You are on page 1of 28

TUGAS BLOK BIOLOGI MOLEKULAR

“ALDH7A1 Inhibits the Intracellular Transport Pathways During Hypoxia and


Starvation to Promote Cellular Energy Homeostasis”

Disusun Oleh

RATU NUR RAMADHANI

J500220037

FAKULTAS KEDOKTERAN

UNIVERSITAS MUHAMMADIYAH SURAKARTA

2023
ARTICLE
https://doi.org/10.1038/s41467-019-11932-0 OPEN

ALDH7A1 inhibits the intracellular transport


pathways during hypoxia and starvation to
promote cellular energy homeostasis
Jia-Shu Yang1, Jia-Wei Hsu1, Seung-Yeol Park1,2, Stella Y. Lee3, Jian Li1, Ming Bai1, Claudia Alves 1,

William Tseng1, Xavier Michelet1, I-Cheng Ho1 & Victor W. Hsu1


1234567890():,;

The aldehyde dehydrogenase (ALDH) family of metabolic enzymes converts aldehydes to


carboxylates. Here, we find that the reductive consequence of ALDH7A1 activity, which
generates NADH (nicotinamide adenine dinucleotide, reduced form) from NAD, underlies
how ALDH7A1 coordinates a broad inhibition of the intracellular transport pathways.
Studying vesicle formation by the Coat Protein I (COPI) complex, we elucidate that NADH
generated by ALDH7A1 targets Brefeldin-A ADP-Ribosylated Substrate (BARS) to inhibit
COPI vesicle fission. Moreover, defining a physiologic role for the broad transport inhibition
exerted by ALDH7A1, we find that it acts to reduce energy consumption during hypoxia and
starvation to promote cellular energy homeostasis. These findings advance the understanding
of intracellular transport by revealing how the coordination of multiple pathways can be
achieved, and also defining circumstances when such coordination is needed, as well as
uncovering an unexpected way that NADH acts in cellular energetics.

1 Division
of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, and Department of Medicine, Harvard Medical School, Boston, MA
02115, USA. 2 Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea. 3 Division of Biology,
Kansas State University, Manhattan, KS 66506, USA. Correspondence and requests for materials should be addressed to J.-S.Y. (email: jyang@rics.bwh.
harvard.edu) or to V.W.H. (email: vhsu@bwh.harvard.edu)

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 1


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

M
uch of our current understanding of vesicular transport COPI vesicle formation to be dissected out16,17,20. When
comes from studies on model pathways1–8. One such ALDH7A1 was added as another purified component (Supple-
pathway involves transport by the coat protein I (COPI) mentary Fig. 2c), we found that COPI vesicle formation is
complex9–11. Early studies identified a multimeric complex, inhibited (Fig. 1f). We also found that the catalytic mutation
known as coatomer, as the core components of the COPI com- prevents the inhibition of COPI vesicle formation in the
plex, and ADP-ribosylation factor 1 (ARF1) as the small GTPase reconstitution system (Fig. 1f). Examination by electron micro-
that regulates the recruitment of coatomer from the cytosol to scopy (EM) revealed that ALDH7A1 induces the accumulation of
Golgi membrane to initiate COPI vesicle formation12,13. Subse- buds with constricted necks on Golgi membrane (Fig. 1g). Thus,
quently, additional key factors have been identified to act in this these results suggested that ALDH7A1 acts as a negative regulator
process. These include the GTPase-activating protein (GAP) for of COPI transport by inhibiting the fission stage of vesicle
ARF1, known as ARFGAP1, which acts as another component of formation.
the COPI complex14,15, and Brefeldin-A ADP-ribosylated sub- We next considered that ALDH7A1 activity has been
strate (BARS), which acts in COPI vesicle fission16,17, the stage of characterized to convert multiple aldehydes to their correspond-
vesicle formation when coated buds undergo neck constriction to ing carboxylates22. Examining these metabolic products, we
become released from the membrane as transport vesicles. found that none could inhibit COPI vesicle formation (Fig. 1h).
To gain further insight into how COPI vesicle formation is We then noted that nicotinamide adenine dinucleotide in reduced
achieved, we have been seeking to identify additional factors that form (NADH) has been shown previously to inhibit the function
act in this process. In this study, we show that a member of the of BARS in COPI vesicle fission16. Thus, we explored whether
aldehyde dehydrogenase (ALDH) family of metabolic enzymes, NADH generated by ALDH7A1 activity could target BARS in
ALDH7A1, also known as antiquitin18, inhibits COPI vesicle explaining how ALDH7A1 inhibits COPI vesicle fission.
fission by targeting BARS. As BARS acts in the fission stage of We first confirmed that simply adding NADH to the
multiple transport pathways, we then uncover a broader set of reconstitution system inhibits COPI vesicle formation (Fig. 1h).
intracellular pathways inhibited by ALDH7A1. We also elucidate We also sought confirmation that NADH can directly target
a physiologic role for this global inhibition, showing that it pro- BARS. We had previously incubated recombinant BARS with
motes cellular energy homeostasis by reducing energy con- liposomes generated from defined pure lipids to demonstrate that
sumption during hypoxia and starvation, situations when cellular BARS can directly induce membrane deformation17. Re-visiting
energy production is impaired. this assay, we found that the addition of NADH prevents BARS
from inducing liposome tubulation (Supplementary Fig. 2d).
To further confirm that ALDH7A1 inhibits BARS through the
Results generation of NADH, we next examined the effect of a point
ALDH7A1 inhibits COPI vesicle fission by targeting BARS. mutation in BARS, glycine at position 172 to glutamate (G172E),
When BARS is fused to glutathione-S-transferase (GST), and then which has been shown previously to render BARS insensitive to
incubated with cytosol in a pulldown experiment, we identified NADH inhibition16. When wild-type BARS was replaced with
multiple interacting proteins (Supplementary Table 1). Besides this G172E mutant, we found that ALDH7A1 could no longer
transport factors, we also identified metabolic enzymes. In this inhibit COPI vesicle formation (Fig. 1i). Moreover, the G172E
regard, we have recently uncovered an unconventional role for mutation prevents the ability of NADH to inhibit liposome
glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in reg- tubulation by BARS (Supplementary Fig. 2d). We also sought
ulating COPI vesicle fission by targeting ARFGAP119. Thus, we physiologic confirmation by pursuing cell-based studies. We
explored whether any of the metabolic enzymes implicated to replaced endogenous wild-type BARS with the mutant (G172E)
interact with BARS could also have a role in COPI transport. BARS, which was achieved by treating cells with siRNA against
A quantitative assay that tracks COPI transport in cells BARS and then transfecting the mutant BARS, and found that
has been established, which examines the redistribution of a ALDH7A1 overexpression could no longer inhibit COPI trans-
COPI cargo protein (known as VSVG-KDELR) from the Golgi port (Fig. 1j). Thus, multiple lines of evidence all pointed to
complex to the endoplasmic reticulum (ER)16,17,20. Performing ALDH7A1 activity inhibiting COPI vesicle fission through the
this assay, we found that siRNA against ALDH7A1 (Supplemen- generation of NADH that targets BARS.
tary Fig. 1a), but not against catalase (Supplementary Fig. 1b)
or formimidoyltransferase-cyclodeminase (Ftcd) (Supplementary
Fig. 1c), enhances COPI transport (Supplementary Fig. 1d). We ALDH7A1 also inhibits multiple other intracellular pathways.
also confirmed that ALDH7A1 can interact directly with BARS, We next considered that BARS also acts at the fission stage in
as assessed by a pulldown experiment using purified components other transport pathways23. Thus, we explored whether
(Fig. 1a). Moreover, ALDH7A1 interacts with BARS in cells, as ALDH7A1 regulates additional pathways besides COPI transport.
assessed by a co-precipitation study (Fig. 1b). We had previously pursued a quantitative screen of the major
We then examined whether the catalytic activity is needed for intracellular pathways, which tracks the transit of model cargoes
ALDH7A1 to affect COPI transport. We targeted a residue in the in transport pathways through their colocalization with organelle
catalytic domain that is conserved across ALDH members21, markers in time-course studies24. Pursuing this approach, we
mutating glutamate at position 268 to glutamine (E268Q), and found that siRNA against ALDH7A1 does not affect transport
found that the point mutant could not rescue the effect of siRNA from the ER to the Golgi (Fig. 2a and Supplementary Fig. 3a). In
against ALDH7A1 (Fig. 1c) on COPI transport (Fig. 1d and contrast, siRNA against ALDH7A1 enhanced transport from the
Supplementary Fig. 2a). Moreover, we found that ALDH7A1 Golgi to the plasma membrane (PM) (Fig. 2b and Supplementary
overexpression had the opposite effect of inhibiting COPI Fig. 3b).
transport, which was also prevented by the point mutation All three major endocytic routes were also enhanced by siRNA
(Fig. 1e and Supplementary Fig. 2b). Thus, these results suggested against ALDH7A1, as reflected by the enhanced recycling of
that ALDH7A1 acts as a negative regulator of COPI transport. transferrin receptor (TfR) to the PM (Fig. 2c and Supplementary
To gain further insight into how ALDH7A1 inhibits COPI Fig. 3c), the enhanced retrograde transport of internalized cholera
transport, we next pursued the reconstitution of COPI vesicles toxin (CT) to the Golgi (Fig. 2d and Supplementary Fig. 3d), and
from Golgi membrane, as this approach enables the details of the enhanced delivery of internalized dextran to the lysosome

2 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0 ARTICLE

Control
a b c d 100 *

Colocalization of VSVG-KDELr
si ALDH7A1
si-ALDH7A1 Rescue w/ ALDH7A1 (wt)

ST RS
Transfection 80 *
Rescue Rescue w/ ALDH7A1 (E268Q)

Scramble

with giantin (%)


-B

yc
pu
ST

E268Q
*

m
In
60
G

S-
e

WT
R
49

on

BA
Blot: ALDH7A1

N
64 40
49 IP: Myc WB: ALDH7A1 49
26 Blot: GST ALDH7A1 20
49 Lysate
β-COP 82
0
0 20 40 60
Time (min)
e Control
f NS g
100 * 125
* 300
Colocalization of VSVG-KDELr

Overexpress ALDH7A1 wt *
Overexpress ALDH7A1 (E268Q)
80 100

Vesicle formation
*
with giantin (%)

(% control)

Buds per mesh


75 200
60

50
40
100
25
20 Bar: 50 nm
0
0 0
Ctl

WT

E268Q
0 20 40 60 Ctl ALDH7A1
Time (min)
ALDH7A1

h 125 NS i j Control
NS
125 Overexpress ALDH7A1(wt)
100 * 100 Overexpress ALDH7A1 (wt) and BARS (G172E) *
Colocalization of VSVG-KDELr
Vesicle formation

100
(% control)

Vesicle formation

75 80
(% control)

with giantin (%)


75 *
50 60
50
25 40
25
0
20
None

Hexanoic acid

Octanoic acid

2-aminoadipic acid

Betaine

NADH

0
BARS WT WT G172E 0
ALDH7A1 – + + 0 20 40 60
Time (min)

Fig. 1 ALDH7A1 inhibits COPI transport. Quantitative results are shown as mean with standard deviation; *p < 0.05, NS not significant, paired two-tailed
Student’s t-test. Source data are provided as a Source Data file. a GST–BARS on beads was incubated with recombinant ALDH7A1, followed by
immunoblotting with antibodies against indicated proteins; n = 2 independent experiments with a representative result shown. b HeLa cells were
transfected with construct as indicated, followed by immunoprecipitation for the myc tag and then immunoblotting for ALDH7A1; n = 2 independent
experiments with a representative result shown. c HeLa cells were treated as indicated and then whole-cell lysates were immunoblotted for proteins as
indicated; n = 2 independent experiments with a representative result shown. Rescues for siRNA-treated cells were performed using tagged forms of
ALDH7A1. d HeLa cells were treated as indicated, and then colocalization of VSVG-KDELR with a cis-Golgi marker (giantin) coupled with kinetic analysis
was performed; n = 3 independent experiments. e HeLa cells were treated as indicated, and then colocalization of VSVG-KDELR with a cis-Golgi marker
(giantin) coupled with kinetic analysis was performed; n = 3 independent experiments. f The COPI reconstitution system was performed followed by
quantitation; n = 3 independent experiments. g Golgi membrane after the incubation in the reconstitution system was examined by EM. Representative
images are shown on the left (scale bar, 50 nm), and quantitation is shown on the right; n = 3 independent experiments. h The COPI reconstitution system
was performed in the presence of different metabolic products (carboxylates) of ALDH7A1 or NADH; NS not significant, ANOVA; n = 3 independent
experiments. i The COPI reconstitution system was performed followed by quantitation; n = 3 independent experiments. j HeLa cells were treated as
indicated, and then the colocalization of VSVG-KDELR with a cis-Golgi marker (giantin) coupled with kinetic analysis was performed; n = 3 independent
experiments

(Fig. 2e and Supplementary Fig. 3e). Fluid-phase endocytosis was siRNA against ALDH7A1. The epidermal growth factor (EGF)
also enhanced, as reflected by increased uptake of dextran (Fig. 2f receptor (EGFR) is predicted to be one such cargo, as it is
and Supplementary Fig. 3f). However, clathrin-mediated endo- internalized through clathrin-mediated endocytosis, and then
cytosis was unaffected, as tracked by the internalization of TfR to transported through the endosome to reach the lysosome6. We
the early endosome (Fig. 2g). Altogether, the results revealed that first confirmed that the endocytosis of EGFR is unaffected by
six out of the eight major intracellular pathways are targeted by siRNA against ALDH7A1 (Fig. 2i). Subsequently, tracking the fate
ALDH7A1 for inhibition (summarized in Fig. 2h). of internalized EGFR to lysosomes, we found that this transport is
We also considered that the enhanced delivery of dextran to also enhanced by siRNA against ALDH7A1 (Fig. 2j). Thus, these
the lysosome induced by siRNA against ALDH7A1 (see Fig. 2e) results further confirmed that ALDH7A1 inhibits endocytic
could be an indirect effect of fluid-phase endocytosis having been transport to the lysosome.
enhanced by the siRNA treatment (see Fig. 2f). Thus, we We next found that the overexpression of ALDH7A1 has the
examined another endocytic cargo that is transported to the opposite effect of the siRNA treatment in affecting the identical
lysosome, but whose internalization at the PM is not affected by pathways (Fig. 3a–i). These effects also require the catalytic

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 3


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

a b c d
VSVG colocalization with 100 Control 100 Control
30
NS * 100 Control Control

CT colocalization with
VSVG colocalization
si ALDH7A1 si ALDH7A1 * *
si ALDH7A1 si ALDH7A1

with TGN 46 (%)


80 80 25

Tf colocalization
with Rab11 (%)
80

TGN 46 (%)
*
giantin (%)

60 20
60 60 *
NS
15 *
40 40 40 10
20 20 20 5
0 0 0 0
0 1 2 3 4 5 0 20 40 60 0 5 10 15 20 25 5 15 25 35
Time (min) Time (min) Time (min) Time (min)

e 100 Control f Control g h


* 100 si ALDH7A1 * 50 Control
Dextran colocalization

Dextran colocalization
si ALDH7A1 NS

Tf colocalization with
si ALDH7A1
* 40
with Lamp1 (%)

with EEA1 (%)


75 75

EEA1 (%)
30
50 * 50 PM
20
NS
25 25 10
Endosome
0 0 0
10 20 30 40 50 60 70 0 10 20 30 0 1 2 3 4 5 6
Time (min) Time (min) Time (min)
Lysosome

i j EGF colocalization with


60
Control
* Golgi
50 Control si ALDH7A1
EGF colocalization with

NS
si ALDH7A1 50
40 *
lamp1 (%)

40
EEA1 (%)

30
30
20 20
NS
10 10
ER
0 0
0 1 2 3 4 5 0 10 20 30 40 50
Time (min) Time (min)

