 mM Tris, pH 7.4, 150 mM NaCl, 1% NP-40
Shijia Ma http://orcid.org/0000-0001-9409-9272
(Sangon Biotech, A100109), 0.25% sodium deoxycholate) Liang Fang http://orcid.org/0000-0003-4502-1756
with protease inhibitor cocktail and immunoprecipitated Zhong Chen http://orcid.org/0000-0003-4755-9357
using EZview Red ANTI-Flag M2 Affinity Gel beads Xiangnan Zhang http://orcid.org/0000-0002-7603-7403
(Sigma-Aldrich, F2426). Then the immunoprecipitates
were eluted with 100 μl 3× Flag peptide solution (Sigma-
Aldrich, F4799). For MYC IP, the supernatant was incu­ References
bated with 4 μl anti-MYC (MBL; M192-3) and mixed [1] Gallacher KI, Jani BD, Hanlon P, et al. Multimorbidity in stroke.
overnight with gentle rotation at 4°C. Then 50 μl protein Stroke. 2019;50(7):1919–1926
AUTOPHAGY 1945

[2] Wang W, Jiang B, Sun H, et al. Prevalence, incidence, and mor­ [24] Imazu T, Shimizu S, Tagami S, et al. Bcl-2/E1B 19 kDa-interacting
tality of stroke in china: results from a nationwide protein 3-like protein (Bnip3L) interacts with bcl-2/Bcl-xL and
population-based survey of 480 687 adults. Circulation. 2017;135 induces apoptosis by altering mitochondrial membrane
(8):759–771 permeability. Oncogene. 1999;18(32):4523–9
[3] Kislin M, Sword J, Fomitcheva IV, et al. Reversible disruption of [25] Chen G, Cizeau J, Vande Velde C, et al. Nix and Nip3 form
neuronal mitochondria by ischemic and traumatic injury revealed a subfamily of pro-apoptotic mitochondrial proteins. J Biol
by quantitative two-photon imaging in the neocortex of anesthe­ Chem. 1999;274(1):7–10
tized mice. J Neurosci. 2017;37(2):333–348 [26] Rodrigo R, Mendis N, Ibrahim M, et al. Knockdown of BNIP3L or
[4] Anzell AR, Maizy R, Przyklenk K, et al. Mitochondrial quality SQSTM1 alters cellular response to mitochondria target drugs.
control and disease: insights into ischemia-reperfusion injury. Autophagy. 2019;15(5):900–907
Mol Neurobiol. 2018;55(3):2547–2564 [27] Esteban-Martinez L, Sierra-Filardi E, McGreal RS, et al.
[5] Um JH, Yun J. Emerging role of mitophagy in human diseases Programmed mitophagy is essential for the glycolytic switch dur­
and physiology. BMB Rep. 2017;50(6):299–307. ing cell differentiation. Embo J. 2017;36(12):1688–1706
[6] Yuan Y, Zheng Y, Zhang X, et al. BNIP3L/NIX-mediated mito­ [28] Klionsky DJ, Abdelmohsen K, Abe A, et al. Guidelines for the use
phagy protects against ischemic brain injury independent of and interpretation of assays for monitoring autophagy (3rd
PARK2. Autophagy. 2017;13(10):1754–1766 edition). Autophagy. 2016;12(1):1–222.
[7] Zhang X, Yan H, Yuan Y, et al. Cerebral ischemia-reperfusion- [29] Komatsu M, Waguri S, Ueno T, et al. Impairment of
induced autophagy protects against neuronal injury by mitochon­ starvation-induced and constitutive autophagy in Atg7-deficient
drial clearance. Autophagy. 2013;9(9):1321–33 mice. J Cell Biol. 2005;169(3):425–34
[8] Yuan Y, Zhang X, Zheng Y, et al. Regulation of mitophagy in [30] McWilliams TG, Prescott AR, Allen GFG, et al. mito-QC illumi­
ischemic brain injury. Neurosci Bull. 2015;31(4):395–406 nates mitophagy and mitochondrial architecture in vivo. J Cell
[9] Galluzzi L, Bravo-San Pedro JM, Blomgren K, et al. Biol. 2016;214(3):333–45
Autophagy in acute brain injury. Nat Rev Neurosci. [31] Shen Z, Zheng Y, Wu J, et al. PARK2-dependent mitophagy
2016;17(8):467–84 induced by acidic postconditioning protects against focal cerebral
[10] Tang YC, Tian H-X, Yi T, et al. The critical roles of mitophagy in ischemia and extends the reperfusion window. Autophagy.