Fig. 2 Knocking down ALDH7A1 enhances intracellular pathways. Quantitative results are shown as mean with standard deviation; *p < 0.05, NS not
significant, paired two-tailed Student’s t-test; n = 3 independent experiments. Source data are provided as a Source Data file. HeLa cells were assessed for the
following transport pathways: a Transport of VSVG from the ER to the Golgi was assessed through the quantitative colocalization of VSVG with a cis-Golgi
marker (giantin). b Transport of VSVG from the Golgi to the plasma membrane was assessed through the quantitative colocalization of VSVG with a TGN
marker (TGN46). c Endocytic recycling of Tf from the recycling endosome (RE) to the plasma membrane was assessed through the quantitative colocalization
of internalized Tf with a recycling endosome marker (Rab11). d Endocytic transport of cholera toxin (CT) to the Golgi was assessed through the quantitative
colocalization of internalized CT with a TGN marker (TGN46). e Endocytic transport of internalized dextran to the lysosome was assessed through the
quantitative colocalization of internalized dextran with a lysosome marker (Lamp1). f Fluid-phase endocytosis of dextran was assessed through the
quantitative colocalization of internalized dextran with an early endosome marker (EEA1). g Clathrin-mediated endocytosis of Tf was assessed through
the quantitative colocalization of internalized transferrin (Tf) with an early endosome marker (EEA1). h Summary of the major intracellular pathways regulated
by ALDH7A1. Red arrows indicate pathways inhibited by ALDH7A1. Green arrows indicate pathways unaffected by ALDH7A1. i Endocytosis of EGFR was
assessed through the quantitative colocalization of internalized EGF with an early endosome marker (EEA1). j Endocytic transport of internalized EGFR to the
lysosome was assessed through the quantitative colocalization of internalized EGF with a lysosome marker (Lamp1)

activity of ALDH7A1, as the catalytic mutation (E268Q) (Supplementary Fig. 5b), and fluid-phase endocytosis (Supple-
prevented the ability of ALDH7A1 overexpression from exerting mentary Fig. 5c). In contrast, this siRNA treatment did not affect
transport inhibition (Fig. 3a–i). Moreover, ALDH7A1 over- endocytic recycling (Supplementary Fig. 5d), endocytic transport
expression could no longer inhibit the BARS-dependent path- to the lysosome (Supplementary Fig. 5e), and endocytic transport
ways, when endogenous wild-type BARS was replaced with the to the Golgi (Supplementary Fig. 5f). Thus, ALDH7A1 is
mutant (G172E) BARS that is defective in binding to NADH predicted to target additional factor(s) besides BARS in
(Supplementary Fig. 3g and h). Consistent with these findings, we explaining how it exerts a broad inhibition of the transport
also found that enhanced COPI transport induced by siRNA pathways.
against ALDH7A1 is prevented when cells were treated with Next, to rule out that any metabolic enzyme that generates
siRNA against BARS (Supplementary Fig. 3i). NADH could exert transport inhibition, we examined the effect of
We also confirmed that these effects of ALDH7A1 are not reducing the cellular level of two dehydrogenases, aldolase
restricted to HeLa cells. Examining human embryonic kidney 293 (Supplementary Fig. 5g) and malate dehydrogenase 2 (MDH2)
(HEK293) cells, we observed similar effects, as reducing (Supplementary Fig. 5h). However, transport was not affected by
ALDH7A1 level through siRNA treatment enhances transport siRNA targeting against either enzyme (Supplementary Fig. 5i).
in the identical pathways and increasing ALDH7A1 level through We also examined another member of the ALDH family, and
overexpression inhibits transport in the identical pathways found that siRNA against ALDH1A1 (Supplementary Fig. 6a)
(Supplementary Fig. 4a–i). does not affect the transport pathways (Supplementary Fig.
We next examined whether the affected transport pathways 6b–h). Moreover, consistent with how ALDH7A1 affects the
could all be explained by ALDH7A1 targeting BARS. Upon BARS-dependent pathways, we found that ALDH1A1, which
siRNA against BARS, we observed transport inhibition in Golgi- does not affect the BARS-dependent pathways, also does not
to-ER transport (Supplementary Fig. 5a), Golgi-to-PM transport interact with BARS (Supplementary Fig. 6i).

4 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0 ARTICLE

Control
a Control b 100 Control c ALDH7A1 (wt) OE
100
ALDH7A1 (wt) OE
*
ALDH7A1 (wt) OE 100 ALDH7A1 (E268Q) OE
NS *

VSVG colocalization
ALDH7A1 (E268Q) OE ALDH7A1 (E268Q) OE

VSVG colocalization
80 75

with TGN 46 (%)


*
with giantin (%)

Tf colocalization
with Rab11 (%)
75
60
NS 50 *
50
40
25 25
20

0 0 0
0 1 2 3 4 5 0 20 40 60 0 5 10 15 20 25
Time (min) Time (min) Time (min)

d 30 e 100 f 100
Control Control Control

Dextran colocalization with

Dextran colocalization with


ALDH7A1 (wt) OE ALDH7A1 (wt) OE
CT colocalization with

ALDH7A1 (wt) OE
* 75 ALDH7A1 (E268Q) OE * 75 *
ALDH7A1 (E268Q) OE ALDH7A1 (E268Q) OE
20
TGN 46 (%)

lamp1 (%)

EEA1 (%)
50 50
*
*
10
* 25 25

0 0 0
5 15 25 35 10 20 30 40 50 60 70 0 10 20 30
Time (min) Time (min) Time (min)

g h i 60 Control
Control 60 Control ALDH7A1 (wt) OE
60 *
ALDH7A1 (wt) OE ALDH7A1 (E268Q) OE

EGF colocalization with


ALDH7A1 (wt) OE NS
NS
EGF colocalization with

ALDH7A1 (E268Q) OE
Tf colocalization with

ALDH7A1 (E268Q) OE
40 *

lamp1 (%)
40 NS 40
EEA1 (%)
EEA1 (%)

NS

20 20 20

0 0 0
0 1 2 3 4 5 6 0 1 2 3 4 5 6 0 10 20 30 40 50
Time (min) Time (min) Time (min)

Fig. 3 ALDH7A1 overexpression inhibits intracellular pathways. Quantitative results are shown as mean with standard deviation; *p < 0.05, NS not
significant, paired two-tailed Student’s t-test; n = 3 independent experiments. Source data are provided as a Source Data file. HeLa cells were assessed for
the following transport pathways: a Transport of VSVG from the ER to the Golgi was assessed through the quantitative colocalization of VSVG with a cis-
Golgi marker (giantin). b Transport of VSVG from the Golgi to the plasma membrane was assessed through the quantitative colocalization of VSVG with a
TGN marker (TGN46). c Endocytic recycling of Tf from the recycling endosome (RE) to the plasma membrane was assessed through the quantitative
colocalization of internalized Tf with a recycling endosome marker (Rab11). d Endocytic transport of CT to the Golgi was assessed through the quantitative
colocalization of internalized CT with a TGN marker (TGN46). e Endocytic transport of internalized dextran to the lysosome was assessed through
the quantitative colocalization of internalized dextran with a lysosome marker (Lamp1). f Fluid-phase endocytosis of dextran was assessed through the
quantitative colocalization of internalized dextran with an early endosome marker (EEA1). g Clathrin-mediated endocytosis of Tf was assessed through
the quantitative colocalization of internalized transferrin (Tf) with an early endosome marker (EEA1). h Endocytosis of EGFR was assessed through the
quantitative colocalization of internalized EGF with an early endosome marker (EEA1). i Endocytic transport of internalized EGFR to the lysosome was
assessed through the quantitative colocalization of internalized EGF with a lysosome marker (Lamp1)

To further characterize transport inhibition by ALDH7A1, we and subcellular fractionation (Fig. 4e) that appending NES to
next fractionated the cell into total membrane and cytosol, and BARS markedly reduces its nuclear localization. Next, we replaced
found that ALDH7A1 interacts with BARS on the membrane but endogenous wild-type BARS with NES-BARS, which was achieved
not in the cytosol (Fig. 4a). Moreover, we found by confocal by knocking down BARS followed by rescue using a siRNA-
microscopy that ALDH7A1 is broadly distributed among the resistant form of NES-BARS. We then confirmed functionally that
intracellular membrane compartments (Fig. 4b). Notably, NES-BARS has markedly reduced the ability to inhibit transcrip-
ALDH7A1 level is markedly reduced at the ER (Fig. 4c), which tion, as assessed through the mRNA level of E-cadherin (Fig. 4f),
is consistent with our observation above that ALDH7A1 does not which has been a model transcript for studies on transcriptional
affect transport from the ER. repression by CtBP25. Notably, we then found that NES-BARS
We also considered that BARS acts not only as a fission factor in mediates the inhibition of transport pathways by ALDH7A1, as
transport pathways, but also as a transcription repressor. In this reflected by effects on COPI transport (Fig. 4g) and fluid-phase
latter role, it is known as C-terminal binding protein (CtBP)25. uptake (Fig. 4h). Thus, the collective results suggested that
Thus, we examined whether the ALDH7A1 could affect the transport inhibition by ALDH7A1 is unlikely to involve the
transport pathways indirectly through the transcription function of targeting of the transcription function of BARS/CtBP.
BARS. To act in transcription, BARS needs to localize to the
nucleus. Thus, to prevent the nuclear localization of BARS, we
ALDH7A1 promotes cellular energy homeostasis. We then
appended a nuclear export signal (NES) to BARS (NES-BARS). We
sought to elucidate the physiologic significance of the broad
initially confirmed by immunofluorescence microscopy (Fig. 4d)
transport inhibition exerted by ALDH7A1. In plants, ALDH7A1

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 5


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

a b ALDH7A1/calnexin ALDH7A1/CM ALDH7A1/TGN46

e
an

ol
br

os
em

yt
M

C
BARS-Myc

BARS-Myc
None

None
ALDH7A1/EEA1 ALDH7A1/Rab11 ALDH7A1/Lamp1

49 ALDH7A1
64 IP: Myc
BARS
49

c
with membrane marker (%)

30
Percentage of ALDH7A1

d e

S
AR
colocalization

BARS NES-BARS

-B
20

S
ES

R
BA
N
10 C N C N

49 GFP-BARS

0 64 Lamin B
ER Golgi TGN EE RE LE 49
Actin

f 6 g Control h 100
ALDH7A1 + BARS NS Control
100

Dextran colocalization with


VSVG-KDELr colocalization with

5 ALDH7A1 + NES-BARS
E-cad mRNA levels

* ALDH7A1 + BARS
75 NS
(fold change)

4 75 NS ALDH7A1 + NES-BARS

EEA1 (%)
3
giantin (%)

50
50
2
NS
1 25 25
0
Control – BARS NES-BARS 0 0
siBARS 0 20 40 60 0 5 10 15 20 25 30 35
Time (min) Time (min)

Fig. 4 ALDH7A1 targets the transport function of BARS. Source data are provided as a Source Data file. a HeLa cells were subjected to subcellular
fractionation to obtain total membrane vs. cytosol fractions. Both fractions were then subjected to co-immunoprecipitation experiments to detect the
association of ALDH7A1 with BARS; n = 2 independent experiments with a representative result shown. b Confocal microscopy was performed to detect
the colocalization of ALDH7A1 with different organelle markers; scale bar, 10 µm (2 µm in inset); n = 3 independent experiments with representative
images shown. c Quantitation of the colocalization results above; n = 3 independent experiments with ten fields of cells examined in each experiment.
d HeLa cells were transfected with GFP-tagged NES-BARS followed by confocal imaging across the nuclear portion of the cell; scale bar, 10 µm; n = 2
independent experiments with a representative image shown. e HeLa cells were fractionated into nuclear vs. cytoplasmic fractions, followed by
immunoblotting for proteins as indicated. Lamin B serves as a marker of the nucleus, while actin serves as a marker of the cytoplasm; n = 2 independent
experiments with a representative result shown. f HeLa cells were treated as indicated, and then the mRNA level of E-cadherin, which has been a model
transcript for studies on transcription repression by BARS/CtBP, is quantified; n = 3 independent experiments. g HeLa cells were treated as indicated, and
then colocalization of VSVG-KDELR with a cis-Golgi marker (giantin) coupled with kinetic analysis was performed. Mean with standard deviation is shown;
*p < 0.05, paired two-tailed Student’s t-test; n = 3 independent experiments. h HeLa cells were treated as indicated, and then colocalization of dextran with
an early endosome marker (EEA1) coupled with kinetic analysis was performed. Mean with standard deviation is shown; *p < 0.05, paired two-tailed
Student’s t-test; n = 3 independent experiments

has been suggested to protect cells from osmotic stress18. How- We next examined whether ALDH7A1 is critical for cellular
ever, mammalian cells are not typically exposed to this form of energy homeostasis during these two major states of energy
stress. Instead, because the intracellular pathways require the deprivation. We initially found that viability is not appreciably
participation of GTPases and ATPases, which consume sub- compromised when cells are subjected to siRNA against
stantial energy, we explored whether ALDH7A1 could have a ALDH7A1 in the normal (non-hypoxic) condition (Fig. 7a).
protective role during energy stress. Two major pathologic However, when the siRNA-treated cells are subjected to hypoxia,
examples of energy stress are hypoxia and starvation. Initially, we we observed enhanced cell death (Fig. 7a). Consistent with these
found that hypoxia inhibits the identical set of transport pathways findings, whereas the total ATP level is maintained when cells
as those targeted by ALDH7A1 (Fig. 5a–h). Moreover, siRNA were treated with siRNA against ALDH7A1 in the normal (non-
against ALDH7A1 prevents these inhibitions (Fig. 5a–h). Exam- hypoxic) condition, we found that ATP level drops dramatically
ining starvation, we found that this condition also inhibits the when cells are subjected to both hypoxia and siRNA against
identical set of pathways as those targeted by ALDH7A1, with ALDH7A1 (Fig. 7b).
siRNA against ALDH7A1 similarly preventing these inhibitions To examine starvation, we switched cells into a medium that
(Fig. 6a–h). lacks glucose and amino acids for nutrient deprivation, but

6 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0 ARTICLE

a 100 Control b c 100


Control

VSVG-KDELr colocalization
Hypoxia 100 Control NS
* Hypoxia
Hypoxia + si ALDH7A1

VSVG colocalization
Hypoxia

VSVG colocalization
75 * 75

with giantin (%)

with giantin (%)


Hypoxia + si ALDH7A1

with TGN 46 (%)


75
*
50 50
50 NS
*
25 25 25

0 0 0
0 10 20 30 40 50 60 0 10 20 30 40 50 60 0 1 2 3 4 5
Time (min) Time (min) Time (min)

d Control e 20
Control
100 Hypoxia Hypoxia
Hypoxia + si ALDH7A1 * Hypoxia + si ALDH7A1 *

CT colocalization with
Tf colocalization with

15 *
75

TGN 46 (%)
Rab 11 (%)

*
50 10

25 5

0 0
0 5 10 15 20 25 0 10 20 30
Time (min) Time (min)

f 50
Control g 80
Control h 30
Hypoxia Hypoxia
* Control
*
Dextran colocalization with

Hypoxia + si ALDH7A1 Hypoxia + si ALDH7A1 NS


EGF colocalization with

* 25 Hypoxia

Tf colocalization with
40
60
20
Lamp1 (%)

EEA1 (%)
EEA1 (%)

30 *
40 15
20 NS
10
20
10 5

0 0 0
0 10 20 30 40 50 60 0 10 20 30 0 1 2 3 4 5
Time (min) Time (min) Time (min)

Fig. 5 Hypoxia inhibits the identical pathways targeted by ALDH7A1. Quantitative results are shown as mean with standard deviation; *p < 0.05, NS not
significant, paired two-tailed Student’s t-test; n = 3 independent experiments. Source data are provided as a Source Data file. HeLa cells were assessed for
the following transport pathways: a COPI transport from the Golgi to the ER was assessed through the quantitative colocalization of VSVG-KDELR with a
cis-Golgi marker (giantin). b Transport of VSVG from the Golgi to the plasma membrane was assessed through the quantitative colocalization of VSVG
with a TGN marker (TGN46). c Transport of VSVG from the ER to the Golgi was assessed through the quantitative colocalization of VSVG with a cis-Golgi
marker (giantin). d Endocytic recycling of Tf from the recycling endosome (RE) to the plasma membrane was assessed through the quantitative
colocalization of internalized Tf with a recycling endosome marker (Rab11). e Endocytic transport of CT to the Golgi was assessed through the quantitative
colocalization of internalized CT with a TGN marker (TGN46). f Endocytic transport of internalized EGFR to the lysosome was assessed through
the quantitative colocalization of internalized EGF with a lysosome marker (Lamp1). g Fluid-phase endocytosis of dextran was assessed through the
quantitative colocalization of internalized dextran with an early endosome marker (EEA1). h Clathrin-mediated endocytosis of Tf was assessed through the
quantitative colocalization of internalized transferrin (Tf) with an early endosome marker (EEA1)

contains serum for cell survival. Upon this switch, we observed expression of the siRNA-resistant mutant BARS (Fig. 7e). We first
the total cellular ATP level to drop immediately (Fig. 7c). confirmed that the expression of the mutant BARS prevents
Notably, this drop was magnified when the starved cells were also hypoxia from exerting transport inhibition, as assessed by COPI
subjected to siRNA against ALDH7A1 (Fig. 7c). Consistent with transport (Fig. 7f) and fluid-phase endocytosis (Fig. 7g). We then
this result, cell death also increased when cells were subjected to found that the expression of the BARS mutant reduces total ATP
both starvation and siRNA against ALDH7A1 (Fig. 7d). level (Fig. 7h) and cell viability (Fig. 7i) during hypoxia. Similar
Besides HeLa cells, we also examined HEK293 cells. Upon results were also seen in starvation, as the reduced total ATP level
siRNA against ALDH7A1, we again observed total ATP level (Fig. 7j) and cell viability (Fig. 7k) were magnified by the
(Supplementary Fig. 7a) and cell viability to be reduced during expression of the mutant BARS.
hypoxia (Supplementary Fig. 7b). Moreover, siRNA against We next considered that AMP-activated protein kinase
ALDH7A1 also further enhanced the drop in the total ATP level (AMPK) acts as a master coordinator of cellular energy home-
(Supplementary Fig. 7c) and cell viability (Supplementary Fig. 7d) ostasis, which involves its ability to sense energy deficit and then
seen during starvation. target effectors to restore energy balance26. Thus, a potential role
Next, to link transport inhibition by ALDH7A1 to its ability to for AMPK in regulating ALDH7A1 would further support
promote energy homeostasis, we examined the effect of ALDH7A1 acting in energy homeostasis, as well as gaining
expressing the mutant BARS (G172E), which prevents the further insights into how ALDH7A1 could be regulated in this
BARS-dependent transport pathways from being inhibited by process. Initially, we found that the inhibition of transport by
ALDH7A1. To express a physiologic level of the mutant BARS, hypoxia also requires AMPK, as reflected by siRNA against
we treated cells with siRNA against BARS followed by limited AMPKα1 that prevented hypoxia from inhibiting both COPI

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 7


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

a b 100 Control c
100 Control

VSVG-KDELr colocalization
Starvation
Starvation * 80

VSVG colocalization
si ALDH7A1/starvation Control
* 75

with TGN 46 (%)

VSVG colocalization
si ALDH7A1/starvation NS

with giantin (%)


75 Starvation NS

with giantin (%)


60
*
50 *
50 40

25 25
20

0 0 0
0 20 40 60 0 20 40 60 0 1 2 3 4 5
Time (min) Time (min) Time (min)

d Control e Control
20 Starvation
100 Starvation
si ALDH7A1/starvation * si ALDH7A1/starvation *

CT colocalization with
Tf colocalization with

15
75

TGN 46 (%)
Rab 11 (%)

* *
50 10

25 5

0 0
0 5 10 15 20 25 0 10 20 30
Time (min) Time (min)

f Control g 100 Control


h
Starvation
si ALDH7A1/starvation * Starvation 30 Control
60 *
Dextran colocalization

si ALDH7A1/starvation NS
*
EGF colocalization with

Starvation

Tf colocalization with
75 25
50
with EEA1 (%)

* 20

EEA1 (%)
Lamp1 (%)

40
50
30 15 NS

20 10
25
10 5

0 0 0
0 10 20 30 40 50 0 10 20 30 0 1 2 3 4 5
Time (min) Time (min) Time (min)

Fig. 6 Starvation inhibits the identical pathways targeted by ALDH7A1. Quantitative results are shown as mean with standard deviation; *p < 0.05, NS not
significant, paired two-tailed Student’s t-test; n = 3 independent experiments. Source data are provided as a Source Data file. HeLa cells were assessed for
the following transport pathways: a COPI transport from the Golgi to the ER was assessed through the quantitative colocalization of VSVG-KDELR with a
cis-Golgi marker (giantin). b Transport of VSVG from the Golgi to the plasma membrane was assessed through the quantitative colocalization of VSVG
with a TGN marker (TGN46). c Transport of VSVG from the ER to the Golgi was assessed through the quantitative colocalization of VSVG with a cis-Golgi
marker (giantin). d Endocytic recycling of Tf from the recycling endosome (RE) to the plasma membrane was assessed through the quantitative
colocalization of internalized Tf with a recycling endosome marker (Rab11). e Endocytic transport of CT to the Golgi was assessed through the quantitative
colocalization of internalized CT with a TGN marker (TGN46). f Endocytic transport of internalized EGFR to the lysosome was assessed through
the quantitative colocalization of internalized EGF with a lysosome marker (Lamp1). g Fluid-phase endocytosis of dextran was assessed through the
quantitative colocalization of internalized dextran with an early endosome marker (EEA1). h Clathrin-mediated endocytosis of Tf was assessed through the
quantitative colocalization of internalized transferrin (Tf) with an early endosome marker (EEA1)

transport (Fig. 8a) and fluid-phase endocytosis (Fig. 8b). Similar fluid-phase endocytosis (Fig. 8j). Thus, the collective results
results were also obtained for starvation, as the inhibition of suggested that AMPK acts mechanistically upstream of ALDH7A1
COPI transport (Fig. 8c) and fluid-phase endocytosis (Fig. 8d) by and BARS in regulating the transport pathways.
starvation also require AMPK. Next, to examine how AMPK regulates ALDH7A1, we pursued
We next queried the relationship between AMPK and an algorithm that predicts consensus sites on substrates targeted
ALDH7A1. AMPK activity can be activated in cells by by different kinases30. This approach suggested two residues in
pharmacologic treatment using 5-aminoimidazole-4-carboxa- ALDH7A1, serine at position 102 (S102) and serine at position
mide-1-β-D-ribofuranoside (AICAR)27,28. We found that this 146 (S146), as potential sites of phosphorylation by AMPK. To
treatment is sufficient to inhibit transport in the normal condition, determine whether AMPK phosphorylates ALDH7A1 at these
as assessed by COPI transport (Fig. 8e) and fluid-phase residues, we next performed in vitro kinase assays by incubating
endocytosis (Fig. 8f). We also found that transport inhibition AMPK and ALDH7A1 as purified components. To detect
induced by AICAR is prevented by siRNA against ALDH7A1 phosphorylation, we used an antibody that recognizes consensus
(Fig. 8e, f). These effects are selective, as siRNA against ALDH7A1 sites of AMPK phosphorylation (referred hereon as the phospho-
does not affect another cellular process targeted by AMPK, the AMPK substrate antibody). We found that wild-type ALDH7A1 is
activation of sirtuin 1 activity29 (Fig. 8g). Moreover, siRNA against phosphorylated by AMPK (Fig. 9a). Moreover, this phosphoryla-
AMPKα1 does not affect clathrin-mediated endocytosis (Fig. 8h), tion is prevented when the S102 residue in ALDH7A1 is mutated
a BARS-independent pathway that is not targeted by ALDH7A1. to alanine (S102A), but not when the S146 residue is mutated to
We also found that the expression of the mutant BARS (G172E) alanine (S146A) (Fig. 9a). Thus, these results suggested that
prevents AICAR from inhibiting COPI transport (Fig. 8i) and AMPK only phosphorylates ALDH7A1 at the S102 residue.

8 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0 ARTICLE

a 100
b 125 c d 60 Control
* si ALDH7A1
*
* 125 Control
Cell death (% total)
100 * 50
75 si ALDH7A1

ATP (% control)
*

Cell death (%)


100 40

ATP (% control)
75 *
50 75 30
50
50 20
25
25 25
NS 10

0 0 0 0
si ALDH7A1 – + – + si ALDH7A1 – + – + 0 10 20 30 40 0 4 8 12 16 20 24
Hypoxia Hypoxia Time (min) Time (h)

e f Control g Control
Hypoxia *

VSVG-KDELr colocalization with


100 Hypoxia
Hypoxia + BARS (G172E) 100 *
BARS (G172E)

Hypoxia + BARS (G172E)

Dextran colocalization
75 * 75
si-BARS

with EEA1 (%)


Rescue

*
Control

giantin (%)
50 50

49 BARS
25 25

15 ARF1
0 0
0 20 40 60 0 10 20 30
Time (min) Time (min)

h 125 i 100 j k 60 Control


* 125 Control BARS (G172E) *
100
Cell death (% total)

BARS (G172E) *
ATP (% control)

75 50

Cell death (% total)


75
* ATP (% control) 100
*
* 40
50
75
50 30
25 50
25 20

0 0 25 10
– –
WT

G172E

WT

G172E

WT

G172E

WT

G172E

BARS – – BARS
0 0
0 10 20 30 0 4 8 12 16 20 24
Hypoxia Hypoxia Time (min) Time (h)

Fig. 7 ALDH7A1 promotes energy homeostasis during hypoxia and starvation. Quantitative results are shown as mean with standard deviation; *p < 0.05,
NS not significant, paired two-tailed Student’s t-test. Source data are provided as a Source Data file. a HeLa cells were treated as indicated, and then cell
death was quantified; n = 3 independent experiments. b HeLa cells were treated as indicated, and then total ATP level was quantified; n = 3 independent
experiments. c Starved HeLa cells were treated as indicated, and then total ATP level was quantified; n = 3 independent experiments. d Starved HeLa cells
were treated as indicated, and then cell death was quantified; n = 3 independent experiments. e Immunoblotting of HeLa cell lysates showing the efficacy of
siRNA against BARS, and the level of mutant BARS expressed for rescue studies; n = 2 independent experiments with a representative result shown. f COPI
transport from the Golgi to the ER in HeLa cells was assessed through the quantitative colocalization of VSVG-KDELR with a cis-Golgi marker (giantin); n =
3 independent experiments. g Fluid-phase endocytosis in HeLa cells was assessed through the quantitative colocalization of internalized dextran with an
early endosome marker (EEA1); n = 3 independent experiments. h HeLa cells were treated as indicated, and then total ATP level was quantified; n = 3
independent experiments. i HeLa cells were treated as indicated, and then cell death was quantified; n = 3 independent experiments. j HeLa cells were
treated with starvation conditions as indicated, and then total ATP level was quantified; n = 3 independent experiments. k HeLa cells were treated with
starvation conditions as indicated, and then cell death was quantified; n = 3 independent experiments

This conclusion was supported by another approach. We found phosphorylation of ALDH7A1 induced by hypoxia is abrogated
that radio-labeled phosphate is incorporated into wild-type, but by expressing the S102A mutation, but not the S146A mutation,
not the S102A mutant, of ALDH7A1 in the kinase assay of ALDH7A1 (Fig. 9c).
(Supplementary Fig. 8a). We also assessed the stoichiometry of We then examined how the phosphorylation status of
this phosphorylation by tracking the rate of ADP generation ALDH7A1 regulates the transport pathways by mutating the
during the kinase assay. When compared with a known optimal S102 residue to alanine (S102A), which prevents its phosphoryla-
substrate of AMPK, a peptide derived from acetyl CoA tion, or to aspartate (S102D), which mimics its constitutive
carboxylase (ACC)26, we found that AMPK phosphorylates phosphorylation. To avoid the overexpression of these ALDH7A1
ALDH7A1 with reasonable efficiency (Supplementary Fig. 8b mutants, we treated cells with siRNA against ALDH7A1 followed
and Supplementary Table 2). Thus, ALDH7A1 is likely a relevant by the limited expression of the transfected mutants (Fig. 9d). In
substrate of AMPK. cells that expressed the S102A mutant, we found that hypoxia can
We also sought to confirm this conclusion through cell-based no longer inhibit COPI transport (Fig. 9e) or fluid-phase
studies. First, we found that conditions that activate AMPK endocytosis (Fig. 9f). On the other hand, we found that the
(hypoxia or AICAR treatment) enhance AMPK-mediated expression of the S102D mutant in the normal (non-energy
phosphorylation of ALDH7A1 in cells (Fig. 9b). Second, this stress) condition is sufficient to inhibit COPI transport (Fig. 9g)
phosphorylation occurs at the S102 residue, as the enhanced and fluid-phase endocytosis (Fig. 9h).

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 9


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

a Control
b Control
c Control d
Starvation Control
Hypoxia Hypoxia
100 *

VSVG-KDELr colocalization
* * Starvation + si AMPK 100 Starvation
VSVG-KDELr colocalization

Dextran colocalization with


Hypoxia + si AMPK Hypoxia + si AMPK

Dextran colocalization with


100 80 *
Starvation + si AMPK

with giantin (%)


75 75
with giantin (%)

75 60

EEA1 (%)
* *

EEA1 (%)
*
* 50 50
50 40

25 20 25 25

0 0 0 0
0 20 40 60 0 10 20 30 0 20 40 60 0 10 20 30
Time (min) Time (min) Time (min) Time (min)

e Control f Control g NS
h
AICAR AICAR 40 50 Control
*
VSVG-KDELr colocalization

100 80 * NS
Dextran colocalization with
AICAR + siALDH7A1 AICAR + si ALDH7A1

Relative Sirt1 activity


si AMPK

EGF colocalization
40 NS
30

with EEA1 (%)


with giantin (%)

75 60 *
30
EEA1 (%)

*
20
50 40
20

25 20 10
10

0 0 0 0
0 20 40 60 0 10 20 30 Ctl AICAR Ctl AICAR 0 1 2 3 4 5
Time (min) Time (min) si ALDH7A1 Time (min)

i 100
Control j Control
AICAR
VSVG-KDELr colocalization

* 80 AICAR
AICAR + BARS (G172E) Dextran colocalization with AICAR + BARS (G172E) *
75
with giantin (%)

60
EEA1 (%)

* *
50
40

25 20

0 0
0 20 40 60 0 10 20 30
Time (min) Time (min)

Fig. 8 AMPK inhibits the transport pathways through ALDH7A1. Quantitative results are shown as mean with standard deviation; *p < 0.05, NS not
significant, paired two-tailed Student’s t-test; n = 3 independent experiments. Source data are provided as a Source Data file. a COPI transport from the
Golgi to the ER in HeLa cells was assessed through the quantitative colocalization of VSVG-KDELR with a cis-Golgi marker (giantin). b Fluid-phase
endocytosis of dextran in HeLa cells was assessed through the quantitative colocalization of internalized dextran with an early endosome marker (EEA1).
c COPI transport from the Golgi to the ER in HeLa cells was assessed through the quantitative colocalization of VSVG-KDELR with a cis-Golgi marker
(giantin). d Fluid-phase endocytosis of dextran in HeLa cells was assessed through the quantitative colocalization of internalized dextran with an early
endosome marker (EEA1). e COPI transport from the Golgi to the ER in HeLa cells was assessed through the quantitative colocalization of VSVG-KDELR
with a cis-Golgi marker (giantin). f Fluid-phase endocytosis of dextran in HeLa cells was assessed through the quantitative colocalization of internalized
dextran with an early endosome marker (EEA1). g HeLa cells were treated as indicated, and then Sirt1 activity was quantified. h EGF endocytosis in HeLa
cells was assessed through the quantitative colocalization of internalized EGF with an early endosome marker (EEA1). i COPI transport from the Golgi to
the ER in HeLa cells was assessed through the quantitative colocalization of VSVG-KDELR with a cis-Golgi marker (giantin). j Fluid-phase endocytosis of
dextran in HeLa cells was assessed through the quantitative colocalization of internalized dextran with an early endosome marker (EEA1)

The effects of the phosphorylation mutants suggested yet when taken altogether, the effects of the ALDH7A1 phosphoryla-
another way of linking transport inhibition by ALDH7A1 to its tion mutants provided substantial further support that ALDH7A1
ability to promote energy homeostasis. We found that the promotes in cellular energy homeostasis during situations of
expression of the S102A mutant results in markedly decreased energy deprivation by reducing energy consumption through the
cellular ATP level during hypoxia, but not in the normal inhibition of transport pathways.
condition (Fig. 9i). Cell viability is also only reduced, when the
S102A mutant is expressed in conjunction with hypoxia (Fig. 9j).
In contrast, the expression of the S102D mutant promotes the Phosphorylation by AMPK recruits ALDH7A1 to membranes.
ability of the cell to maintain total ATP level (Fig. 9i) and viability We next examined how the phosphorylation of ALDH7A1 at its
(Fig. 9j) during hypoxia. Similar results were obtained when we S102 residue regulates its ability to exert transport inhibition. We
examined starvation. The ability of the cell to maintain its ATP initially found that the catalytic activity is unaffected by mutating
level during starvation is worsened by the expression of the the S102 residue (Fig. 10a). We then considered that the
S102A mutant, while the S102D mutant shows a protective effect recruitment from the cytosol to the membrane provides a major
(Fig. 9k). Moreover, expression of the S102A mutant worsens cell way of regulating transport factors. Thus, we examined whether
viability during starvation, while expression of the S102D is phosphorylation of the S102 residue regulates the recruitment of
protective (Fig. 9l). We also expressed the G172E mutant of BARS cytosolic ALDH7A1 to membrane. Fractionating cells into total
in cells, and found that the S102D mutant of ALDH7A1 can no membrane and cytosol, we found that, compared to the wild-type
longer protect the total ATP level (Supplementary Fig. 8c) and form, the S102A mutant has reduced distribution on the mem-
cell viability (Supplementary Fig. 8d) during starvation. Thus, brane, while the S102D mutant has enhanced distribution on the