cerebral ischemia. Protein Cell. 2016;7(10):699–713 2017;13(3):473–485
[11] Sliter DA, Martinez J, Hao L, et al. Parkin and PINK1 mitigate [32] Liu L, Feng D, Chen G, et al. Mitochondrial outer-membrane
STING-induced inflammation. Nature. 2018;561(7722):258–262 protein FUNDC1 mediates hypoxia-induced mitophagy in mam­
[12] Yin W, Signore AP, Iwai M, et al. Rapidly increased neuronal malian cells. Nat Cell Biol. 2012;14(2):177–85
mitochondrial biogenesis after hypoxic-ischemic brain injury. [33] Wei Y, Chiang W-C, Sumpter R, et al. Prohibitin 2 is an inner
Stroke. 2008;39(11):3057–63 mitochondrial membrane mitophagy receptor. Cell. 2017;168(1–­
[13] Zuo W, Liu Z, Yan F, et al. Hyperglycemia abolished 2):224–238 e10
Drp-1-mediated mitophagy at the early stage of cerebral [34] Otsu K, Murakawa T, Yamaguchi O. BCL2L13 is a mammalian
ischemia. Eur J Pharmacol. 2019;843(p):34–44 homolog of the yeast mitophagy receptor Atg32. Autophagy.
[14] Zheng Y, Zhang X, Wu X, et al. Somatic autophagy of axonal 2015;11(10):1932–3.
mitochondria in ischemic neurons. J Cell Biol. 2019;218 [35] Kuramori C, Azuma M, Kume K, et al. Capsaicin binds to prohi­
(6):1891–1907 bitin 2 and displaces it from the mitochondria to the nucleus.
[15] Novak I, Kirkin V, McEwan DG, et al. Nix is a selective autophagy Biochem Biophys Res Commun. 2009;379(2):519–25
receptor for mitochondrial clearance. EMBO Rep. 2010;11 [36] Jiang D, Sun X, Wang S, et al. Upregulation of miR-874-3p
(1):45–51 decreases cerebral ischemia/reperfusion injury by directly target­
[16] Schweers RL, Zhang J, Randall MS, et al. NIX is required for ing BMF and BCL2L13. Biomed Pharmacother. 2019;117:108941.
programmed mitochondrial clearance during reticulocyte [37] Wu W, Tian W, Hu Z, et al. ULK1 translocates to mitochondria
maturation. Proc Natl Acad Sci U S A. 2007;104(49):19500–5 and phosphorylates FUNDC1 to regulate mitophagy. EMBO Rep.
[17] Zhang X, Yuan Y, Jiang L, et al. Endoplasmic reticulum stress 2014;15(5):566–75
induced by tunicamycin and thapsigargin protects against transi­ [38] Nakamura Y, Kitamura N, Shinogi D, et al. BNIP3 and NIX
ent ischemic brain injury: involvement of PARK2-dependent mediate Mieap-induced accumulation of lysosomal proteins
mitophagy. Autophagy. 2014;10(10):1801–13 within mitochondria. PLoS One. 2012;7(1):e30767
[18] Lampert MA, Orogo AM, Najor RH, et al. BNIP3L/NIX and [39] Sulistijo ES, MacKenzie KR. Sequence dependence of BNIP3
FUNDC1-mediated mitophagy is required for mitochondrial net­ transmembrane domain dimerization implicates side-chain
work remodeling during cardiac progenitor cell differentiation. hydrogen bonding and a tandem GxxxG motif in specific
Autophagy. 2019;15(7):1182–1198 helix-helix interactions. J Mol Biol. 2006;364(5):974–90.
[19] Ma J, Ni H, Rui Q, et al. Potential roles of NIX/BNIP3L pathway [40] Ding WX, Ni HM, Chen X, et al. A coordinated action of Bax, PUMA,
in rat traumatic brain injury. Cell Transplant. 2019;28(5):585-595. and p53 promotes MG132-induced mitochondria activation and apop­
[20] Zhang M, Shi R, Zhang Y, et al. Nix/BNIP3L-dependent mito­ tosis in colon cancer cells. Mol Cancer Ther. 2007;6(3):1062–9
phagy accounts for airway epithelial cell injury induced by cigar­ [41] Gandolfi S, Laubach JP, Hideshima T, et al. The proteasome and
ette smoke. J Cell Physiol. 2019;234(8):14210–14220 proteasome inhibitors in multiple myeloma. Cancer Metastasis
[21] Lazarini M, Machado-Neto JA, Duarte ADSS, et al. BNIP3L in Rev. 2017;36(4):561–584
myelodysplastic syndromes and acute myeloid leukemia: impact [42] Vij R, Wang M, Kaufman JL, et al. An open-label, single-arm,
on disease outcome and cellular response to decitabine. phase 2 (PX-171-004) study of single-agent carfilzomib in
Haematologica. 2016;101(11):e445–e448 bortezomib-naive patients with relapsed and/or refractory multi­
[22] Yasuda M, Han J, Dionne C, et al. BNIP3alpha: a human homolog ple myeloma. Blood. 2012;119(24):5661–70
of mitochondrial proapoptotic protein BNIP3. Cancer Res. [43] Lin Y-H, Dong J, Tang Y, et al. Opening a new time window for
1999;59(3):533–7. treatment of stroke by targeting HDAC2. J Neurosci. 2017;37
[23] Yussman MG, Toyokawa T, Odley A, et al. Mitochondrial death (28):6712–6728
protein Nix is induced in cardiac hypertrophy and triggers apop­ [44] Zhu M-Y, Zhang D-L, Zhou C, et al. Mild acidosis protects
totic cardiomyopathy. Nat Med. 2002;8(7):725–30 neurons during oxygen-glucose deprivation by reducing loss of
1946 X. WU ET AL.

mitochondrial respiration. ACS Chem Neurosci. 2019;10 [56] Caldeira MV, Curcio M, Leal G, et al. Excitotoxic stimula­
(5):2489–2497 tion downregulates the ubiquitin-proteasome system
[45] Solenski NJ, diPierro CG, Trimmer PA, et al. Ultrastructural through activation of NMDA receptors in cultured hippo­
changes of neuronal mitochondria after transient and permanent campal neurons. Biochim. Biophys Acta-Mol Basis Dis.