10 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0 ARTICLE

a b c d

Si ALDH7A1
Rescue
ALDH7A1-myc

S146A

S102A

S146A

S102A

Control
ia

S102D

S102A
AR
ox
e
WT

on

yp

WT
C
AI
N

H
P-S/T 49 P-S/T P-S/T ALDH7A1
49 49 49
49 ALDH7A1
ALDH7A1 ALDH7A1
49
49 β-COP
IP: ALDH7A1 82
IP: myc

e Control
f Control g h WT
WT/hypoxia *
* WT 80

Dextran colocalization with


WT/hypoxia S102A/hypoxia S102D
VSVG-KDELr colocalization

80
Dextran colocalization with
100

VSVG-KDELr colocalization
* 100 *
S102A/hypoxia S102D *
60
with giantin (%)

75 60

with giantin (%)


75

EEA1 (%)
*
EEA1 (%)
* *
50 40 40
50

25 20 25 20

0 0 0 0
0 20 40 60 0 10 20 30 0 20 40 60 0 10 20 30
Time (min) Time (min) Time (min) Time (min)

i j k No treatment l
125 ALDH7A1 (S102A)
* 125 Control
ALDH7A1 (S102D)
*
100 60 S102D
100 S102A * *
* 100 * 50
ATP (% control)

Cell death (% total)

Total cell death (%)


ATP (% control)
75
75 75 40
50 30
50 50
20
25 25 25
10
0 0 0 0
si ALDH7A1 – + + + – + + + si ALDH7A1 – + + + + 0 10 20 30 0 4 8 12 16 20 24
Rescue – – S102A S102D – – S102A S102D Rescue – – – S102A S102D Time (min) Time (h)
Hypoxia Hypoxia

Fig. 9 AMPK phosphorylates ALDH7A1. Quantitative results are shown as mean with standard deviation; *p < 0.05, NS not significant, paired two-tailed
Student’s t-test. Source data are provided as a Source Data file. a The in vitro kinase assay was performed, followed by immunoblotting for phospho-AMPK
substrate (upper panel) or ALDH7A1 (lower panel); n = 2 independent experiments with a representative result shown. b HeLa cells were treated as
indicated. ALDH7A1 was then immunoprecipitated from whole-cell lysates, followed by immunoblotting for phospho-AMPK substrate (upper panel) or
ALDH7A1 (lower panel); n = 2 independent experiments with a representative result shown. c HeLa cells were transfected with constructs as indicated.
Cells were than subjected to hypoxia, followed by immunoprecipitation of ALDH7A1 and then immunoblotting for ALDH7A1 or the phospho-AMPK
substrate antibody; n = 2 independent experiments with a representative result shown. d Immunoblotting of whole-cell lysates showing efficacy of siRNA
against ALDH7A1, and the level of ALDH7A1 mutants expressed for rescue studies; n = 2 independent experiments with a representative result shown.
e COPI transport from the Golgi to the ER in HeLa cells was assessed through the quantitative colocalization of VSVG-KDELR with a cis-Golgi marker
(giantin); n = 3 independent experiments. f Fluid-phase endocytosis of dextran in HeLa cells was assessed through the quantitative colocalization of
internalized dextran with an early endosome marker (EEA1); n = 3 independent experiments. g COPI transport from the Golgi to the ER in HeLa cells was
assessed through the quantitative colocalization of VSVG-KDELR with a cis-Golgi marker (giantin); n = 3 independent experiments. h Fluid-phase
endocytosis of dextran in HeLa cells was assessed through the quantitative colocalization of internalized dextran with an early endosome marker (EEA1);
n q= 3 independent experiments. i HeLa cells were treated as indicated, and then total ATP level was quantified; n = 3 independent experiments. j HeLa
cells were treated as indicated, and then cell death was quantified; n = 3 independent experiments. k HeLa cells were treated with starvation conditions as
indicated, and then total ATP level was quantified; n = 3 independent experiments. l HeLa cells were treated with starvation conditions as indicated, and
then cell death was quantified; n = 3 independent experiments

membrane (Fig. 10b). To ascertain that the phosphorylation observed for the ER, which is consistent with our findings above
of the S102 residue promotes membrane recruitment, rather that starvation and hypoxia do not inhibit transport from the ER.
than inhibiting membrane release, we incubated the phosphor- To gain further insight into how membrane recruitment
ylation mutants as recombinant proteins with Golgi membrane, promotes transport inhibition by ALDH7A1, we next considered
and found that the S102D mutant becomes membrane-bound that an isoform of nicotinamide mononucleotide adenylyltrans-
after the incubation, while the S102A mutant remains soluble ferase (NMNAT), which catalyzes the synthesis of NAD from
(Fig. 10c). NMN (nicotinamide mononucleotide), has been found to exist at
Complementing the above results, we found by subcellular the Golgi31. We first confirmed that Golgi membrane contains an
fractionation that hypoxia and starvation, conditions that activate appreciable level of NAD (Fig. 10d). We then found that simply
AMPK phosphorylation of ALDH7A1, enhance the membrane incubating recombinant ALDH7A1 with Golgi membrane results
distribution of ALDH7A1 (Supplementary Fig. 9a). Moreover, in this NAD being converted to NADH (Fig. 10d). Notably, this
confocal microscopy revealed that the localization of ALDH7A1 result suggested that Golgi membrane contains not only NAD,
to multiple intracellular compartments is enhanced in these but also substrates of ALDH7A1, as its catalytic activity requires
conditions (Supplementary Fig. 9b). Notably, no enhancement is both substrate and cofactor to drive the reaction.

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 11


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

a 2.0

1.5

NADH (pmol)
b c S102D S102A
1.0 S102A S102D WT
P S P S
NS M C M C M C
0.5
49
49

0.0
WT S102D S102A

d O NADH e 300 *
70 NAD

60
Nucleotide (pmol)

NADH (pmol)
50 200

40
30 100
20
10
0
0 S102A S102D
Golgi Golgi + ALDH7A1

f g 40
*

% cell with tubulated Golgi


S102A S102D 30
S P S P
20
49 BARS

19
Cbv 10

6
0
Ctl S102D S102A

Fig. 10 Phosphorylation of ALDH7A1 regulates its membrane recruitment. Source data are provided as a Source Data file. a Recombinant ALDH7A1 was
incubated with a known substrate (octanal) and NAD, and the catalytic activity was tracked through the generation of NADH. Mean with standard
deviation is shown; NS not significant, ANOVA; n = 3 independent experiments. b HeLa cells were transfected with different myc-tagged ALDH7A1
constructs as indicated, followed by subcellular fractionation to obtain total cytoplasmic membrane (M) and cytosol (C), and then immunoblotting using
anti-myc antibody; n = 2 independent experiments with a representative result shown. c Recombinant forms of ALDH7A1 mutants were incubated with
Golgi membrane, followed by centrifugation to obtain membrane (P) vs. soluble (S) fractions, and then immunoblotting for ALDH7A1; n = 2 independent
experiments with a representative result shown. d Golgi membrane were treated as indicated, and then the levels of NAD and NADH on the membrane
were quantified; n = 3 independent experiments with a representative result shown. e Liposomes were generated using purified lipids to mimic the
composition of Golgi membrane. A substrate of ALDH7A1 (octanal) and NAD were then added, followed by incubation with different forms of recombinant
ALDH7A1 as indicated. The level of NADH was then quantified. Mean with standard deviation is shown; *p < 0.05, paired two-tailed Student’s t-test; n = 3
independent experiments. f Golgi membrane was incubated with mutant forms of ALDH7A1 as indicated, followed by centrifugation to re-isolate Golgi
membrane (P, pellet) vs. soluble (S) fraction. Both fractions were then immunoblotted for endogenous BARS; n = 2 independent experiments with a
representative result shown. g HeLa cells that expressed the different mutant forms of ALDH7A1 as indicated, achieved by siRNA against endogenous
ALDH7A1 followed by transfection of mutant forms, were examined by immunofluorescence microscopy that tracked VSVG leaving the TGN. Cells that
showed VSVG-positive Golgi tubules were then quantified; n = 3 independent experiments

We next considered that native membrane is complex, of liposomes enhances ALDH7A1 activity by nearly 100-fold
containing a myriad of membrane-bound proteins that could (Supplementary Table 3).
potentially mediate an indirect way by which ALDH7A1 We also further scrutinized how NADH generated by ALDH7A1
converts NAD on Golgi membrane to NADH. Thus, to confirm inhibits BARS. When the Golgi membrane was incubated with the
that ALDH7A1 on the membrane can directly convert NAD to S102A mutant of ALDH7A1, which has reduced capacity to
NADH, we generated liposomes with a lipid composition that generate NADH (see Fig. 10e), we observed BARS to remain on the
mimics the lipid profile of the Golgi membrane. To this mix, we membrane (Fig. 10f). In contrast, when the Golgi membrane was
added known aldehyde substrates of ALDH7A1 along with incubated with the S102D mutant, which generates elevated level of
NAD. Upon the incubation of recombinant ALDH7A1 with NADH (see Fig. 10e), we observed BARS to become released from
these liposomes, we observed the generation of NADH, and the membrane (Fig. 10f). Gel filtration revealed that the release
notably, this generation exhibits physiologic specificity, as only BARS exists as a dimer (Supplementary Fig. 10b). We also
the S102D mutant induces NADH generation, but not the considered that BARS has been shown to induce the fission of
S102A mutant (Fig. 10e). We also confirmed that the tubular carriers that emanate from the trans-Golgi network (TGN)
reconstituted ALDH7A1 activity requires the presence of both for transport to the PM23. We found that the expression of the
NAD and substrate (Supplementary Fig. 10a). We then S102D mutant of ALDH7A1 prevents this fission role of BARS,
performed enzyme kinetics, which revealed that the presence while the S102A mutant had no appreciable effect (Fig. 10g and

12 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0 ARTICLE

Supplementary Fig. 10c). Thus, the collective results further oxidative stress, which is mediated through the metabolic con-
supported that phosphorylation of the S102 residue in ALDH7A1 version of aldehydes to carboxylates. This possibility predicts that
promotes its role in transport inhibition by enhancing its ALDH7A1 activity may be particularly important for cell survival
recruitment from the cytosol to the membranes. in pathologic conditions when both types of stresses are present.
We further note that hereditary deficiency in ALDH7A1
activity has been identified32. As we have found that ALDH7A1 is
Discussion not critical under normal (non-stressed) conditions, this finding
ALDH7A1 activity is currently known to act in lysine metabo- explains how such humans can exist. However, there are situa-
lism32 and osmotic stress18. In this study, we find that situations tions when organs of the human body can encounter substantial
of energy stress induce AMPK to phosphorylate ALDH7A1, hypoxia and starvation. A major pathologic example is cardiac
which results in its redistribution from the cytosol to intracellular ischemia. As such, our finding that ALDH7A1 protects the cell
membrane compartments to exert a broad inhibition of the against situations of energy deprivation not only predicts that
intracellular transport pathways. Studying COPI transport, we cardiac ischemia should be more devastating for patients with
have further elucidated that rather than the metabolic conversion deficient ALDH7A1 activity, but also suggests a therapeutic target
of aldehydes to carboxylates, it is the reductive consequence of for the treatment of this major cause of human morbidity and
this reaction, which generates NADH from NAD, that underlies mortality.
how ALDH7A1 inhibits COPI transport, as NADH generated by
ALDH7A1 targets BARS to inhibit COPI vesicle fission. Methods
Many metabolic enzymes generate NADH from NAD through Chemicals and proteins. NAD, NADH, AICAR, hexanoic acid, octanoic acid,
their dehydrogenase activity. However, whereas the NADH octanal, betaine aldehyde, betaine, and 2-aminoadipic acid were obtained (Sigma).
generated by these enzymes is typically cytosolic, we find that Cholesterol, sphingomyelin, DOPC, DOPE, DOPS, and phosphatidic acid (PA)
were obtained (Avanti). Alexa 546-conjugated transferrin (Tf), Alexa 555-
ALDH7A1 also generates membrane-bound NADH. Thus, conjugated forms of EGF, dextran, and CT were also obtained (Invitrogen). Fugene
because BARS only interacts with ALDH7A1 on membranes, 6 Transfection Reagent was from Promega. RNAiMax was from Invitrogen. Pre-
these observations suggest how ALDH7A1 targets BARS with parations of coatomer, ARF1, ARFGAP1, BARS, Golgi membrane, and cytosol
efficiency and specificity. We further note that, whereas NADH is have been described14,16. To generate recombinant ALDH7A1, the human cDNA
was expressed in bacteria (BL21) using the pET-15b vector (Novagen), followed by
well known to act in cellular energetics through its participation isolation of the 6×-his tagged form of ALDH7A1 using a nickel column. AMPK
in glycolysis and mitochondrial oxidative phosphorylation, we was purified from cells by transfection of a flag-tagged construct into HeLa cells,
have uncovered a fundamentally different way that NADH gen- followed by immunoprecipitation of the tagged construct from cell lysates using an
erated by ALDH7A1 on membranes promotes energy home- anti-flag antibody (M2).
ostasis, by reducing energy consumption through transport
inhibition. Cells. HeLa and HEK293 cells were obtained from American Type Culture Col-
Our findings also advance a mechanistic understanding of COPI lection (ATCC), which were documented to be free of mycoplasma contamination.
Cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM) with 10% fetal
vesicle formation. Early studies identified a major mechanism of bovine serum and supplemented with glutamine. Starvation medium consisted of
regulation, which involves the small GTPase ARF1 regulating the phosphate-buffered saline supplemented with 10% dialyzed bovine serum. Hypoxia
recruitment of coatomer, the core components of the COPI com- studies were performed by incubating cells in a hypoxia chamber (filled with pre-
plex, from the cytosol to membranes9. In the current study, we find mixed air that contains 1% oxygen) at 37 °C for 16 h.
that ALDH7A1 regulates the fission stage of COPI vesicle forma-
tion. We further note that we have recently identified another Plasmids. Mammalian expression vectors, which contain temperature-sensitive
metabolic enzyme, GAPDH, also to inhibit COPI vesicle fission19. mutations (ts-045) of VSVG and VSVG-KDELR, have been described24. The BARS
mutant (G172E) has been described previously16, and was subcloned into
Whereas ALDH7A1 targets BARS, GAPDH targets ARFGAP1. pcDNA3.1 for transfection studies. Flag-AMPK in pcDNA3 was a gift from Dr.
Thus, the collective findings from these two studies suggest that the Bing Zheng (Columbia University, NY). ALDH7A1 was subcloned into mamma-
fission stage represents another major node by which COPI lian expression vectors, pcDNA3.1 and pEGFP-N1. Mutant ALDH7A1 (E268Q)
transport can be targeted for regulation. was generated using QuikChange Site-Directed-Mutagenesis (Stratagene) with
paired oligonucleotides: 5′-gggagaagtctgttgcaacttggaggaaacaatgcc-3′ and 5′-
It is further notable that the intracellular transport pathways ggcattgtttcctccaagttgcaacagacttctccc-3′. Mutant ALDH7A1 (S102A) was generated
have not been known to be a major target by which significant with paired oligonucleotides: 5′-aagatccaagtactaggagccttggtgtctttggagatgggg-3′ and
reduction in cellular energy consumption can be achieved. An 5′-ccccatctccaaagacaccaaggctcctagtacttggatctt-3′. Mutant ALDH7A1 (S102D) was
explanation is suggested by the consideration that studies on generated with paired oligonucleotides: 5′-aagatccaagtactaggagacttggtgtctttgga-
vesicular transport have typically focused on single pathways. gatgggg-3′ and 5′-ccccatctccaaagacaccaagtctcctagtacttggatctt-3′. Mutant ALDH7A1
(S146A) was generated with paired oligonucleotides: 5′-atcttgccttctgaaa-
In contrast, we have found that, by coordinating inhibition across gatctggccatgcactgattgag-3′ and 5′- ctcaatcagtgcatggccagatctttcagaaggcaagat-3′.
multiple pathways, a meaningful reduction in energy consump-
tion can be achieved to promote energy homeostasis during Antibodies. Rabbit antibody against human ALDH7A1 (WB: 1:1000, IF: 1:500)
situations of energy stress. This explanation is further supported was generated by injecting the recombinant protein into rabbits using a com-
by our recent elucidation that GAPDH also relieves energy stress mercial service (Proteintech). The following antibodies have been described in our
by exerting a broad inhibition of the transport pathways19. previous studies14,16,24,35: ARF1 (WB 1:1000), BARS (WB 1:1000), coatomer (IF
Besides having roles in lysine metabolism32 and osmotic 1:10), β-COP (WB 1:10), calnexin (IF 1:200), cellubrevin (WB 1:1000), giantin
(IF 1:2000), GM130 (IF 1:500), Lamp1 (IF 1:200), Sec61p (IF 1:500), TGN46 (IF
stress18, ALDH7A1 activity has also been suggested to protect the 1:200), VSVG (IF 1:10), HA epitope tag (WB 1:200), and Myc epitope tag (WB
cell against lipid peroxidation22,33, which is a form of oxidative 1:200). The following antibodies were obtained from commercial sources: actin
stress that occurs in multiple pathologic conditions34. As we have (WB 1:1000, Ambion, AM4302), EEA1 (1:100 IF, BD Transduction Laboratories,
elucidated that transport inhibition by ALDH7A1 requires its 610456), GFP (WB 1:1000, Thermo Scientific, MA5-15256), GST (WB 1:1000,
Santa Cruz Biotechnology, sc-138), and Rab11 (IF 1:200, BD Biosciences, 610656).
recruitment to membranes, and lipid peroxidation also occurs on Other primary antibodies were obtained from Cell Signaling: ALDH1A1 (12035S,
membranes, an intriguing possibility is that ALDH7A1 activity WB 1:1000), AMPKα1 (2603S, WB 1:500), phospho-substrate-AMPK antibody
on membranes coordinates two general processes to protect (5759S, WB 1:500), Lamin B (12586S, WB 1:1000), and Sirt1 (2493S, WB 1:500).
against two major types of cellular stress: (i) inducing transport Conjugated secondary antibodies were obtained from Jackson ImmunoResearch:
horseradish peroxidase-conjugated donkey antibodies against mouse IgG (715-035-
inhibition to reduce energy consumption in protecting against 150, WB 1:10,000) and against rabbit IgG (711-035-152, WB 1:10,000), Cy2 donkey
energy stress, which is mediated through the generation of antibodies against mouse IgG (715-225-151, IF 1:200) and against rabbit IgG (711-
NADH, and (ii) reducing lipid peroxidation to protect against 225-152, IF 1:200), Cy3 goat antibody against mouse IgG (115-165-062, IF 1:200),

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 13


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

Cy3 donkey antibodies against rabbit IgG (711-165-152, IF 1:200) and against For the endocytosis of EGF, cells were incubated with Alexa 555-conjugated
sheep IgG (713-165-147, IF 1:200). EGF (1 μg/ml in DMEM) for 1 h at 4 °C. Cells were then washed to clear unbound
EGF, followed by shifting to 37 °C for times indicated in the figures. Cells were
stained for EEA1, followed by confocal microscopy to assess the arrival of EGF to
Mass spectrometry. Cytosol was purified from rat livers by homogenization in the early endosome.
lysis buffer (1 mM Tris pH 7.4, 800 mM sucrose, 5 mM EDTA, and protease
inhibitors) at 4 °C, followed by centrifugation at 80,000×g for 90 min. The resulting Confocal microscopy. Colocalization studies were performed using two confocal
supernatant was dialyzed against dialysis buffer (25 mM Tris at pH 8.0, 50 mM systems. The Nikon system is equipped with the Nikon Eclipse TE2000U Inverted
KCl, and 1 mM DTT), followed by centrifugation at 150,000×g for 90 min at 4 °C. Microscope having a Plan Apo 60×/1.40 oil objective, Nikon D-Eclipse C1 confocal
Aliquots were stored at −80 °C. Pulldown experiment was performed by incu- package with a 488 Laser (having 515/30 emission filter) and a 543 Laser (having
bating GST or GST-BARS on beads (5 μg) with 50 μg rat liver cytosol in traffic 590/50 emission filter), and Nikon EZ-C1 version 3.90 acquisition software. The
buffer (25 mM HEPES pH 7.2, 50 mM KCl, and 2.5 mM Mg(OAc)2) for 1 h fol- Zeiss system is equipped with the Zeiss Axio Observer Z1 Inverted Microscope
lowed by extensive washing. Samples were separated by SDS-PAGE and then having a Plan-Apochromat 63× objective, the Zeiss LSM 800 with Airyscan con-
stained with Coomassie blue. Specific bands were excised for protein identification focal package with Zeiss URGB (488 and 561 nm) laser lines, and Zen 2.3 blue
by mass spectrometry (Taplin Mass Spectrometry Facility, Harvard Medical edition confocal acquisition software.
School, USA). Interacting proteins were identified for both GST and GST-BARS. For quantitation of colocalization, ten fields of cells were examined, with each
Proteins that only interacted with GST-BARS were considered specific. field typically containing five cells. Images were imported into the NIH Image J
version 1.50e software, and then analyzed through a plugin software (https://
imagej.net/Coloc_2). Under the “Image” tab, the “Split Channels” option was
Transfections and siRNA. Transfection of DNA plasmids was performed using selected. Under the “Plugins” tab, “Colocalization Analysis” option was selected,
FuGene6 (Roche). Transfection of siRNA was performed using Lipofectamine and within this option, the “Colocalization Threshold” option was selected.
RNAiMAX (Invitrogen). Plasmid transfections occurred over 48 h and siRNA Colocalization values were then calculated by the software, and expressed as the
transfections occurred over 72 h. The siRNA sequences used to target human fraction of protein of interest (cargo or ALDH7A1) colocalized with an organelle
ALDH7A1, 5′-gcagugagcauguuucuugtt-3′ (sense strand) and 5′-caagaaacaugcuca marker.
cugctt-3′ (complement strand), were obtained (Dharmacon). Rescue plasmids for
wild-type and catalytic dead mutant of human ALDH7A1 were generated by
targeting this siRNA sequence using QuikChange Site-Directed-Mutagenesis In vitro reconstitution of COPI vesicle formation. The reconstitution system was
(Stratagene) with paired oligonucleotides: 5′-cacaccaagcaggcagtgtcgatgtttcttggag performed essentially as previously described14,16. Briefly, Golgi membrane
cagtg-3′ and 5′-cactgctccaagaaacatcgacactgcctgcttggtgtg-3′. Oligonucleotides that (0.2 mg/ml) was washed with 3 M KCl, and then incubated with ARF1 (6 μg/ml)
target human AMPKα1, 5′-agugaagguuggcaaacautt-3′ (sense strand) and 5′- and coatomer (6 μg/ml) for the first-stage incubation that reproduces the ARF-
auguuugccaaccuucacutt-3′ (complement strand), human ALDH1A1, 5′-guagccuu dependent recruitment of coatomer onto the Golgi membrane. The Golgi mem-
cacaggaucaauu-3′ (sense strand) and 5′-uugauccugugaaggcuacuu-3′ (complement brane was re-isolated and then incubated with ARFGAP1 (2 μg/ml) and BARS
strand), human catalase, 5′-uggauauggaucacauacu-3′ (sense strand) and 5′- (2 μg/ml) for the second stage that results in vesicle formation. ALDH7A1 (1 μg/
aguaugugauccauaucca-3′ (complement strand), human Ftcd, 5′-ccaaucuucugga ml) was added at the second stage. Products of ALDH7A1 activity were also added
cuuuga-3′ (sense strand) and 5′-ucaaaguccagaagauugg-3′ (complement strand), at this stage. These include: hexanoic acid (20 μM), octanoic acid (20 μM), 2-
human malate dehydrogenase 2, 5′-uggugagccgccugacccu-3′ (sense strand) and 5′- aminoadipic acid (20 μM), or NADH (20 μM).
agggucaggcggcucacca-3′ (complement strand), and human aldolase, 5′-ccgagaa EM examination of Golgi membrane using the whole-mount technique has
caccgaggagaa-3′ (sense strand) and 5′-uucuccucgguguucucgg-3′ (complement been described previously14,16. In brief, membrane samples from the COPI
strand) were also obtained (Dharmacon). Targeting specificities of these siRNAs reconstitution system were spotted onto EM grids and then fixed with 2% PFA/PBS
have been documented previously36,37. for 10 min. Grids were rinsed three times with water, followed by 1% uranyl acetate
staining, and then examined using JEOL 1200EX Transmission electron
microscope.
In vivo transport assays. Quantitative microscopy-based transport assays of the
different intracellular pathways has been described previously24. In brief, they are Subcellular fractionation. Cells were washed with PBS, resuspended in homo-
done as follows. genization buffer (0.25 M sucrose, 1 mM EDTA, and 20 mM HEPES-KOH, pH 7.4
For anterograde transport from ER to Golgi, cells were transfected with pROSE- and protease inhibitor cocktail) and then disrupted by passing through 28-gauge
VSVG-ts045-Myc for 1 day, and then incubate at 39 °C for 4 h to accumulate needles. After low-speed centrifugation (800×g for 6 min) to spin out nuclei and
VSVG in the ER. Cells were then shifted to 32 °C for different times as indicated in unbroken cells, the resulting post-nuclear supernatant was centrifuged at
figures. Cells were then stained for giantin, followed by confocal microscopy to 100,000×g for 1 h to obtain cytosol and total membrane fractions.
assess the arrival of VSVG to the Golgi. To obtain nuclear vs. cytoplasmic fractions, cells were washed with PBS and
For retrograde transport from the Golgi to the ER, cells were transfected with resuspended into hypotonic buffer containing 10 mM HEPES, pH 8.0, 10 mM KCl,
pROSE-VSVG-ts045-KDELR-Myc for 1 day, and then incubated at 32 °C for 8 h to 3 mM MgCl2, 0.5 mM DTT, and protease inhibitors. The suspension was then
achieve steady-state distribution at the Golgi. Cells were then shifted to 39 °C for incubated on ice for 10 min and Triton X-100 was added to a final concentration of
different times as indicated in the figures. Cells were then stained for giantin, 0.3%. After low-speed centrifugation (800×g for 5 min), the supernatant was
followed by confocal microscopy to assess the exit of VSVG-KDELR from collected as the cytoplasmic fraction. The pellet was washed twice with hypotonic
the Golgi. buffer again and resuspended in the lysis buffer containing 100 mM Tris-HCl, pH
For anterograde transport from the Golgi to the PM, cells were transfected with 8.0, 1% Triton X-100, 100 mM NaCl, 0.5 mM EDTA, and protease inhibitors. After
pROSE-VSVG-ts045-Myc for 1 day, and then incubated at 20 °C for 2 h to centrifugation at 15,000×g for 10 min, the supernatant was collected as the nuclear
accumulate VSVG at the TGN. Cells were then shifted to 32 °C for different times fraction.
as indicated in the figures. Cells were then stained for TGN46, followed by confocal
microscopy to assess the exit of VSVG from the Golgi.
In vitro studies using liposomes. Liposomes were generated from pure defined
For the recycling of Tf from the early recycling endosome to the PM, cells were
lipids to mimic the composition of native membrane (mol%): dioleoyl-
incubated with Alexa 546-conjugated Tf (5 μg/ml in DMEM) at 37 °C for 2 h to
phosphatidylcholine (DOPC) 50%, dioleoyl-phosphatidylethanolamine (DOPE)
allow the steady-state accumulation of Tf in endosomes. Subsequently, cells were
16%, dioleoyl-phosphatidylserine (DOPS) 5%, PA 5%, cholesterol 17%, and
incubated with a medium without Tf for different times as indicated in the figures.
sphingomyelin 7%. These pure lipids were mixed in a glass tube, dried under
Cells were then stained for Rab11, followed by confocal microscopy to assess the
nitrogen, and then resuspended in buffer (25 mM Hepes–KOH, pH 7.2, 50 mM
exit of Tf from the early recycling endosome.
KCl, 2.5 mM Mg(OAc)2, and 0.2 M sucrose), which involved ten times of rapid
For the retrograde transport of CT from the PM to the Golgi, cells were
freeze and thaw with occasional vortexing. The generated giant liposomes were
incubated with Alexa 555-conjugated CT for 30 min at 4 °C (0.5 μg/ml in DMEM).
then passed through a mini-extruder (400-nm filter) 21 times.
After washing to clear unbound CT, cells were shifted to 37 °C for different times as
To reconstitute ALDH7A1 activity in the context of liposomes, liposomes
indicated in the figures. Cells were then stained for TGN46, followed by confocal
(20 μg) were mixed with NAD (10 μM) and substrate (octanal (10 μM) or betaine
microscopy to assess the arrival of CT to the Golgi.
aldehyde (10 μM)) in 200 μl of PBS, and then recombinant ALDH7A1 (1 μg) was
For endocytic transport of EGF to the lysosome, cells were incubated with Alexa
added. Incubation was performed at 37 °C for 1 h. Subsequently, NADH
555-conjugated EGF (1 μg/ml in DMEM) for 1 hour at 4 °C. Cells were then
measurement was performed using the enzyme cycling assay. Enzyme kinetics of
washed to clear unbound EGF, followed by shifting to 37 °C for times indicated in
ALDH7A1 were determined as previously described22. To calculate Km, incubation
the figures. Cells were stained for Lamp1, followed by confocal microscopy to
was performed by varying the level of substrate in the presence of excess NAD.
assess the arrival of EGF to the lysosome.
For the fluid-phase uptake of dextran, cells were incubated with Alexa 555-
conjugated dextran (0.2 mg/ml) at 37 °C for different times as indicated in the In vitro kinase assay. The kinase assay was performed essentially as previously
figures. Cells were then stained for EEA1, followed by confocal microscopy to described19. For phosphorylation detection using the AMPK phospho-substrate
assess the arrival of dextran to the early endosome. antibody, myc-tagged AMPK1 was isolated from transfected cells that had been