cerebral ischemia. Stroke. 2002;33(3):816–24 2013;1832(1):263–274.
[46] Xiang G, Yang L, Long Q, et al. BNIP3L-dependent mitophagy [57] Herndon TM, Deisseroth A, Kaminskas E, et al. U.S. Food and
accounts for mitochondrial clearance during 3 factors-induced drug administration approval: carfilzomib for the treatment of
somatic cell reprogramming. Autophagy. 2017;13(9):1543–1555 multiple myeloma. Clin Cancer Res. 2013;19(17):4559–63
[47] O’Sullivan TE, Johnson L, Kang H, et al. BNIP3- and [58] Karademir B, Liu L, Feng D, et al. Proteomic approach for under­
BNIP3L-mediated mitophagy promotes the generation of natural standing milder neurotoxicity of carfilzomib against bortezomib.
killer cell memory. Immunity. 2015;43(2):331–42 Sci Rep. 2018;8(1):16318
[48] Chen G, Ray R, Dubik D, et al. The E1B 19K/Bcl-2-binding [59] Zhang L, Zhang ZG, Buller B, et al. Combination treatment with
protein Nip3 is a dimeric mitochondrial protein that activates VELCADE and low-dose tissue plasminogen activator provides
apoptosis. J Exp Med. 1997;186(12):1975–83 potent neuroprotection in aged rats after embolic focal ischemia.
[49] Hanna RA, Quinsay MN, Orogo AM, et al. Microtubule- Stroke. 2010;41(5):1001–7
associated protein 1 light chain 3 (LC3) interacts with [60] Kolev K, Skopál J, Simon L, et al. Matrix
Bnip3 protein to selectively remove endoplasmic reticulum metalloproteinase-9 expression in post-hypoxic human
and mitochondria via autophagy. J Biol Chem. 2012;287 brain capillary endothelial cells: H2O2 as a trigger and
(23):19094–104 NF-kappaB as a signal transducer. Thromb Haemost.
[50] Lee S, Zhang C, Liu X. Role of glucose metabolism and ATP in 2003;90(3):528–37
maintaining PINK1 levels during Parkin-mediated mitochondrial [61] Jeong EI, Chung HW, Lee WJ, et al. E2-25K SUMOylation inhi­
damage responses. J Biol Chem. 2015;290(2):904–17. bits proteasome for cell death during cerebral ischemia/reperfu­
[51] Allen GF, Toth R, James J, et al. Loss of iron triggers PINK1/ sion. Cell Death Dis. 2016;7(12):e2573
Parkin-independent mitophagy. EMBO Rep. 2013;14(12):1127–35. [62] Qiu JH, Asai A, Chi S, et al. Proteasome inhibitors induce cyto­
[52] Kanki T, Klionsky DJ. Mitophagy in yeast occurs through chrome c-caspase-3-like protease-mediated apoptosis in cultured
a selective mechanism. J Biol Chem. 2008;283(47):32386–93. cortical neurons. J Neurosci. 2000;20(1):259–65
[53] Marinkovic M, Sprung M, Novak I. Dimerization of mitophagy [63] Herrero-Mendez A, Almeida A, Fernandez E, et al. The bioener­
receptor BNIP3L/NIX is essential for recruitment of autophagic getic and antioxidant status of neurons is controlled by contin­
machinery. Autophagy. 2020;1–12. DOI:10.1080/ uous degradation of a key glycolytic enzyme by APC/C-Cdh1. Nat
15548627.2020.1755120 Cell Biol. 2009;11(6):747–52
[54] Frazier DP, Wilson A, Graham RM, et al. Acidosis regulates the [64] Baek SH, Noh AR, Kim K-A, et al. Modulation of mitochondrial
stability, hydrophobicity, and activity of the BH3-only protein function and autophagy mediates carnosine neuroprotection
Bnip3. Antioxid Redox Signal. 2006;8(9–10):1625–34 against ischemic brain damage. Stroke. 2014;45(8):2438–2443
[55] Bingol B, Tea JS, Phu L, et al. The mitochondrial deubiquitinase USP30 [65] Ran FA, Hsu PD, Wright J, et al. Genome engineering using the
opposes parkin-mediated mitophagy. Nature. 2014;510(7505):370–5 CRISPR-Cas9 system. Nat Protoc. 2013;8(11):2281–2308.

You might also like