14 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0 ARTICLE

starved and treated with AICAR. For phosphorylation detection using γP32-ATP or 6. Sorkin, A. & von Zastrow, M. Endocytosis and signalling: intertwining
using the ADP-Glo Kinase assay (Promega), recombinant active AMPK was molecular networks. Nat. Rev. Mol. Cell Biol. 10, 609–622 (2009).
obtained (Sinobiological). In all cases, AMPK was incubated with recombinant 7. Henne, W. M., Buchkovich, N. J. & Emr, S. D. The ESCRT pathway. Dev. Cell
ALDH7A1 in the presence of ATP and phosphatase inhibitors. To assess the 21, 77–91 (2011).
stoichiometry of phosphorylation, the SAMS peptide, a sequence from an optimal 8. Cullen, P. J. Endosomal sorting and signalling: an emerging role for sorting
AMPK substrate, ACC, was used for comparison. nexins. Nat. Rev. Mol. Cell Biol. 9, 574–582 (2008).
9. Hsu, V. W., Lee, S. Y. & Yang, J. S. The evolving understanding of COPI
Other assays. Total cellular ATP level was detected using ATPlite Luminescence vesicle formation. Nat. Rev. Mol. Cell Biol. 10, 360–364 (2009).
Detection Assay System (Perkin Elmer), and was performed according to manu- 10. Glick, B. S. & Nakano, A. Membrane traffic within the Golgi apparatus. Annu.
facturer’s instructions, with final values normalized to number of viable cells. Cell Rev. Cell Dev. Biol. 25, 113–132 (2009).
viability was assessed by incubating cells with propidium iodine (1 ng/μl) and then 11. Shiba, Y. & Randazzo, P. A. ArfGAP1 function in COPI mediated membrane
quantified by flow cytometry. NAD/NADH measurements were performed using the traffic: currently debated models and comparison to other coat-binding
standard enzyme cycling assay according to manufacturer’s instruction (BioVision). ArfGAPs. Histol. Histopathol. 27, 1143–1153 (2012).
12. Waters, M. G., Serafini, T. & Rothman, J. E. ‘Coatomer’: a cytosolic protein
complex containing subunits of non-clathrin-coated Golgi transport vesicles.
mRNA quantification. Total RNA was isolated using the PureLink kit (Ambion).
Total mRNA was then isolated using the MicroPolyAPurist kit (Ambion). Both Nature 349, 248–251 (1991).
were done according to manufacturer’s instructions. Quantitation was performed 13. Donaldson, J. G., Cassel, D., Kahn, R. A. & Klausner, R. D. ADP-ribosylation
by measuring optical density at 260 nm and normalizing for cell number. Quan- factor, a small GTP-binding protein, is required for binding of the coatomer
titation of RNA transcript using real-time PCR (qPCR) has been described38. The protein beta-COP to Golgi membranes. Proc. Natl Acad. Sci. USA 89,
cycling conditions were: denaturation at 95 °C for 30 s, annealing at 56 °C for 60 s, 6408–6412 (1992).
and extension at 72 °C for 30 s. Primer sequences used to detect the mRNA for E- 14. Yang, J. S. et al. ARFGAP1 promotes the formation of COPI vesicles,
cadherin, 5′-tcgacacccgattcaaagtgg-3′ (forward) and 5′-gtgggttatgaaaccgtagagg-3′ suggesting function as a component of the coat. J. Cell Biol. 159, 69–78 (2002).
(reverse), for ALDH7A1, 5′-tttccctgtggcagtgtatg-3′ (forward) and 5′-cctcca- 15. Shiba, Y. et al. ArfGAP1 promotes COPI vesicle formation by facilitating
gaaccttggctattatc-3′ (reverse), for Ftcd, 5′-aacctgctcggcacaaa-3′ (forward) and 5′- coatomer polymerization. Cell. Logist. 1, 139–154 (2011).
ctgagccaggttcttctcatc-3′ (reverse), for Catalase, 5′-ctccactgttgctggagaat-3′ (forward) 16. Yang, J. S. et al. A role for BARS at the fission step of COPI vesicle formation
and 5′-cgagatcccagttaccatcttc-3′ (reverse), for ALDO, 5′-cctcttccatgagacactctac-3′ from Golgi membrane. EMBO J. 24, 4133–4143 (2005).
(forward) and 5′-cgcccttgtctaccttgat-3′ (reverse), and for MDH2, 5′-agccacttt 17. Yang, J. S. et al. A role for phosphatidic acid in COPI vesicle fission yields
cacttctcctg-3′ (forward) and 5′-tttggtctcgatgtggctc-3′ (reverse), were purchased insights into Golgi maintenance. Nat. Cell Biol. 10, 1146–1153 (2008).
from Integrated DNA Technologies, Inc. (IDT). 18. Fong, W. P., Cheng, C. H. & Tang, W. K. Antiquitin, a relatively unexplored
member in the superfamily of aldehyde dehydrogenases with diversified
physiological functions. Cell. Mol. Life Sci. 63, 2881–2885 (2006).
Gel filtration. Analysis was performed using the AKTA UPC-900 FPLC system
(GE Healthcare) with a Superdex 200 10/200GL column, with the following 19. Yang, J. S. et al. GAPDH inhibits intracellular pathways during starvation for
parameters: sample volume 500 µl, eluent buffer 20 mM Tris-HCl, 100 mM NaCl, cellular energy homeostasis. Nature 561, 263–267 (2018).
pH 7.2, flow rate 0.5 ml/min. 20. Yang, J. S. et al. COPI acts in both vesicular and tubular transport. Nat. Cell
Biol. 13, 996–1003 (2011).
21. Wang, X. & Weiner, H. Involvement of glutamate 268 in the active site of
Statistical analysis. Sample size is noted in the figure legends. Sample size used human liver mitochondrial (class 2) aldehyde dehydrogenase as probed by
was based on our previous familiarity with the assays. For comparison between two site-directed mutagenesis. Biochemistry 34, 237–243 (1995).
conditions, significance was tested by the paired two-tailed Student’s t-test. For 22. Brocker, C. et al. Aldehyde dehydrogenase 7A1 (ALDH7A1) is a novel enzyme
comparison among multiple conditions, significance was tested by the analysis of involved in cellular defense against hyperosmotic stress. J. Biol. Chem. 285,
variance (ANOVA). These tests were performed using Excel or Prism software. No 18452–18463 (2010).
inclusion/exclusion criteria were pre-established. The experiments were not ran-
23. Bonazzi, M. et al. CtBP3/BARS drives membrane fission in dynamin-
domized. The investigators were not blinded to the group allocation during
independent transport pathways. Nat. Cell Biol. 7, 570–580 (2005).
experiments and in outcome assessment.
24. Bai, M. et al. ARFGAP1 promotes AP-2 dependent endocytosis. Nat. Cell Biol.
13, 559–567 (2011).
Reporting Summary. Further information on research design is available in the 25. Zhang, Q. et al. Redox sensor CtBP mediates hypoxia-induced tumor cell
Nature Research Reporting Summary linked to this article. migration. Proc. Natl Acad. Sci. USA 103, 9029–9033 (2006).
26. Hardie, D. G., Ross, F. A. & Hawley, S. A. AMPK: a nutrient and energy
sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 13,
Data availability
251–262 (2012).
The data that support the findings of this study are available from the corresponding
27. Carling, D., Mayer, F. V., Sanders, M. J. & Gamblin, S. J. AMP-activated
author upon reasonable request. A reporting summary for this Article is available as a
protein kinase: nature’s energy sensor. Nat. Chem. Biol. 7, 512–518 (2011).
Supplementary Information file. The source data for Figs. 1a–j, 2a–g, 2i, j, 3a–i, 4a, 4c,
28. Mihaylova, M. M. & Shaw, R. J. The AMPK signalling pathway coordinates
4e–h, 5a–h, 6a–h, 7a–k, 8a–j, 9a–l, 10a–g, Supplementary Figs. 1a–d, 2d, 3g–i, 4a–i, 5a–i,
cell growth, autophagy and metabolism. Nat. Cell Biol. 13, 1016–1023 (2011).
6a–i, 7a–d, 8a–d, 9a, b, 10a, b, and Supplementary Tables 2 and 3 are provided as a
29. Chang, C. et al. AMPK-dependent phosphorylation of GAPDH riggers Sirt1
Source Data file. Uncropped scans of immunoblots are also provided in this Source Data
file. The mass spectrometry proteomics data have been deposited to the
activation and is necessary for autophagy upon glucose starvation. Mol. Cell
60, 930–940 (2015).
ProteomeXchange Consortium via the PRIDE39 partner repository with the data set
30. Manning, B. D., Tee, A. R., Logsdon, M. N., Blenis, J. & Cantley, L. C.
identifier PXD014645.
Identification of the tuberous sclerosis complex-2 tumor suppressor gene
product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol.
Received: 20 July 2018 Accepted: 27 July 2019 Cell 10, 151–162 (2002).
31. Mayer, P. R. et al. Expression, localization, and biochemical characterization
of nicotinamide mononucleotide adenylyltransferase 2. J. Biol. Chem. 285,
40387–40396 (2010).
32. Mills, P. B. et al. Mutations in antiquitin in individuals with pyridoxine-
dependent seizures. Nat. Med. 12, 307–309 (2006).
References 33. Brocker, C., Cantore, M., Failli, P. & Vasiliou, V. Aldehyde dehydrogenase
1. Bonifacino, J. S. & Glick, B. S. The mechanisms of vesicle budding and fusion. 7A1 (ALDH7A1) attenuates reactive aldehyde and oxidative stress induced
Cell 116, 153–166 (2004). cytotoxicity. Chem. Biol. Interact. 191, 269–277 (2011).
2. Cai, H., Reinisch, K. & Ferro-Novick, S. Coats, tethers, Rabs, and SNAREs 34. Reed, T. T. Lipid peroxidation and neurodegenerative disease. Free Radic. Biol.
work together to mediate the intracellular destination of a transport vesicle. Med. 51, 1302–1319 (2011).
Dev. Cell 12, 671–682 (2007). 35. Dai, J. et al. ACAP1 promotes endocytic recycling by recognizing recycling
3. De Matteis, M. A. & Luini, A. Exiting the Golgi complex. Nat. Rev. Mol. Cell sorting signals. Dev. Cell 7, 771–776 (2004).
Biol. 9, 273–284 (2008). 36. Kwon, H. J. et al. Activation of AMP-activated protein kinase stimulates the
4. Haucke, V., Neher, E. & Sigrist, S. J. Protein scaffolds in the coupling of nuclear localization of glyceraldehyde 3-phosphate dehydrogenase in human
synaptic exocytosis and endocytosis. Nat. Rev. Neurosci. 12, 127–138 (2011). diploid fibroblasts. Exp. Mol. Med. 42, 254–269 (2010).
5. Conner, S. D. & Schmid, S. L. Regulated portals of entry into the cell. Nature 37. Moreb, J. S. et al. ALDH isozymes downregulation affects cell growth, cell
422, 37–44 (2003). motility and gene expression in lung cancer cells. Mol. Cancer 7, 87 (2008).

NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications 15


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-11932-0

38. Moisan, J., Grenningloh, R., Bettelli, E., Oukka, M. & Ho, I. C. Ets-1 is a negative Reprints and permission information is available online at http://npg.nature.com/
regulator of Th17 differentiation. J. Exp. Med. 204, 2825–2835 (2007). reprintsandpermissions/
39. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in
2019: improving support for quantification data. Nucleic Acids Res. 47, Peer review information: Nature Communications thanks Vasilis Vasiliou and other
D442–D450 (2019). anonymous reviewer(s) for their contribution to the peer review of this work. Peer
reviewer reports are available.

Acknowledgements Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
We thank Joao Paulo and Steve Gygi for advice and discussion. This work is supported published maps and institutional affiliations.
by grants from the National Institutes of Health to V.W.H. (GM058615 and GM115683)
and to I.C.H. (AR070171), from the Brigham and Women’s Hospital to J.S.Y. (the
Evergreen Innovation Fund), and from the Department of Defense to I.C.H. (W81XWH-
Open Access This article is licensed under a Creative Commons
11-1-0492).
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
Author contributions appropriate credit to the original author(s) and the source, provide a link to the Creative
J.S.Y., J.W.H., S.Y.P., S.Y.L., J.L., M.B., C.A., W.T., X.M., and I.C.H. performed the Commons license, and indicate if changes were made. The images or other third party
experiments. All authors participated in analyzing the data. V.W.H. supervised the work material in this article are included in the article’s Creative Commons license, unless
and wrote the manuscript with help from J.S.Y. indicated otherwise in a credit line to the material. If material is not included in the
article’s Creative Commons license and your intended use is not permitted by statutory
Additional information regulation or exceeds the permitted use, you will need to obtain permission directly from
Supplementary Information accompanies this paper at https://doi.org/10.1038/s41467- the copyright holder. To view a copy of this license, visit http://creativecommons.org/
019-11932-0. licenses/by/4.0/.

Competing interests: The authors declare no competing interests.


© The Author(s) 2019

16 NATURE COMMUNICATIONS | (2019)10:4068 | https://doi.org/10.1038/s41467-019-11932-0 | www.nature.com/naturecommunications


LAPORAN TUGAS JURNAL READING REMIDI BLOK MOLECULAR BIOLOGY
2022/2023

NO Komponen Pemaparan
1 Identitas Judul Artikel: ALDH7A1 Inhibits the Intracellular Transport Pathways
Jurnal During Hypoxia and Starvation to Promote Cellular Energy Homeostasis

Judul Jurnal: Intracellular Communication

DOI : https://doi.org/10.1038/s41467-019-11932-0

Penulis: Jia-Shu Yang, Jia-Wei Hsu, Seung-Yeol Park, Stella Y. Lee, Jian Li,
Ming Bai, Claudia Alves, William Tseng, Xavier Michelet, I-Cheng Ho &
Victor W. Hsu

2 Abstrak ALDH adalah enzim metabolik yang mengubah aldehida


menjadi karboksilat. Dalam penelitian ini, ALDH7A1 menghasilkan NADH
dari NAD, menyebabkan penghambatan transport intraseluler. NADH
menargetkan BARS untuk menghambat pemisahan vesikel COPI. Selain
itu, ALDH7A1 mengurangi konsumsi energi selama hipoksia dan kelaparan
untuk homeostasis energi seluler. Temuan ini meningkatkan pemahaman
tentang transport intraseluler dan peran NADH dalam energetika seluler.
3 Introduction/ Latar belakang penelitian ini adalah memahami mekanisme dan regulasi jalur
Pendahuluan transportasi intraseluler, khususnya transportasi vesikel COPI. Transportasi
vesikel merupakan salah satu proses kunci dalam sel, di mana vesikel (kantong
membran kecil) digunakan untuk mengangkut berbagai molekul dan zat di
antara berbagai kompartemen seluler, seperti dari golgi ke retikulum
endoplasma, atau dari golgi ke membran plasma.

Dipenelitian ini, banyak pemahaman tentang jalur transportasi intraseluler


berasal dari studi pada jalur model, termasuk jalur COPI. Jalur COPI
melibatkan kompleks mantel protein I (COPI) yang berperan dalam
membentuk vesikel untuk mengangkut molekul dari retikulum endoplasma ke
golgi dan antara kompartemen golgi.

Studi awal telah mengidentifikasi beberapa komponen utamai dalam jalur


COPI, termasuk kompleks multimerik (katomer) dan ARF1 (GTPase kecil)
yang mengatur rekrutmen katomer dari sitosol ke membran golgi untuk
membentuk vesikel COPI. Namun, meskipun banyak yang telah diketahui
tentang jalur COPI, masih ada banyak aspek yang perlu dipahami untuk
mendapatkan gambaran yang lebih lengkap tentang regulasi dan koordinasi
proses ini.

Penelitian ini bertujuan untuk mengeksplorasi dan mengidentifikasi faktor


tambahan yang berperan dalam pembentukan vesikel COPI. Dalam penelitian
ini, ditemukan bahwa enzim ALDH7A1, yang termasuk dalam keluarga
aldehid dehidrogenase (ALDH), memiliki peran dalam menghambat
pembelahan vesikel COPI dengan menargetkan BARS. Selain itu, penelitian ini
mengungkapkan bahwa ALDH7A1 memiliki peran luas dalam menghambat
beberapa jalur transportasi intraseluler lainnya. Temuan ini memberikan
wawasan baru tentang bagaimana koordinasi jalur intraseluler terjadi dan
bagaimana ALDH7A1 berperan dalam regulasi homeostasis energi seluler,
khususnya selama kondisi hipoksia dan kelaparan dimana produksi energi
seluler terpengaruh.

4 Material dan 1. Bahan kimia dan protein. NAD, NADH, AICAR, asam heksanoat, asam
Method oktanoat, oktanal, betain aldehida, betain, dan 2-aminoadipat asam diperoleh
dari Sigma. Kolesterol, sfingomielin, DOPC, DOPE, DOPS, dan asam
fosfatidat (PA) diperoleh dari Avanti. Alexa 546-terkonjugasi transferrin (Tf),
Alexa 555-terkonjugasi bentuk EGF, dekstran, dan CT juga diperoleh dari
Invitrogen. Reagen Fugene 6 Transfection diperoleh dari Promega. RNAiMax
diperoleh dari Invitrogen. Preparasi katomer, ARF1, ARFGAP1, BARS,
membran golgi, dan sitosol telah dijelaskan sebelumnya. Untuk menghasilkan
ALDH7A1 rekombinan, cDNA manusia diekspresikan dalam bakteri (BL21)
menggunakan vektor pET-15b (Novagen), diikuti dengan isolasi bentuk tag 6x
dari ALDH7A1 menggunakan kolom nikel. AMPK dimurnikan dari sel melalui
transfeksi konstruksi yang ber-tag flag ke sel HeLa, diikuti dengan
imunopresipitasi dari konstruksi ber-tag dari lisat sel menggunakan antibodi
anti-flag (M2).

2. Sel. Sel HeLa dan HEK293 diperoleh dari American Type Culture
Collection (ATCC), yang didokumentasikan bebas dari kontaminasi
mikoplasma. Sel-sel dibiakkan dalam Dulbecco's Modified Eagle Medium
(DMEM) dengan 10% serum fetal bovin dan ditambahkan glutamin. Medium
kelaparan terdiri dari saline-buffered fosfat dengan tambahan 10% serum bovin
yang difiltrasi. Studi hipoksia dilakukan dengan menginkubasi sel dalam ruang
hipoksia (berisi campuran udara yang mengandung 1% oksigen) pada suhu
37°C selama 16 jam.

3. Plasmid. Vektor ekspresi mamalia, yang mengandung mutasi suhu-sensitive


(ts-045) dari VSVG dan VSVG-KDELR, telah dijelaskan. Mutan BARS
(G172E) telah dijelaskan sebelumnya dan di-subkloning ke dalam pcDNA3.1
untuk studi transfeksi. Flag-AMPK dalam pcDNA3 diberikan oleh Dr. Bing
Zheng (Columbia University, NY). ALDH7A1 di-subkloning ke dalam vektor
ekspresi mamalia, pcDNA3.1 dan pEGFP-N1. Mutan ALDH7A1 (E268Q),
(S102A), (S102D), dan (S146A) dihasilkan menggunakan QuikChange Site-
Directed-Mutagenesis (Stratagene) dengan sepasang oligonukleotida tertentu.

4. Antibodi. Antibodi kelinci terhadap ALDH7A1 manusia (WB: 1:1000, IF:


1:500) dihasilkan dengan menyuntikkan protein rekombinan ke dalam kelinci
menggunakan layanan komersial (Proteintech). Antibodi lainnya dijelaskan
sebelumnya.

5. Spektrometri massa. Sitosol dihasilkan dari hati tikus dengan homogenisasi


dalam larutan lisis (1 mM Tris pH 7.4, 800 mM sukrosa, 5 mM EDTA, dan
inhibitor protease) pada 4°C, diikuti dengan sentrifugasi pada 80.000xg selama
90 menit. Supernatan yang dihasilkan diaduk dengan buffer lisis (25 mM Tris
pH 8.0, 50 mM KCl, dan 1 mM DTT), diikuti dengan sentrifugasi pada
150.000xg selama 90 menit pada 4°C. Sampel disimpan pada -80°C.
Eksperimen pulldown dilakukan dengan menginkubasi GST atau GST-BARS
pada kapsul (5 μg) dengan 50 μg sitosol hati tikus dalam larutan transportasi
(25 mM HEPES pH 7.2, 50 mM KCl, dan 2.5 mM Mg(OAc)2) selama 1 jam,
diikuti dengan pencucian ekstensif. Sampel dipisahkan dengan SDS-PAGE dan
diwarnai dengan Coomassie biru. Pita spesifik diiris untuk identifikasi protein
dengan spektrometri massa. Protein-protein yang berinteraksi diidentifikasi
baik untuk GST maupun GST-BARS. Protein-protein yang hanya berinteraksi
dengan GST-BARS dianggap spesifik.

6. Transfeksi dan siRNA. Transfeksi plasmid DNA dilakukan menggunakan


FuGene6 (Roche). Transfeksi siRNA dilakukan menggunakan Lipofectamine
RNAiMAX (Invitrogen). Transfeksi plasmid berlangsung selama 48 jam dan
transfeksi siRNA berlangsung selama 72 jam. Sekuen siRNA yang digunakan
untuk mentargetkan ALDH7A1 manusia adalah 5'-gcagugagcauguuucuugtt-3'
(untai sense) dan 5'-caagaaacaugcucacugctt-3' (untai komplementer), yang
diperoleh dari Dharmacon. Plasmid penyelamat untuk wildtype dan mutan mati
katalitik dari ALDH7A1 manusia dihasilkan dengan mentargetkan urutan
siRNA ini menggunakan QuikChange Site-Directed-Mutagenesis (Stratagene)
dengan oligonukleotida berpasangan tertentu. Spesifisitas target siRNA-siRNA
ini sudah dijelaskan sebelumnya.

7. Pengujian transportasi in vivo. Assay transportasi berbasis mikroskopi


kuantitatif dari jalur intraseluler yang berbeda telah dijelaskan sebelumnya.
Dilakukan dengan cara yang berbeda untuk tiap kompartemen

8. Mikroskop konfokal. Studi kolokalisasi dilakukan menggunakan dua sistem


mikroskop konfokal. Sistem Nikon dilengkapi dengan Mikroskop Terbalik
Nikon Eclipse TE2000U dengan objektif Plan Apo 60×/1.40 oil, paket
konfokal Nikon D-Eclipse C1 dengan Laser 488 (dengan filter emisi 515/30)
dan Laser 543 (dengan filter emisi 590/50), serta perangkat lunak akuisisi
Nikon EZ-C1 versi 3.90. Sistem Zeiss dilengkapi dengan Mikroskop Terbalik
Zeiss Axio Observer Z1 dengan objektif Plan-Apochromat 63×, Zeiss LSM
800 dengan paket konfokal Airyscan dengan garis laser Zeiss URGB (488 dan
561 nm), serta perangkat lunak akuisisi konfokal Zen 2.3 edisi biru. Untuk
kuantifikasi kolokalisasi, sepuluh bidang sel diuji, dan setiap bidang biasanya
mengandung lima sel. Gambar diimpor ke perangkat lunak NIH Image J versi
1.50e, dan kemudian dianalisis melalui perangkat lunak plugin
(https://imagej.net/Coloc_2). Di bawah tab "Image," opsi "Split Channels"
dipilih. Di bawah tab "Plugins," opsi "Colocalization Analysis" dipilih, dan
dalam opsi ini, opsi "Colocalization Threshold" dipilih. Nilai kolokalisasi
kemudian dihitung oleh perangkat lunak dan diungkapkan sebagai fraksi
protein yang diminati (kargo atau ALDH7A1) yang kolokalisasi dengan
penanda organel.

9. Rekonstruksi in vitro pembentukan vesikel COPI. Sistem rekonstruksi


dilakukan secara essensial seperti yang telah dijelaskan sebelumnya14,16.
Secara singkat, membran golgi (0,2 mg/ml) dicuci dengan KCl 3 M, dan
kemudian diinkubasi dengan ARF1 (6 μg/ml) dan coatomer (6 μg/ml) untuk
inkubasi tahap pertama yang mereproduksi rekruitmen coatomer yang
tergantung pada ARF pada membran golgi. Membran golgi kemudian diisolasi
kembali dan diinkubasi dengan ARFGAP1 (2 μg/ml) dan BARS (2 μg/ml)
untuk tahap kedua yang menghasilkan pembentukan vesikel. ALDH7A1 (1
μg/ml) ditambahkan pada tahap kedua. Produk aktivitas ALDH7A1 juga
ditambahkan pada tahap ini. Termasuk di antaranya: asam heksanoat (20 μM),
asam oktanoat (20 μM), 2-aminoadipat (20 μM), atau NADH (20 μM).
Pemeriksaan EM pada membran golgi menggunakan teknik seluruh-sampel
telah dijelaskan sebelumnya. Secara singkat, sampel membran dari sistem
rekonstruksi COPI dititiskan pada grid EM dan kemudian difiksasi dengan 2%
PFA/PBS selama 10 menit. Grid dibilas tiga kali dengan air, diikuti dengan
pewarnaan asetat uranil 1%, dan kemudian diperiksa menggunakan mikroskop
elektron transmisi JEOL 1200EX.

10. Fraksinasi subseluler. Sel-sel dicuci dengan PBS, diresuspensi dalam


buffer homogenisasi (0,25 M sukrosa, 1 mM EDTA, dan 20 mM HEPES-KOH,
pH 7,4, serta inhibitor protease), kemudian dihancurkan dengan melewati
jarum 28-gauge. Setelah sentrifugasi kecepatan rendah (800×g selama 6 menit)
untuk mengendapkan inti dan sel yang tidak pecah, supernatan pasca-inti yang
dihasilkan sentrifugasi dengan kecepatan tinggi (100.000×g selama 1 jam)
untuk mendapatkan fraksi sitosol dan membran total. Untuk mendapatkan
fraksi nukleus vs. sitoplasma, sel-sel dicuci dengan PBS dan diresuspensi
dalam buffer hipotonik yang mengandung 10 mM HEPES, pH 8,0, 10 mM
KCl, 3 mM MgCl2, 0,5 mM DTT, dan inhibitor protease. Suspensi kemudian
diinkubasi pada suhu es selama 10 menit dan Triton X-100 ditambahkan hingga
konsentrasi akhir 0,3%. Setelah sentrifugasi kecepatan rendah (800×g selama 5
menit), supernatan dikumpulkan sebagai fraksi sitoplasma. Endapan dicuci dua
kali dengan buffer hipotonik lagi dan diresuspensi dalam buffer lisis yang
mengandung 100 mM Tris-HCl, pH 8,0, 1% Triton X-100, 100 mM NaCl, 0,5
mM EDTA, dan inhibitor protease. Setelah sentrifugasi pada 15.000×g selama
10 menit, supernatan dikumpulkan sebagai fraksi nukleus.

11. Studi in vitro menggunakan liposom. Liposom dihasilkan dari lipid


murni yang telah ditentukan untuk meniru komposisi membran alami (mol%):
dioleoyl-fosfatidilkolin (DOPC) 50%, dioleoyl-fosfatidiletanolamin (DOPE)
16%, dioleoyl-fosfatidilserin (DOPS) 5%, PA 5%, kolesterol 17%, dan
sfingomielin 7%. Lipid murni ini dicampur dalam tabung kaca, dikeringkan
dengan nitrogen, lalu diresuspensi dalam buffer (25 mM Hepes–KOH, pH 7,2,
50 mM KCl, 2,5 mM Mg(OAc)2, dan 0,2 M sukrosa), dengan dilakukan
sepuluh kali pembekuan cepat dan pencairan dengan pengadukan sesekali.
Liposom raksasa yang dihasilkan kemudian diapitkan melalui mini-extruder
(saringan 400 nm) sebanyak 21 kali. Untuk merekonstruksi aktivitas
ALDH7A1 dalam konteks liposom, liposom (20 μg) dicampur dengan NAD
(10 μM) dan substrat (oktanal (10 μM) atau betain aldehida (10 μM)) dalam
200 μl PBS, kemudian ditambahkan ALDH7A1 rekombinan (1 μg). Inkubasi
dilakukan pada 37 °C selama 1 jam. Selanjutnya, pengukuran NADH dilakukan
menggunakan metode pengujian siklus enzim. Kinetika enzim ALDH7A1
ditentukan seperti yang telah dijelaskan sebelumnya. Untuk menghitung Km,
inkubasi dilakukan dengan memvariasikan tingkat substrat dalam keberadaan
NAD berlebih.

12. Pengujian kinase in vitro. Pengujian kinase dilakukan secara prinsip


seperti yang telah dijelaskan sebelumnya. Untuk deteksi fosforilasi
menggunakan antibodi fosfo-substrat AMPK, AMPK1 yang berlabel myc
diisolasi dari sel-sel yang di-transfeksi yang sebelumnya telah kelaparan dan
diobati dengan AICAR. Untuk deteksi fosforilasi menggunakan γP32-ATP atau
menggunakan pengujian kinasa ADP-Glo (Promega), AMPK aktif rekombinan
diperoleh (Sinobiological). Dalam semua kasus, AMPK diinkubasi dengan
ALDH7A1 rekombinan dalam keberadaan ATP dan inhibitor fosfatase. Untuk
menilai stoikiometri fosforilasi, peptida SAMS, suatu urutan dari substrat
AMPK yang optimal, ACC, digunakan untuk perbandingan.
13. Pengujian lainnya. Tingkat ATP seluler total dideteksi menggunakan
ATPlite Luminescence Detection Assay System (Perkin Elmer), dan dilakukan
sesuai petunjuk produsen, dengan nilai akhir dinormalisasi dengan jumlah sel
yang dapat hidup. Viabilitas sel dinilai dengan menginkubasi sel dengan iodin
propidium (1 ng/μl) dan kemudian dihitung menggunakan flow cytometry.
Pengukuran NAD/NADH dilakukan menggunakan pengujian siklus enzim
standar sesuai petunjuk produsen (BioVision).

14. Kuantifikasi mRNA. RNA total diisolasi menggunakan kit PureLink


(Ambion). mRNA total kemudian diisolasi menggunakan kit MicroPolyAPurist
(Ambion). Keduanya dilakukan sesuai petunjuk produsen. Kuantifikasi
dilakukan dengan mengukur kepadatan optik pada 260 nm dan dinormalisasi
untuk jumlah sel. Kuantifikasi transkrip RNA menggunakan real-time PCR
(qPCR) telah dijelaskan38. Kondisi siklusnya adalah: denaturasi pada 95 °C
selama 30 detik, annealing pada 56 °C selama 60 detik, dan ekstensi pada 72
°C selama 30 detik. Urutan primer yang digunakan untuk mendeteksi mRNA
untuk E-kadherin, ALDH7A1, Ftcd, Catalase, ALDO, dan MDH2, dibeli dari
Integrated DNA Technologies, Inc.

15. Gel filtration. Analisis dilakukan menggunakan sistem AKTA UPC-900


FPLC (GE Healthcare) dengan kolom Superdex 200 10/200GL, dengan
parameter berikut: volume sampel 500 μl, eluen buffer 20 mM Tris-HCl, 100
mM NaCl, pH 7.2, laju aliran 0.5 ml/menit.

16. Analisis statistik. Ukuran sampel dicatat dalam bentuk gambar. Ukuran
sampel yang digunakan didasarkan pada keakraban kami sebelumnya dengan
pengujian. Untuk perbandingan antara dua kondisi, signifikansi diuji dengan uji
t dua sisi berpasangan (paired two-tailed Student's t-test). Untuk perbandingan
di antara beberapa kondisi, signifikansi diuji dengan analisis varians
(ANOVA). Pengujian ini dilakukan menggunakan perangkat lunak Excel atau
Prism. Tidak ada kriteria inklusi/eksklusi yang telah ditetapkan sebelumnya.
Eksperimen tidak diacak. Para peneliti tidak dibutakan terhadap alokasi
kelompok selama eksperimen dan dalam penilaian hasil.

5 Result/Hasil 1. ALDH7A1 menghambat fisi vesikel COPI dengan mentargetkan BARS.


SiRNA terhadap ALDH7A1 meningkatkan transportasi COPI, sedangkan
ALDH7A1 berlebihan menghambatnya. ALDH7A1 menghambat pembentukan
vesikel COPI dengan menghambat tahap fisi. Aktivitas katalitik ALDH7A1
diperlukan untuk efeknya. ALDH7A1 menghasilkan NADH yang menghambat
BARS, sehingga menghambat fisi vesikel COPI. Namu, ketika BARS wildtype
digantikan dengan mutan (G172E) BARS, kami menemukan bahwa ALDH7A1
tidak lagi dapat menghambat pembentukan vesikel COPI. Selain itu, mutasi
G172E mencegah kemampuan NADH untuk menghambat tubulasi liposom
oleh BARS

2. ALDH7A1 menghambat banyak jalur intraseluler selain transportasi


COPI. Hasil penelitian menunjukkan bahwa siRNA terhadap ALDH7A1
meningkatkan transportasi dari golgi ke membran plasma dan tiga jalur
endositik utama lainnya, termasuk transportasi endositik ke lisosom. Namun,
endositosis termediasi klatrin tidak terpengaruh oleh ALDH7A1. Enam dari
delapan jalur intraseluler utama menjadi target ALDH7A1 untuk inhibisi.
Ketika ekspresi ALDH7A1 berlebihan, efek-efek ini berbalik, tetapi
memerlukan aktivitas katalitik ALDH7A1. Ekspresi berlebihan ALDH7A1
tidak lagi menghambat jalur-jalur yang tergantung pada BARS ketika BARS
endogen digantikan dengan BARS mutan yang cacat dalam pengikatan NADH.
Ini menunjukkan bahwa ALDH7A1 menghambat jalur-jalur tersebut dengan
menghasilkan NADH yang mentargetkan BARS. Penelitian juga menguji
apakah jalur-jalur yang terpengaruh dapat dijelaskan hanya dengan interaksi
ALDH7A1 dengan BARS. Hasil menunjukkan bahwa jalur-jalur yang
melibatkan BARS dalam tahap fisi terpengaruh oleh siRNA terhadap BARS,
sementara jalur-jalur yang tidak melibatkan BARS tidak terpengaruh. Ini
menandakan bahwa ALDH7A1 mungkin menargetkan faktor lain selain BARS
untuk memberikan penghambatan yang luas pada jalur-jalur transportasi.
Fraksinasi sel juga menunjukkan bahwa ALDH7A1 berinteraksi dengan BARS
pada membran, tetapi tidak di sitosol. ALDH7A1 juga terdistribusi luas di
antara kompartemen membran intraseluler, dengan kadar yang berkurang di
RE, yang sesuai dengan temuan bahwa ALDH7A1 tidak mempengaruhi
transportasi dari RE. Dalam mencari lebih lanjut mengenai penghambatan
transportasi oleh ALDH7A1, penelitian menambahkan sinyal ekspor nukleus
pada BARS untuk mencegah lokalisasinya di nukleus. Hasil menunjukkan
bahwa penghambatan transportasi oleh ALDH7A1 tidak melibatkan fungsi
transkripsi BARS.
3. ALDH7A1 berperan dalam meningkatkan homeostasis energi seluler.
Penelitian ini mencari tahu makna fisiologis dari penghambatan transportasi
yang dilakukan oleh ALDH7A1 selama stres energi, khususnya hipoksia dan
kelaparan. Hasil penelitian menunjukkan bahwa penghambatan transportasi
oleh hipoksia dan kelaparan memerlukan kehadiran ALDH7A1. Ketika
ALDH7A1 dihilangkan melalui siRNA, sel mengalami penurunan tingkat ATP
dan peningkatan kematian sel selama kondisi stres energi. Hal ini menunjukkan
bahwa ALDH7A1 memiliki peran pelindung selama stres energi dan
berkontribusi pada homeostasis energi seluler. Selanjutnya, penelitian ini
mengeksplorasi hubungan antara AMPK dan ALDH7A1. AMPK merupakan
koordinator utama homeostasis energi seluler dan terlibat dalam mendeteksi
defisit energi dan menargetkan efektor untuk mengembalikan keseimbangan
energi. Penelitian menunjukkan bahwa AMPK berperan dalam mengatur jalur
transportasi yang ditargetkan oleh ALDH7A1. Aktivasi AMPK dengan
pengobatan AICAR menghambat transportasi dalam kondisi normal, dan
penghambatan ini memerlukan kehadiran ALDH7A1. Hasil ini menunjukkan
bahwa AMPK berfungsi mekanistik di atas ALDH7A1 dan BARS dalam
mengatur jalur transportasi. Penelitian ini juga menemukan bahwa AMPK
dapat mengfosforilasi ALDH7A1 pada residu S102. Fosforilasi ini
mempengaruhi fungsi ALDH7A1 dalam menghambat transportasi selama stres
energi. Mutasi S102A pada ALDH7A1 mencegah fosforilasi dan mengurangi
kemampuan ALDH7A1 untuk menghambat transportasi selama stres energi,
sementara mutasi S102D yang meniru fosforilasi konstitutif meningkatkan
kemampuan ALDH7A1 dalam menghambat transportasi selama stres energi.
Hal ini menunjukkan bahwa fosforilasi S102 pada ALDH7A1 berperan penting
dalam regulasi fungsi ALDH7A1 selama kondisi stres energi.

4. Fosforilasi oleh AMPK mentargetkan ALDH7A1 ke membran.


Kemudian, penelitian dilakukan untuk menguji bagaimana fosforilasi residu
S102 pada ALDH7A1 mengatur kemampuannya untuk menghambat
transportasi. Awalnya, diketahui bahwa aktivitas katalitik ALDH7A1 tidak
dipengaruhi oleh mutasi residu S102. Namun, ditemukan bahwa rekrutmen
ALDH7A1 dari sitoplasma ke membran merupakan cara utama dalam
mengatur faktor-faktor transportasi. Oleh karena itu, penelitian menunjukkan
bahwa fosforilasi residu S102 mengatur rekrutmen ALDH7A1 dari sitoplasma
ke membran. Mutan S102A memiliki distribusi yang lebih rendah pada
membran dibandingkan dengan bentuk tipe liar, sedangkan mutan S102D
memiliki distribusi yang meningkat pada membran. Selanjutnya, penelitian
menemukan bahwa kondisi hipoksia dan kelaparan, yang mengaktifkan
fosforilasi AMPK pada ALDH7A1, meningkatkan distribusi ALDH7A1 pada
membran serta meningkatkan lokalisasi ALDH7A1 pada kompartemen
intraseluler. Hasil ini menunjukkan bahwa rekrutmen ALDH7A1 ke membran
terjadi dalam kondisi stres energi tersebut. Selain itu, penelitian juga
mengungkapkan bahwa membran golgi mengandung NAD, yang dapat diubah
menjadi NADH oleh ALDH7A1. Hal ini menandakan bahwa membran golgi
memiliki substrat dan kofaktor yang diperlukan untuk aktivitas katalitik
ALDH7A1. Eksperimen lebih lanjut dengan liposom menunjukkan bahwa
ALDH7A1 pada membran dapat langsung mengubah NAD menjadi NADH,
dan hal ini terjadi khususnya pada mutan S102D. Selanjutnya, penelitian
mendetail bagaimana NADH yang dihasilkan oleh ALDH7A1 menghambat
BARS (enzim lain yang terlibat dalam transportasi seluler). Ketika membran
golgi diinkubasi dengan mutan S102A, BARS tetap berada pada membran,
sedangkan ketika diinkubasi dengan mutan S102D, BARS menjadi dilepaskan
dari membran. Eksperimen ini menunjukkan bahwa fosforilasi residu S102
pada ALDH7A1 mempengaruhi peran ALDH7A1 dalam menghambat
transportasi dengan meningkatkan rekrutmennya dari sitoplasma ke membran,
dan NADH yang dihasilkan oleh ALDH7A1 berperan dalam mekanisme ini

5 Discussion Hasil penelitian relevan dengan latar belakang serta tujuan dari penelitian ini.
Latar belakang adanya penelitian ini adalah untuk mengeksplorasi peran
ALDH7A1 dalam menghambat fisi vesikel COPI dan jalur intraseluler lainnya,
serta memahami mekanisme di balik penghambatan ini. Selain itu, penelitian
ini juga bertujuan untuk mengeksplorasi peran ALDH7A1 dalam menjaga
homeostasis energi seluler selama kondisi stres energi seperti hipoksia dan
kelaparan.

Hasil penelitian tersebut menyajikan bukti bahwa ALDH7A1 menghambat


pembentukan vesikel COPI dengan mentargetkan BARS dan menghasilkan
NADH yang menghambat BARS, sehingga menghambat fisi vesikel COPI.
Selain itu, ALDH7A1 juga menghambat banyak jalur intraseluler selain
transportasi COPI, dan penghambatan ini tampaknya melibatkan interaksi
dengan faktor-faktor lain selain BARS.

Penelitian juga menunjukkan bahwa ALDH7A1 berperan dalam meningkatkan


homeostasis energi seluler selama stres energi, seperti hipoksia dan kelaparan,
dan bahwa AMPK berperan dalam mengatur jalur transportasi yang ditargetkan
oleh ALDH7A1. Fosforilasi oleh AMPK mentargetkan ALDH7A1 ke
membran, di mana NADH yang dihasilkan oleh ALDH7A1 mempengaruhi
fungsi ALDH7A1 dalam menghambat transportasi selama kondisi stres energi.

Persamaan:
1. Penelitian ini melibatkan peran ALDH7A1 dalam menghambat pembentukan
vesikel COPI dan jalur intraseluler lainnya.
2. ALDH7A1 menghasilkan NADH, yang berkontribusi dalam menghambat
fisi vesikel COPI melalui penghambatan BARS.
3. Penelitian ini menyoroti pentingnya aktivitas katalitik ALDH7A1 dalam
efeknya dalam menghambat transportasi intraseluler.
4. ALDH7A1 berperan dalam menjaga homeostasis energi seluler, terutama
selama kondisi stres energi seperti hipoksia dan kelaparan.
5. ALDH7A1 berinteraksi dengan BARS pada membran seluler dan memiliki
distribusi yang luas di antara kompartemen membran intraseluler.

Perbedaan:
1. Dalam penelitian ini, ALDH7A1 ditemukan menghambat COPI vesicle
fission dengan menghasilkan NADH dari reaksi metabolik yang melibatkan
BARS. Sementara itu, dalam penelitian lain yang melibatkan enzim GAPDH,
ditemukan bahwa GAPDH menghambat fisi vesikel COPI dengan menargetkan
ARFGAP1.
2. ALDH7A1 menghasilkan NADH yang terlokalisasi pada membran seluler,
sementara banyak enzim metabolik lainnya menghasilkan NADH yang
terutama berada di sitosol.
3. Hasil penelitian ini mengidentifikasi ALDH7A1 sebagai regulator kunci
dalam menghambat beberapa jalur transportasi intraseluler lainnya, sedangkan
banyak enzim metabolik hanya mengatur jalur khusus yang terkait dengan
fungsi katalitik mereka.
4. Dalam penelitian ini, ditemukan bahwa ALDH7A1 berperan dalam
melindungi sel selama kondisi stres energi, sementara dalam beberapa
penelitian lain, peran ALDH7A1 lebih terkait dengan metabolisme asam amino
dan stres oksidatif.
5. Dampak dari defisiensi ALDH7A1 dalam kondisi normal diketahui kurang
signifikan, tetapi dalam kondisi stres energi seperti hipoksia dan kelaparan,
peran protektif ALDH7A1 menjadi lebih penting.

Temuan Unik:
1. ALDH7A1 memiliki peran unik dalam mengatur jalur transportasi
intraseluler dan homeostasis energi seluler melalui dengan menghasilkan
NADH yang terlokalisasi pada membran seluler.
2. Penelitian ini menemukan bahwa ALDH7A1 berperan dalam menghambat
jalur intraseluler yang berbeda selain transportasi COPI, dan pemahaman ini
diperluas dengan mencakup koordinasi inhibisi pada banyak jalur transportasi
untuk mencapai pengurangan yang bermakna dalam konsumsi energi seluler.
3. Penemuan ini juga mengungkapkan potensi terapeutik ALDH7A1 dalam
mengatasi gangguan jalur transportasi seluler dan homeostasis energi seluler
dalam kondisi patologis, seperti dalam kasus cardiac ischemia.
4. Fokus pada peran ALDH7A1 dalam melindungi sel selama kondisi stres
energi memberikan wawasan baru tentang adaptasi seluler terhadap kondisi
lingkungan yang menuntut dan peran penting ALDH7A1 dalam menjaga
kelangsungan hidup sel selama situasi tersebut.

Hasil penelitian ini memiliki beberapa dampak penting terhadap ilmu


pengetahuan:

1. Memperluas pemahaman tentang peran ALDH7A1 dalam regulasi


transportasi intraseluler: Penelitian ini telah mengidentifikasi peran baru dari
enzim ALDH7A1 dalam menghambat pembentukan vesikel COPI dan jalur
intraseluler lainnya. Ini membantu memperluas pemahaman kita tentang
kompleksitas regulasi transportasi intraseluler dan memberikan wawasan
tentang cara-cara baru di mana sel mengatur lalu lintas substansi antara
kompartemen seluler.

2. Penemuan mekanisme unik penghambatan COPI vesicle fission: Penemuan


bahwa ALDH7A1 menghambat COPI vesicle fission melalui dengan
menghasilkan NADH dari reaksi metabolik yang melibatkan BARS,
menawarkan wawasan baru tentang mekanisme regulasi jalur transportasi
intraseluler. Hal ini memberikan pandangan yang lebih komprehensif tentang
bagaimana sel mengatur pembentukan dan transportasi vesikel intraseluler.

3. Menyoroti peran NADH membran-bound dalam regulasi jalur intraseluler:


Temuan bahwa ALDH7A1 menghasilkan NADH yang terlokalisasi pada
membran selular menunjukkan bahwa reaksi metabolik dan kofaktor yang
dihasilkan oleh enzim dapat berfungsi secara lokal dan spesifik dalam regulasi
jalur intraseluler. Ini membuka potensi penelitian lebih lanjut tentang peran
NADH membran-bound dalam regulasi aktivitas enzim dan jalur seluler
lainnya.

4. Implikasi pada homeostasis energi seluler dan adaptasi terhadap stres energi:
Penelitian ini mengungkapkan bahwa ALDH7A1 memiliki peran penting
dalam menjaga homeostasis energi seluler selama kondisi stres energi seperti
hipoksia dan kelaparan. Temuan ini dapat berkontribusi pada pemahaman
tentang mekanisme adaptasi seluler terhadap kondisi lingkungan yang
menuntut.

5. Potensi terapeutik dalam kondisi patologis: Penelitian ini menunjukkan


bahwa ALDH7A1 memiliki peran dalam melindungi sel selama kondisi stres
energi, dan ini dapat memberikan potensi terapeutik dalam mengatasi penyakit
dan kondisi patologis yang melibatkan gangguan dalam regulasi transportasi
intraseluler dan homeostasis energi seluler. Hasil ini dapat mendorong
pengembangan terapi yang berfokus pada pengaturan ALDH7A1 untuk
memperbaiki gangguan jalur transportasi seluler dalam berbagai kondisi
penyakit.

Secara keseluruhan, penelitian ini memberikan sumbangan signifikan terhadap


ilmu pengetahuan dengan memperluas pemahaman tentang peran ALDH7A1
dalam regulasi transportasi intraseluler dan homeostasis energi seluler. Hasil ini
dapat membuka pintu bagi penelitian lebih lanjut tentang fungsi dan potensi
terapeutik ALDH7A1 dalam konteks biologi seluler dan patologi.

Namun, seperti setiap penelitian, ada beberapa keterbatasan yang perlu diakui:
1. Penelitian in vitro: sebagian besar penelitian ini dilakukan dalam model sel
in vitro, yang berarti bahwa hasilnya mungkin tidak sepenuhnya
merepresentasikan kondisi yang terjadi di tubuh manusia secara keseluruhan.
Oleh karena itu, perlu penelitian lanjutan di model hewan atau manusia untuk
mengonfirmasi temuan ini dan mengevaluasi dampaknya pada organisme
hidup.

2. Keterbatasan model sel: penelitian ini mungkin menggunakan model sel


tertentu yang tidak sepenuhnya mencerminkan kondisi di seluruh tubuh. Sifat
dan perilaku sel dapat bervariasi berdasarkan jenis sel, lingkungan, dan
interaksi dengan sel-sel lain. Oleh karena itu, hasil dari satu jenis sel mungkin
tidak dapat langsung diterapkan pada jenis sel lainnya.

3. Kompleksitas jalur transportasi intraseluler: jalur transportasi intraseluler


adalah proses yang kompleks dan sangat teratur. Meskipun penelitian ini telah
mengidentifikasi ALDH7A1 sebagai regulator penting dalam penghambatan
jalur transportasi, masih banyak interaksi dan mekanisme lain yang mungkin
berperan. Penelitian lebih lanjut diperlukan untuk memahami interaksi
ALDH7A1 dengan faktor-faktor lain yang terlibat dalam transportasi
intraseluler.

4. Pentingnya in vivo validation: hasil penelitian in vitro harus diuji kembali


dalam setting in vivo untuk mengonfirmasi temuan dan memahami bagaimana
peran ALDH7A1 berkontribusi pada fungsi seluler dan fisiologi tubuh manusia
secara keseluruhan.

5. Relevansi klinis: walaupun penelitian ini memberikan pemahaman yang


lebih dalam tentang peran ALDH7A1 dalam berbagai kondisi stres, termasuk
iskemia jantung, dampak klinis dari temuan ini pada pengobatan penyakit
masih perlu dievaluasi lebih lanjut. Langkah-langkah pengobatan yang efektif
memerlukan lebih dari sekadar memahami peran biologis suatu molekul, dan
studi klinis akan diperlukan untuk menilai potensi ALDH7A1 sebagai target
terapeutik.

6. Penerapan pada manusia: penelitian ini mungkin menemukan efek


ALDH7A1 pada sel dan jalur transportasi intraseluler, tetapi implikasinya pada
manusia secara keseluruhan mungkin lebih kompleks. Faktor-faktor lain seperti
faktor genetik, lingkungan, dan kondisi kesehatan individu dapat
mempengaruhi bagaimana ALDH7A1 berinteraksi dan berkontribusi pada
fungsi seluler dan homeostasis energi dalam tubuh manusia.

Dengan mengakui keterbatasan ini, penelitian ini memberikan dasar yang kuat
untuk penelitian lebih lanjut dalam memahami peran dan potensi terapeutik
dari ALDH7A1 dalam berbagai kondisi patologis dan fisiologis. Hasil ini dapat
memberikan panduan bagi pengembangan terapi baru yang berfokus pada
regulasi transportasi intraseluler dan homeostasis energi seluler untuk
meningkatkan kesehatan dan mengatasi berbagai gangguan dan penyakit.

6 Kesimpulan Semua informasi yang disajikan dalam hasil penelitian ini berhasil menjawab
hipotesis dan tujuan penelitian dengan baik. Berdasarkan seluruh bagian jurnal
ini, dapat disimpulkan bahwa:

1. Latar belakang penelitian menjelaskan bahwa ALDH7A1 berperan dalam


menghambat fisi vesikel COPI dengan mentargetkan BARS, serta memiliki
pengaruh pada banyak jalur intraseluler lainnya, termasuk transportasi dari
golgi ke membran plasma dan jalur endositik.

2. Tujuan penelitian adalah untuk memahami peran ALDH7A1 dalam


mengatur transportasi intraseluler dan homeostasis energi seluler selama
kondisi stres energi, seperti hipoksia dan kelaparan.

3. Hasil penelitian tersebut menyajikan bukti bahwa ALDH7A1 menghambat


pembentukan vesikel COPI dengan mentargetkan BARS dan menghasilkan
NADH yang menghambat BARS, sehingga menghambat fisi vesikel COPI.
Selain itu, ALDH7A1 juga menghambat banyak jalur intraseluler selain
transportasi COPI, dan penghambatan ini tampaknya melibatkan interaksi
dengan faktor-faktor lain selain BARS.

Penelitian juga menunjukkan bahwa ALDH7A1 berperan dalam meningkatkan


homeostasis energi seluler selama stres energi, seperti hipoksia dan kelaparan,
dan bahwa AMPK berperan dalam mengatur jalur transportasi yang ditargetkan
oleh ALDH7A1. Fosforilasi oleh AMPK mentargetkan ALDH7A1 ke
membran, di mana NADH yang dihasilkan oleh ALDH7A1 mempengaruhi
fungsi ALDH7A1 dalam menghambat transportasi selama kondisi stres energi.

4. Temuan unik penelitian ini adalah bahwa ALDH7A1 menghambat jalur


transportasi intraseluler melalui dengan menghasilkan NADH, yang
menyebabkan penghambatan BARS dan transportasi intraseluler secara luas.

Penemuan ini memberikan kontribusi signifikan terhadap pemahaman kita


tentang mekanisme regulasi intraseluler dan potensi terapeutik untuk mengatasi
kondisi patologis yang melibatkan situasi stres energi.

You might also like